Supra R Renal

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

PRIMER

Adrenal insufficiency
Stefanie Hahner   1 ✉, Richard J. Ross   2, Wiebke Arlt   3,4, Irina Bancos5,
Stephanie Burger-​Stritt1, David J. Torpy6, Eystein S. Husebye7,8,9 and Marcus Quinkler10
Abstract | Adrenal insufficiency (AI) is a condition characterized by an absolute or relative
deficiency of adrenal cortisol production. Primary AI (PAI) is rare and is caused by direct adrenal
failure. Secondary AI (SAI) is more frequent and is caused by diseases affecting the pituitary,
whereas in tertiary AI (TAI), the hypothalamus is affected. The most prevalent form is TAI owing to
exogenous glucocorticoid use. Symptoms of AI are non-​specific, often overlooked or misdiagnosed,
and are related to the lack of cortisol, adrenal androgen precursors and aldosterone (especially in
PAI). Diagnosis is based on measurement of the adrenal corticosteroid hormones, their regulatory
peptide hormones and stimulation tests. The goal of therapy is to establish a hormone replacement
regimen that closely mimics the physiological diurnal cortisol secretion pattern, tailored to the
patient’s daily needs. This Primer provides insights into the epidemiology, mechanisms and
management of AI during pregnancy as well as challenges of long-​term management. In addition,
the importance of identifying life-​threatening adrenal emergencies (acute AI and adrenal crisis)
is highlighted and strategies for prevention, which include patient education, glucocorticoid
emergency cards and injection kits, are described.

Glucocorticoids Adrenal insufficiency (AI) is an endocrine disorder caused by the administration of supraphysiological
A class of corticosteroids characterized by adrenal hypofunction, leading to inad- doses of exogenous glucocorticoids for a sustained
that are essential mediators equate production of glucocorticoids, especially cortisol. period of time8 (Fig. 2).
of the stress system and AI can be primary, secondary or tertiary depending on The clinical signs and symptoms of AI are related
major players in the stress
response, that regulate resting
the underlying aetiology. Accordingly, the production to the respective hormone deficiency, which are often
homeostasis (metabolism of mineralocorticoids and adrenal androgens might also manifested gradually over a long period of time. Major
of carbohydrates, proteins be affected1 (Fig. 1). Primary adrenal insufficiency (PAI) symptoms associated specifically with cortisol deficiency
and fats) and modulate the results from direct adrenal gland failure caused by the include fatigue, hypotension and weight loss; however,
immune response.
destruction of or damage to the adrenal glands (Fig. 2). these symptoms are non-​specific and often lead to diag-
Mineralocorticoids Autoimmune adrenalitis, also known as Addison dis- nostic delays and misdiagnosis9. Unexplained hypon-
A class of corticosteroids ease, is the most common cause of PAI, especially in atraemia as well as hyperpigmentation or loss of pubic
involved in the maintenance high-​income countries, whereas the proportion of and axillary hair might more specifically raise suspi-
of the salt and water balance patients with PAI in whom the AI is attributable to cion of AI. AI can also manifest as an acute emergency
in the body.
infectious diseases, such as tuberculosis or HIV, is high known as adrenal crisis. Owing to the crucial protective
Immune checkpoint in countries with high prevalence of these infections2–7. role of cortisol under stressful conditions (physical and
inhibitors Secondary adrenal insufficiency (SAI) occurs secondary psychological), patients with unknown AI and patients
Drugs typically used in to diseases of the pituitary or the hypothalamus such as under established replacement therapy are at risk of
oncology that stimulate
tumours of the hypothalamus and/or pituitary and their developing life-​threatening adrenal crisis10–15. Thus, pre-
antitumour immune response
by blocking checkpoint treatment, hypophysitis or granulomatous infiltration3. vention of adrenal crisis is a principal goal of long-​term
proteins and promoting AI caused by hypothalamic abnormalities or dysfunc- management.
immune-​mediated elimination tion, which results in reduced corticotropin-​releasing Iatrogenic causes of AI are of important relevance
of tumour cells. hormone (CRH) secretion is referred to as tertiary for many medical disciplines. Glucocorticoid phar-
adrenal insufficiency (TAI). Similar to SAI, the con- macotherapy and opiates, which are known to impair
sequent lack of adrenal adrenocorticotropic hormone the function of the HPA axis, are broadly used to
(ACTH) stimulation leads to reduced cortisol and treat multiple conditions. The more widespread use
dehydroepiandrosterone (DHEA) secretion (Fig.  2). of immune checkpoint inhibitors (ICIs), which are asso-
TAI is, therefore, frequently subsumed within the term ciated with autoimmune hypophysitis and rarely with
✉e-​mail: Hahner_S@ukw.de SAI without further differentiation between the two auto­immune adrenalitis, has further increased the inci-
https://doi.org/10.1038/ entities. TAI typically results from the suppression dence of AI15. Hence, knowledge of the management of
s41572-021-00252-7 of the hypothalamic–pituitary–adrenal (HPA) axis AI and the prevention and treatment of adrenal crisis


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 1

0123456789();:
Primer

Adrenal
gland
Kidney

Zona glomerulosa
Mineralocorticoids
• Aldosterone and corticosterone

Zona fasciculata
Glucocorticoids
• Cortisol and cortisone

Zona reticularis
Classic and 11-oxygenated
androgen precursors
• DHEA, DHEAS, androstenedione
and 11-hydroxyandrostenedione
Cortex
Medulla
Catecholamines
• Epinephrine and noreprinephrine
Peptides
• Somatostatin and substance P

Fig. 1 | Adrenal gland hormones. A schematic cross section of adrenal glands showing the different anatomical layers
and the different hormones secreted by each layer. The adrenal glands consist of two parts — an outer steroid-​secreting
adrenal cortex and an inner neuroendocrine adrenal medulla. The adrenal cortex can be further subdivided into three
zones, namely the outer zona glomerulosa, the intermediate zona fasciculata and the inner zona reticularis, which secrete
mineralocorticoids, glucocorticoids and adrenal androgen precursors, respectively. The adrenal medulla secretes the
catecholamine hormones, which include epinephrine and norepinephrine and small amounts of dopamine.
DHEA, dehydroepiandrosterone; DHEAS, dehydroepiandrosterone sulfate.

is relevant for physicians across multiple disciplines. normal physiology is important for optimal patient
In contrast to the rapid improvement in well-​b eing management.
achieved by the initiation of replacement therapy in In this Primer, we summarize the current knowledge
patients with newly diagnosed AI, long-​term manage- of the epidemiology, pathophysiology and diagnosis of
ment is much more challenging. An individually tailored AI and highlight the latest developments in its therapeu-
glucocorticoid replacement therapy that closely mimics tic management. In addition, we describe the specific
challenges in current management, with a particular
focus on the prevention and treatment of adrenal crisis.
Author addresses Furthermore, we discuss the outstanding questions in
1
Department of Medicine I, Division of Endocrinology and Diabetology, University the field and provide directions for future research.
Hospital Wuerzburg, Wuerzburg, Germany.
2
Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK. Epidemiology
3
Institute for Metabolism and Systems Research, University of Birmingham, Primary adrenal insufficiency
Birmingham, UK. The epidemiology of PAI has changed over the past
4
Centre for Endocrinology, Diabetes, and Metabolism, Birmingham Health Partners, century regarding the prevalence and the spectrum
University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK. of underlying causes. PAI is rare, with a prevalence of
5
Division of Endocrinology, Metabolism and Nutrition, Department of Internal Medicine, 10–20 persons per 100,000 population and has multi-
Mayo Clinic, Rochester, MN, USA.
ple aetiologies (Table 1). Although tuberculosis was one
6
Endocrine and Metabolic Unit, Royal Adelaide Hospital, University of Adelaide,
Adelaide, SA, Australia. of the predominant causes of PAI, nowadays Addison
7
Department of Clinical Science, University of Bergen, Bergen, Norway. disease and congenital adrenal hyperplasia (CAH) are
8
K.G. Jebsen Center for Autoimmune Diseases, University of Bergen, Bergen, Norway. emerging as the prominent causes of PAI3,16,17. Addison
9
Department of Medicine, Haukeland University Hospital, Bergen, Norway. disease accounts for ~90% of non-​CAH cases in indus-
10
Endocrinology in Charlottenburg, Berlin, Germany. trialized countries18. By contrast, infections are the

2 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

The hypothalamic–pituitary–adrenal axis


Primary Secondary Tertiary

PVN PVN PVN


Hypothalamus

↑ CRH ↑ CRH ↓ CRH


Pituitary

↑ ACTH ↓ ACTH ↓ ACTH

Adrenal

↓ Cortisol ↓ Cortisol ↓ Cortisol


↓ DHEA ↓ DHEA ↓ DHEA
↓ Aldosterone Normal aldosterone ↓
Normal
Aldosterone
aldosterone

Fig. 2 | Types of adrenal insufficiency. Circadian input and stress inputs converge on the paraventricular nucleus (PVN)
of the hypothalamus, inducing the release of corticotropin-​releasing hormone (CRH) and arginine vasopressin.
CRH in turn stimulates adrenocorticotropic hormone (ACTH) release into the circulation, which induces the secretion
of corticosteroids, mainly cortisol but also dehydroepiandrosterone (DHEA) and, to a minor extent, aldosterone.
The hypothalamic–pituitary–adrenal axis is regulated by cortisol via feedback regulation both in the hypothalamus and the
pituitary. In primary adrenal insufficiency, glucocorticoid deficiency leads to a loss of the negative feedback of cortisol
on the hypothalamus and pituitary, resulting in increased CRH and ACTH levels. In addition, decreased aldosterone
secretion results in increased renin release from the kidney. In secondary adrenal insufficiency (SAI), ACTH synthesis
and secretion are impaired, leading to a loss of adrenal cortisol and DHEA production; however, aldosterone production
is unimpaired as the renin–angiotensin–aldosterone system signalling cascade remains unaffected. Tertiary adrenal
insufficiency (TAI) may be caused by endogenous factors directly affecting the hypothalamic region or following the
treatment of endogenous Cushing syndrome. Most frequently, exogenous administration of glucocorticoid pharmacotherapy
results in TAI. Diminished hypothalamic CRH secretion in TAI results in reduced ACTH stimulation; similar to SAI, secretion
of cortisol and DHEA are impaired, although aldosterone secretion is preserved. Of note, often differentiation between
SAI and TAI is difficult, or mixed forms of SAI and TAI might occur (for example, by medications). As the resulting changes
in peripheral adrenocortical hormones is comparable, the term “secondary AI” is frequently used for both forms. Adapted
with permission from ref.270, Nicolaides NC, Chrousos GP, Charmandari E. Adrenal Insufficiency. 2017 Oct 14. In: Feingold
KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C,
Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, editors.
Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; PMID: 25905309.

predominant cause of PAI in regions with high prev- adrenal metastases, haemorrhage, infiltrative disorders
alence of tuberculosis, HIV infection and opportunis- and infarction26,28.
tic infections such as cytomegalovirus4–7. Overall, the CAH is caused by deficiency of the steroidogenic
prevalence of Addison disease seems to be increasing. enzyme 21-​hydroxylase and is the most common inher-
For example, the prevalence has been reported to have ited form of PAI29. CAH is an autosomal recessive
21-​Hydroxylase increased from 3.9 cases per 100,000 population in 1968 disorder with ~1 in 50 people harbouring a mutation in
An enzyme involved in the (ref.19) to 22 cases per 100,000 population in Iceland in CYP21A2 (encoding 21-​hydroxylase) The prevalence
biosynthesis of cortisol 2016 (ref.20). The prevalence of Addison disease also dif- of classic CAH varies between populations, ranging
and aldosterone.
fers based on the geographical location, ranging from from 0.5 to 1 person per 10,000 population30. Other less
Autoimmune polyglandular 1.4 cases per 100,000 population in South Africa to frequent genetic causes of PAI include adrenoleukodys­
syndromes 9–22 cases per 100,000 population in Europe2,3,17,21–24. trophy, adrenal hypoplasia congenita and autoimmune
Also known as polyglandular Addison disease usually manifests between 20–50 years polyglandular syndrome (APS) type 1 (ref.31) (Table 1).
autoimmune syndromes
of age, but can occur at any age, although onset is rare
or autoimmune polyendocrine
syndromes. These syndromes in children <2 years of age. PAI of any origin is more Autoimmune comorbidities of Addison disease. In more
comprise a heterogeneous prominent in women than in men, with a peak incidence than two thirds of patients, Addison disease occurs in
group of rare immune-​ in the third decade of life, with higher frequencies in association with other autoimmune diseases, such as part
mediated endocrinopathies individuals with other autoimmune disorders, such as of APS type 1 or APS type 2 (ref.15). APS type 1 is a rare,
affecting more than
one endocrine organ.
patients with type 1 diabetes mellitus, than in the gen- monogenic disorder characterized by childhood onset
Non-endocrine organs eral population17,20,25–27. Other rare causes that lead to with sequential manifestation of the main clinical symp-
may also be involved. adrenal destruction and dysfunction include bilateral toms, such as chronic candidiasis, hypoparathyroidism


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 3

0123456789();:
Primer

Table 1 | Causes of primary adrenal insufficiency


Aetiology Pathogenesis Diagnostic tools
Autoimmune T and B cell autoimmunity against adrenocortical cells 21-​Hydroxylase
autoantibodies
Infection Mycobacteria, bacteria (e.g. Neisseria meningitidis, Haemophilus influenzae, Culture, QuantiFERON
Pseudomonas aeruginosa), viruses (e.g. human immunodeficiency virus, test, PCR, adrenal CT
herpes simplex, cytomegalovirus) or fungi (e.g. Pneumocystis jirovecii)
Tumour Primary tumour (bilateral), metastasis (bilateral), adrenal lymphoma Adrenal CT
(bilateral)
Bleeding Anti-​phospholipid syndrome, anticoagulant therapy, disseminated Adrenal CT, phospholipid
intravascular coagulation autoantibodies
Surgery Bilateral adrenalectomy Patient history
Infiltrative Amyloidosis Adrenal CT,
subcutaneous fat biopsy
Haemochromatosis
Ferritin, HFE sequencing
Histiocytosis
Adrenal imaging
Genetica Congenital adrenal hyperplasia, congenital lipoid adrenal hyperplasia, Sequence of relevant
adrenoleukodystrophy (X-​linked), adrenal hypoplasia congenita, gene
autoimmune polyglandular syndrome type 1
Medicationb Enzyme inhibition (ketoconazole, fluconazole, itraconazole, etomidate, Medication and patient
aminoglutethimide, metyrapone, trilostane, osilodrostat); adrenolytic history
effect and increased cortisol metabolism (mitotane); inflammation
(checkpoint inhibitors)
a
See also Table 3. bSee also Table 4. Data from refs15,31.

and Addison disease32,33. Autoimmune comorbidities of patients receiving ICIs targeting CTLA4 and the com-
Addison disease include autoimmune thyroid disease bination of CTLA4 and PD1 (up to 13% of patients)48,49.
(40% of patients), premature ovarian failure (5–16%), Another form of iatrogenic SAI is opioid-​induced AI,
type 1 diabetes mellitus (11%), pernicious anaemia which has been reported in ~9–15% of patients using
(10%), vitiligo (6%), and coeliac disease (2%)3,34,35. In chronic opioids, and has been reported at doses as
contrast to APS type 1, APS type 2 is most frequently low as 20 morphine milligram equivalents per day50,51.
diagnosed during adulthood. Nevertheless, opioid-​induced AI is poorly recognized
and is probably under-​reported52.
Secondary adrenal insufficiency
SAI is more common than PAI, with a prevalence of up Tertiary adrenal insufficiency
to 42 cases per 100,000 population based on European Tertiary AI may be caused by endogenous factors such
studies17,36,37. SAI typically occurs in patients with dis- as tumours, radiation or inflammatory processes affecting
eases affecting the pituitary or hypothalamus such as the hypothalamic region. TAI is also observed after the
tumours, autoimmune hypophysitis or trauma, or occurs treatment of endogenous Cushing syndrome following
following radiotherapy of intracranial tumours, and removal of an ACTH-​producing or cortisol-​producing
only rarely has a genetic cause (for example, mutations tumour.
of the transcription factor TPIT)37–39 (Table 2). SAI will The most common cause of TAI is chronic exoge-
develop in 10–62% of patients with pituitary adenoma nous administration of glucocorticoids. Glucocorticoid-​
as a result of the mass effect of the tumour on the pitu- induced TAI is also termed iatrogenic or exogenous
itary gland; the risk increases with pituitary surgery or AI and is a temporary condition in most patients8. The
radiotherapy37,39–41. The development of hypopituitarism risk of developing TAI varies according to the route of
(also known as pituitary insufficiency) after radio- glucocorticoid administration owing to differences in
therapy is dose-​d ependent and time-​d ependent 42. systemic availability. The risk of TAI is reported to be
A German study found that in patients with acromegaly 4.2% for intranasal administration, 20% for adminis-
who underwent radiotherapy, the incidence of SAI tration by inhalation, 49% for oral administration and
had increased from 39% to 45% at a mean of 9.1 years 52% for intra-​articular administration8,53. The risk of
after treatment43. Furthermore, SAI has been reported AI is lowest with short-​term, low-​dose glucocorticoid
in 26–57% patients with hypophysitis44,45, whereas in use and highest with long-​term, high-​dose glucocorti-
pituitary stalk lesions of any kind, 23% of patients may coid use8. However, the correlation between TAI and
develop SAI46. glucocorticoid dose or treatment duration is weak,
In addition, ICIs, which are used in various cancer indicating that additional factors might influence indi-
therapies, have been associated with adverse effects on vidual susceptibility8,54. Every patient on glucocorti-
Hypopituitarism the HPA axis. ICIs were associated with an increased inci- coid pharmacotherapy has to be regarded as at risk of
Dysfunction of the pituitary
with impaired secretion or a
dence of serious grade SAI (OR 3.19, 95% CI 1.84–5.54) TAI and, therefore, the dosage needs to be carefully
lack of secretion of one or and hypophysitis (OR 4.77, 95% CI 2.60–8.78)47. The tapered and the patient counselled. This recommen-
more pituitary hormones. observed incidence of hypophysitis was highest in dation is further supported by the increased mortality

4 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

observed within the first 3 months following cessation (for example, compounds that induce CYP3A4) has been
of glucocorticoid therapy55. Recovery of the function of associated with the induction or aggravation of corti­
the HPA axis following cessation of glucocorticoid ther- sol deficiency. Although 3% of all adults are using oral
apy is observed within 24 months in most patients but glucocorticoids leading to the suppression of cortisol
recovery may take >4 years in some patients56–58. secretion, the risk of adrenal crisis is generally low78.

Adrenal crisis Precipitating factors. Adrenal emergencies are the


Prevalence and mortality. In those with known AI, consequence of severe cortisol deficiency or relative
adrenal crises have an incidence of 5–10 events per cortisol deficiency in the time of a major stress, in par-
100 patient-​years11,14,59–61 and are more frequent in patients ticular when associated with acute inflammation such as
with PAI than in those with SAI10,14,61,62, presumably sepsis10–12,69,73. Gastroenteritis is a frequent precipitant of
owing to the lack of aldosterone and more severe hypo­ adrenal crisis owing to the reduced absorption of orally
cortisolism. The mortality from adrenal crises is reported administered glucocorticoids in these patients. However,
to be 0.5 per 100 patient-​years11. Mortality from adre- many symptoms of adrenal crisis are abdominal and,
nal emergencies is rare (<1%) after hospital admis- therefore, the diagnosis of gastroenteritis as a cause may
sion, although fatalities from undiagnosed AI may be be difficult. In any case, treatment of gastroenteritis in
missed63,64. The prognosis in patients with AI is gener- AI and of adrenal crisis accompanied by gastrointestinal
ally good if treatment is adequate. However, standard- symptoms is the same — timely parenteral administra-
ized mortality rates are approximately double compared tion of glucocorticoids. Gastroenteritis is the most com-
with controls65,66. Younger patients (that is <40 years of mon precipitating factor when a precipitant is found,
age) were found to be at increased risk of premature although an obvious precipitating factor is lacking in
death67. Adrenal crisis contributes to 15–40% of reported ~10% of cases11. Temporary reductions in hydrocorti-
AI-​related deaths10,65,67,68. sone availability may increase the incidence of adrenal
crises, even when alternative glucocorticoid preparations
Risk factors. Risk factors for adrenal crisis include previ- are available79. In patients with AI, psychological stress
ous crises69, older age (>65 years of age) or younger age can also elicit an adrenal crisis under exceptional cir-
(adolescence and young adulthood)70–73, and comorbidi- cumstances, such as bereavement. Casual dose increases
ties such as diabetes mellitus, diabetes insipidus or other of glucocorticoid replacement because of perceived low-​
non-​endocrine diseases14,69. The use of low-​dose, short-​ level psychological stress should, however, be avoided as
acting glucocorticoids70,74,75 might potentially underlie this could result in chronic over-​replacement.
the increased incidence of adrenal crisis75. In addition,
in patients with thyroid disease, treatment initiation with Mechanisms/pathophysiology
l-​thyroxine and onset of hyperthyroidism owing to The adrenal cortex is under the control of two different reg-
Graves disease can precipitate a crisis through a more ulatory circuits, the HPA axis and the renin–angiotensin–
rapid inactivation of cortisol than in people without aldosterone system (RAAS). In PAI (except CAH),
thyroid disease76,77. Treatment with drugs that reduce all adrenocortical zones are affected, resulting in the
cortisol production (for example, steroidogenesis inhib- deficiency of all adrenocortical hormones. However,
itors) or increase the metabolic clearance of cortisol in SAI, hormone secretion specifically from the zona

Table 2 | Causes of secondary adrenal insufficiency


Aetiology Pathogenesis Clinical manifestations and
diagnostic tools
Tumours Pituitary adenoma, craniopharyngioma, metastasis (carcino- Pituitary MRI, screen for hormone
mas of breast, lung, colon, prostate), chondroma, chordoma, deficiencies
germinoma, suprasellar meningioma, astrocytoma of the optic
nerve, ependymoma, pituitary carcinoma, lymphoma
Iatrogenic Pituitary surgery, radiotherapy Pituitary MRI, screen for hormone
deficiencies
Traumatic Traumatic brain injury, postpartum apoplexy (Sheehan Severe headache, visual disturbances,
and vascular syndrome), tumour apoplexy, subarachnoidal bleeding, pituitary MRI, screen for hormone
aneurysm, snake bite deficiencies
Autoimmune T and B cell autoimmunity against pituitary cells Pituitary MRI, confirmation of subtype
(hypophysitis), IgG4-​related hypophysitis by transsphenoidal biopsy, screen for
hormone deficiencies
Genetic Mutations in transcription factors, hormone and hormone Sequence of relevant gene
receptor genesa
Infiltrative Sarcoidosis, histiocytosis, haemochromatosis, amyloidosis Pituitary MRI, specific diagnostic tests
Infectious Tuberculosis, HIV infection, meningitis, actinomycosis, syphilis Pituitary MRI, specific diagnostic tests
Medication Checkpoint inhibitors, interferon-​α, opioids Check medication and patient history
See also Table 3. Table adapted from refs31,155.
a


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 5

0123456789();:
Primer

fasciculata and zona reticularis is impaired owing to a Besides the circadian rhythm of cortisol secretion,
lack of ACTH stimulation, whereas aldosterone secre- ultradian peaks of cortisol occur, which are produced
tion from the zona glomerulosa remains intact (Fig. 2). by a pituitary–adrenal feedback regulation, independent
In this section, we summarize the current knowledge of of the CRH pulses. As cortisol is not stored but must be
pathophysiology of the different types of AI. synthesized upon ACTH stimulation, there is a delay
before the peak secretion of cortisol. This time delay
Physiology of the HPA axis that occurs before the negative feedback on ACTH
The HPA axis is the anatomical unit that regulates the production introduces the ultradian rhythmicity 83.
physiological secretion of cortisol. The regulation of Oscillating glucocorticoid levels have been shown to
cortisol secretion is complex and evident on multiple have an impact on the physiological regulation at the
levels in the HPA axis (for review see ref.80) (Fig. 2). cellular level in many organs. Hence, disruptions in
Circadian input from the suprachiasmatic nucleus in the pulsatile signal of cortisol induces changes in meta-
the hypothalamus as well as stress stimuli from other bolic processes, behaviours, affective state and cognitive
brain regions converge on the paraventricular nuclei of function84.
the hypothalamus, which stimulates the release of CRH
and vasopressin80. CRH travels via the portal veins of the Functions of cortisol. Cortisol is a key modulator of
median eminence to the anterior pituitary and induces physiological processes such as energy metabolism,
a secretory pulse of ACTH into the general circulation. stress response, reproduction, immunity and cognition,
The circulating ACTH binds to and stimulates the and exerts its actions by interacting with the glucocor-
ACTH receptor (also known as MC2R) in the adrenal ticoid receptor and mineralocorticoid receptor85. Upon
cortex, with subsequent release of cortisol80 (Fig. 2). binding, the cortisol–receptor complex translocates to
Cortisol is secreted in pulses of various amplitudes the nucleus and binds to different glucocorticoid and
every 60–90 min creating a tightly regulated circadian mineralocorticoid response elements, thereby modulat-
pattern with elevated secretion in the early hours of the ing gene transcription and repression86 to mediate the
morning prior to waking and a nadir around bedtime diverse physiological functions of cortisol87. The ultra-
in the late evening81. Cortisol levels are lowest within dian oscillation of cortisol enables cyclic glucocorticoid-​
an hour of sleep and increase with a peak shortly after mediated transcriptional regulation. Studies in mouse
awakening. Further short-​term increases in cortisol cell lines and rat models have clearly demonstrated
occur subsequent to specific stimuli, such as meals markedly different transcription patterns depending
and everyday life stressors. Cortisol peaks are generally on ultradian glucocorticoid stimulation or constant
preceded by a secretory surge of pituitary ACTH caused glucocorticoid stimulation, indicating that ultradian
by the corticotropic cells (Fig. 3). Cortisol provides a neg- rhythmicity is of physiological importance88. In humans,
ative feedback loop by binding to glucocorticoid recep- disruption of ultradian rhythmicity of plasma cortisol
tors in the hypothalamus and the pituitary gland, which has been shown to negatively impact emotional and
inhibits ACTH secretion82. cognitive responses84,89.
Most of the cortisol (>95%) is found in the circulation
10 mg HC 5 mg HC 2.5 mg HC bound to corticosteroid-​binding globulin (CBG). The
fraction of free bioavailable cortisol depends on the cor-
800
Night time sleep Night time sleep
tisol binding affinity of CBG, which varies depending on
700 multiple factors, such as body temperature or inflamma-
tion. In the presence of fever or inflammation, decreased
600 CBG affinity might increase the amount of bioavailable
Serum cortisol (nmol/l)

cortisol90,91.
500

400
Renin–angiotensin–aldosterone system
The RAAS is an important hormonal cascade, which is
300 a critical regulator of electrolyte balance, blood volume,
systemic vascular resistance and, consequently, blood
200 pressure. The RAAS is also the primary regulator of
aldosterone secretion from the adrenal cortex, with angi-
100
otensin II and potassium being the principal proximate
0 regulators (Fig. 4). Indeed, increases in potassium blood
1500 1900 2300 0300 0700 1100 1500 1900 2300 0300 0700 levels physiologically stimulate aldosterone secretion and
Time ( 24-h clock) hypokalaemia results in reduced aldosterone secretion92.
The enzyme renin is released by the kidneys and cataly-
Cortisol circadian rhythm Thrice-daily hydrocortisone
ses the conversion of angiotensinogen to angio­tensin I,
which is subsequently converted to angiotensin II by the
Fig. 3 | The physiological rhythm of cortisol and pharmacokinetic profile of
hydrocortisone therapy. Circadian rhythm of cortisol showing early morning peak and
angiotensin-​converting enzyme. Angiotensin II binds
pulses at meal times overlaid with cortisol profile of thrice-​daily hydrocortisone (HC) to angiotensin receptors and stimulates the release
replacement therapy. Dashed lines denote oral intake of conventional hydrocortisone at of aldosterone from the adrenal cortex. The binding of
doses of 10 mg in the morning, 5 mg at noon and 2.5 mg in the afternoon. Adapted with angiotensin II to its receptors induces further effects
permission from ref.184, Oxford University Press, and ref.271, Wiley. such as vasoconstriction. Renin secretion is stimulated

6 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

Adrenal gland
Kidney

Blood sodium concentration Blood potassium Sympathetic nerve


Macula densa mechanism concentration stimulation

Activation of Aldosterone Renal sodium retention Renal perfusion Juxtaglomerular Renin


AT1 receptor release (and potassium excretion) pressure cells release
Circulating blood volume

Vasoconstriction

Angiotensin II Angiotensin I Angiotensinogen

Angiotensin-converting enzyme

Fig. 4 | The renin–angiotensin–aldosterone system. The main stimulus of aldosterone secretion is mediated via the
renin–angiotensin–aldosterone system. Renin secretion is stimulated by decreased renal perfusion pressure (for example,
reduced blood volume, reduced sodium or increased potassium blood levels) and sympathetic nerve stimulation.
Renin catalyses the conversion of angiotensinogen to angiotensin I, which is converted to angiotensin II by the
angiotensin-converting enzyme (ACE). Binding of angiotensin II to AT1 receptor stimulates the release of aldosterone
from the adrenal cortex and induces further cardiovascular effects such as vasoconstriction. Renin release is mainly
inhibited by negative feedback of angiotensin II.

by decreased renal perfusion pressure, reduced blood Lack of aldosterone leads to loss of sodium in
sodium concentrations in the body (sensed by macula the body, with decreased intravascular fluid vol-
densa cells), increased blood potassium load, and sym- ume and hypotension104. The loss of negative feed-
pathetic nerve stimulation of juxtaglomerular cells. Renin back by cortisol in Addison disease results in
release is inhibited by negative feedback of angioten- increased release of pro-opiomelanocortin and pro-
sin II or by atrial natriuretic peptide, which is released opiomelanocortin-derived peptides such as ACTH and
from the cardiac muscle cells of the atria in response to α-melanocyte stimulating hormone (α-​MSH). The
increased stretching of the atrial wall owing to increased increase in α-MSH levels leads to skin hyperpigmen-
atrial blood volume93. ACTH acutely stimulates aldoster- tation through stimulation of MSHR (also known as
one secretion and conversely glucocorticoids suppress melanocortin receptor 1) not only in areas of the body
aldosterone secretion94–96. exposed to sun but also in areas of increased mechanical
friction, such as palmar creases, scars, the groin and the
Macula densa cells
Specialized cells lining the wall
Primary adrenal insufficiency oral mucosa1.
of kidney tubules that detect Addison disease. In Addison disease, autoimmune
changes in distal tubular fluid destruction of adrenocortical tissue is believed to be Congenital adrenal hyperplasia. Congenital AI results
composition and transmit caused by autoantibodies to the most frequently observed from mutations in genes involved in the HPA axis
signals to the glomerular
autoantigen, 21-​hydroxylase97. These autoantibodies are (Table  3) . CAH, the most common genetic form of
vascular elements.
detected in most patients with Addison disease98, even PAI, comprises a group of disorders of adrenal steroid
Juxtaglomerular cells preceding overt adrenal failure by many years99 and, biosynthesis. In ~95% of patients, CAH is caused by
Specialized cells in the kidney therefore, are a highly specific and sensitive biomarker. mutations in CYP21A2 (ref.105); the resultant cortisol
that synthesize, store and Circulating CD4+ and CD8+ T cells with autoreactivity deficiency stimulates increased ACTH secretion from
secrete the enzyme renin.
against 21-​hydroxylase that are found in patients are the pituitary, leading to adrenocortical hyperplasia and
Salt-​wasting syndrome thought to mediate the destruction of the adrenal increased secretion of adrenal androgens upstream
A more severe form of cor­tex100,101 (Fig. 5). Studies have shown that Addison of the enzymatic block, driving increased androgen
congenital adrenal hyperplasia disease is associated with certain major histocompati- production (Fig. 6).
characterized by insufficient
bility complex (MHC) genotypes such as DR3-​DQ2 and The severity of CAH depends on the severity of the
production and action of
aldosterone that lead to renal
DR4-​DQ8 (refs97,102). In addition, variants in CTLA4, disease-​causing mutations and the residual enzyme
sodium loss and consequent PTPN22, CIITA and CLEC16A, which are also impli- activity. Neonates with <2% of the enzyme activity
sodium depletion of the body. cated in other autoimmune diseases, have been shown to will have both cortisol and aldosterone deficiency30.
If undiagnosed, dehydration, increase individual susceptibility to Addison disease102. In approximately two-​thirds of patients, impaired aldos-
hypotension, failure to
thrive, hyponatraemia
Furthermore, heritability of Addison disease is high and terone synthesis leads to a potentially life-​threatening
and hyperkalaemia occur similar to that of coeliac disease. In a twin study, 97% salt-​wasting syndrome and in one-​third of patients, a mild
within days of birth. of disease liability was attributable to genetic factors103. virilizing form.


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 7

0123456789();:
Primer

Adrenal cortex Local draining lymph node


Adrenal
cortex cell Antigen
Virus MHC II TCR
Dendritic Co-stimulation
cell
TH1
Necrosis cell
IFNγ
Uptake of
antigens
IL-12 Activation of
IL-2 autoreactive
Cortisol TH cells
production
Autoantibodies
Activation and
expansion of
autoreactive
TC cell TC cells B cell

BCR

Oxygen free
radicals
Activated Activation of
macrophage autoreactive B cells

Fig. 5 | Mechanisms underlying Addison disease. Addison disease is characterized by destruction of the adrenal cortex
by T cell-​mediated autoimmune mechanisms with contributions from dendritic cells and macrophages. Activation of
B cells in local draining lymph nodes leads to production of autoantibodies against the enzyme 21-​hydroxylase, which is
detected in ~90% of patients with Addison disease. Circulating CD4+ and CD8+ T cells targeting 21-​hydroxylase-​expressing
corticotrophs mediate the destruction of the adrenal cortex. The environmental triggers of Addison disease are not
completely known, but viruses might be involved. BCR, B cell receptor; IFNγ, interferon-​γ; MHC, major histocompatibility
complex; TC cell, cytotoxic T cell; TCR, T cell receptor; TH cell, helper T cell. Adapted with permission from ref.272, Elsevier.

In the salt-​wasting form of CAH, the 46,XX female of steroid-​secreting cells occurs in adrenal and gonadal
Virilization neonate presents with virilization of the external genitalia tissues. Adrenal failure may also occur in isolation; in
A condition in which a
and both sexes may present with salt-​wasting adrenal one series, 8% of young boys with Addison disease had
female develops masculine
characteristics such as excess
crisis, typically in the second and third week of life, underlying X-​linked adrenoleukodystrophy88. Other
facial and body hair, acne, if steroid replacement is not initiated promptly after congenital forms of PAI are typically rare in the general
increased muscle mass and birth30. Patients with less severe mutations present early population and are discussed in detail elsewhere36,109,110
baldness. Virilization is caused or late in childhood with signs of virilization, as well as (Table 3).
by excess production of
endogenous androgens from
growth arrest and precocious pseudopuberty. Mild forms
the adrenals or the ovaries or of CAH with at least 30–50% residual enzyme function Secondary adrenal insufficiency
from exogenous androgen are more prevalent and are subsumed under the term In SAI, the diminished or total lack of ACTH pulses
administration. non-​classic CAH (formerly also known as ‘late-​onset’ results in a loss of adrenal cortisol synthesis and secre-
CAH or cryptic CAH), which is characterized by adre- tion. In contrast to PAI, in which loss of adrenal function
Precocious pseudopuberty
Partial pubertal development
nal androgen excess, mild or absent glucocorticoid often is near complete by the time of clinical presenta-
that results from premature deficiency and normal mineralocorticoid biosynthesis29. tion, all degrees of severity of AI are observed in SAI
appearance of secondary depending on residual ACTH secretion. Furthermore,
sexual characteristics in Other congenital forms of PAI. Next to CAH, the most mineralocorticoid production is usually intact in
prepubertal boys and girls
owing to excess production
prevalent genetic form of PAI is X-​linked adrenoleu- patients with SAI as aldosterone production is primar-
of sex steroids. kodystrophy. Adrenoleukodystrophy is a peroxisomal ily controlled by the RAAS hormonal cascade (which
disorder caused by mutations in ABCD1 that lead requires a functioning adrenal gland and kidney) and
Leydig cells to accumulation of very-​long-​chain fatty acids in all controlled only to a minor, transient degree by ACTH95.
Steroidogenic cells in
tissues, especially the central nervous system, the SAI can manifest as an isolated entity, but SAI often
the testes that produce
testosterone upon stimulation
adrenal cortex and the Leydig cells of the testes106,107. presents in association with the lack of other pitui-
by pituitary luteinizing The incidence of adrenoleukodystrophy is 1 in 17,000 tary hormones. In addition to pituitary tumours, the
hormone. newborns (typically males and very rarely females)108. hypothalamic–pituitary unit can also be damaged by
Neurological symptoms can manifest in childhood with head trauma; up to one third of patients with traumatic
Spastic paraparesis
A group of rare hereditary
central demyelination and rapid neurological decline or brain injury develop chronic pituitary dysfunction, most
disorders that cause gradual in young adults with mixed motor and sensory neuro­ frequently with growth hormone deficiency, followed by
weakness with muscle spasms. pathy leading to spastic paraparesis. Progressive failure gonadotrophin and ACTH deficiency111.

8 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

Corticotroph
Autoimmune hypophysitis, another prominent cause pituitary hormone deficiency121. Patients with PROP-1
Cells in the anterior of SAI, is characterized by infiltration of lymphocytes mutations demonstrate ACTH deficiency; however, it
pituitary that produce into the pituitary gland. It frequently develops in the is unknown whether ACTH deficiency in these patients is
pro-​opiomelanocortin, which peripartum period and can lead to the development of a consequence of corticotroph attrition or pituitary hyper-
undergoes cleavage to form
a mass lesion in the pituitary owing to heavy lympho- trophy, followed by pituitary degeneration. Congenital
adrenocorticotropic hormone
(ACTH) in response to cytic infiltration112. Isolated deficiency of single pituitary forms of SAI often demonstrate atypical pheno­typic
stimulation by hypothalamic hormones may occur, for example, as isolated ACTH presentation, such as post-​pubertal presentation or adult
corticotropin-​releasing deficiency, but more often, multiple pituitary hormone progression of hormonal deficiencies122.
hormone.
deficiencies are observed in patients. Autoimmune
hypophysitis has been reported as an important adverse Adrenomedullary function in AI
effect of ICIs, especially ipilimumab (an anti-​CTLA4 In patients with PAI and SAI, besides a lack of cortico­
antibody)113. Besides autoimmune hypophysitis and steroids, studies have found reduced adrenomedullary
ICI-​induced hypophysitis, IgG4-​related disease also epinephrine production during normal physiological
leads to IgG4-​related hypophysitis owing to the infiltra- conditions and in response to stress123–129. By contrast,
tion of IgG4-​positive plasma cells into the pituitary gland norepinephrine levels were increased in the majority of
alongside infiltration of other organs114,115. Furthermore, patients. Although of distinct developmental origins,
9–15% of patients on long-​term opioid therapy develop the adrenal cortex and the adrenal medulla are func-
SAI50,51,116 probably owing to blunted ACTH responses, tionally closely interconnected. For example, activity of
loss of diurnal rhythm and negative effects of opioid on the enzyme PNMTase, which catalyses the conversion
adrenal glucocorticoid production. Long-​term opioid of norepinephrine to epinephrine, requires high local
analgesia therapy also impairs the hypothalamic– glucocorticoid concentrations from the adrenal cortex
pituitary–gonadal axis, with secondary hypogonadism through a rich blood supply130,131. These intra-​adrenal
observed in 75% of men and 21% of women50,51,117,118. cortisol levels cannot be achieved by standard glucocor-
ticoid replacement therapy. Nevertheless, the clinical
Congenital forms of SAI. Congenital SAI is very rare and relevance of reduced epinephrine levels and diminished
usually results from abnormalities in transcription factors epinephrine response under stressful conditions is yet
involved in the differentiation of anterior pituitary cells to be understood. High levels of norepinephrine and
(for review see refs119,120) (Table 3). For example, mutations respective activation of adrenergic receptors may at
in PROP-1 and TPIT have been shown to result in ACTH least partly compensate for the epinephrine deficiency
deficiency. Mutations in PROP-1 are the most common regarding sympathomimetic effects129. Epinephrine
cause of familial and sporadic congenital combined deficiency has been hypothesized to contribute to the

Table 3 | Genetic causes of adrenal insufficiency


Causes Mechanisms
Primary adrenal insufficiency
Congenital adrenal hyperplasia 21-​Hydroxylase deficiency (>95% of patients)
Rare causes in order of prevalence: 11β-​hydroxylase deficiency,
P450 oxidoreductase deficiency, 17α-​hydroxylase deficiency,
3β-​hydroxysteroid dehydrogenase type 2 deficiency
Congenital lipoid adrenal hyperplasia STAR mutations
Adrenoleukodystrophy (X-​linked) ABCD1 and ABCD2 mutations
Adrenal hypoplasia congenita NR0B1 (DAX1) mutations, deletion of genes linked with Duchenne
muscular dystrophy, glycerol kinase, NR5A1 mutations
Autoimmune polyglandular syndrome type 1 AIRE mutation
IMAGe syndrome CDKN1C mutations
Kearns–Sayre syndrome Mitochondrial DNA deletions
Lysosomal acid lipase deficiency LIPA mutations
(Wolman disease)
Sitosterolaemia ABCG5 and ABCG8 mutations
Familial glucocorticoid deficiency or Mutations in MC2R, MRAP, MCM4 or STAR
corticotropin insensitivity syndromes
Triple A syndrome (Allgrove syndrome) AAAS mutations
Secondary adrenal insufficiency
Congenital isolated corticotropin deficiency TBX19 mutations
Combined pituitary hormone deficiency Mutations in GLI2, HESX1, OTX2, LHX4, LHX3, PROP1, SOX3 or POU1F1
Prader–Willi syndrome Deletion or silencing of genes in imprinting centre for the syndrome
Table adapted from refs36,120,109,110.


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 9

0123456789();:
Primer

Adrenarche reduced general well-​being and impaired vitality, which been identified. The various effects of DHEA can be
Maturation of the zona might further impair resistance to stress. The effects of categorized into direct and indirect139. Indirect effects
reticularis of the adrenal epinephrine deficiency may be more pronounced in are mediated via conversion of these major adrenal sex
resulting in increased patients with classic CAH and in patients with other con- steroid precursors into active androgens and oestrogens
production of adrenal
androgens.
genital forms of AI, as cortisol deficiency may already in the placenta, gonads and peripheral target cells. The
have affected adrenal medullary development 125,132. enzymes involved in the conversion of DHEA into active
Clinical studies have suggested that the lack of epi- sex steroids are expressed in different tissues, enabling
nephrine in patients with AI results in reduced glucose local intracrine action of DHEA, independent of changes
counter-​regulatory response to hypoglycaemia and in in blood hormone levels140. DHEA has been shown to
cognitive impairment133,134. So far, however, the exact non-​specifically bind to cell membranes or receptors.
clinical relevance of catecholamine deficiency in patients Direct actions of DHEA include neuroprotective and
with AI remains to be established. anti-​depressive effects within the brain (for example, via
binding to GABA(A) receptor subunit α-6 and GluN2A)
Adrenal androgens and immunomodulatory effects by interaction with
Although highly abundant in human circulation, the immune cells139,141. The observed impairment of general
physiological role of adrenal androgens and precursors, well-​being in patients with AI has in part been associated
DHEA and dehydroepiandrosterone sulfate (DHEAS), is with lack of neurosteroidal stimulatory activity of DHEA
still not completely understood. In contrast to glucocorti- and androgens139,142.
coid and mineralocorticoid deficiency, the lack of DHEA
or DHEAS is not life-​threatening. In PAI (except CAH), Adrenal crisis
DHEA secretion is impaired owing to the destruction The pathophysiology of adrenal crisis, a potentially fatal
of the zona reticularis and, in SAI, owing to impaired condition caused by acute cortisol deficiency, is not yet
ACTH stimulation. The lack of DHEA results in pro- well understood10. Glucocorticoids have been suggested
nounced androgen deficiency in woman, whereas in to influence stress by permissive, suppressive and stim-
men, androgen production is preserved by testicular ulating actions143. It has been hypothesized that patients
testosterone secretion. with untreated AI have a lack of permissive action of
The physiological secretion of DHEA evolves over glucocorticoids, leading to impaired responsiveness
the lifetime of an individual135,136. DHEA levels increase of the cardiovascular system143–145. However, stable replace-
in the prepubertal years, especially during adrenarche, ment therapy in patients with chronic AI is thought to
reaching peak levels at the end of the third decade of ensure sufficient permissive actions of glucocorticoids,
life, followed by a continuous decline with advancing such as providing sufficient sensitivity to catecholamines
age, supporting a role for DHEA in the reproductive in response to stress146,147. Furthermore, in patients with
system137,138. A specific DHEA receptor has not yet PAI, the hyponatraemia and fluid retention caused by

Cholesterol DHEAS

Androgens (zona reticularis)

Pregnenolone 17-Hydroxy-pregnenolone DHEA Androstenediol

Progesterone 17-Hydroxy-progesterone Androstenedione Testosterone

21-hydroxylase deficiency
11-Deoxycorticosterone 11-Deoxycortisol
Glucocorticoids
(zona fasciculata)
Corticosterone Cortisol

18-Hydroxy-corticosterone
Mineralocorticoids
(zona glomerulosa)
Aldosterone

Fig. 6 | Adrenal steroidogenesis and 21-hydroxylase deficiency. Steroidogenesis in the adrenal cortex is catalysed by a
series of steroidogenic enzymes. Cholesterol is the basic substrate that is converted into mineralocorticoids, glucocorticoids
and sex steroids in the respective adrenocortical zones. Congenital adrenal hyperplasia is caused most frequently by
mutations in CYP21A2 (encoding the enzyme 21-​hydroxylase). This enzyme converts 17-​hydroxy-​progesterone to
11-​deoxycortisol and progesterone to 11-​deoxycorticosterone, which are precursors for cortisol and aldosterone. Impaired
cortisol synthesis leads to increased adrenocorticotropic hormone stimulation of the adrenal cortex, with accumulation
of steroid precursors upstream of the enzyme block and increased conversion to sex steroids, mainly androgens.
DHEA, dehydroepiandrosterone; DHEAS, dehydroepiandrosterone sulfate. Adapted with permission from ref.273, Elsevier.

10 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

Adrenal gland
Stress
Medulla

Zona Zona Epinephrine deficiency


glomerulosa fasciculata
Vasodilation, impaired
effect of catecholamines

Aldosterone Cortisol
deficiency deficiency Hypotension

Capillary

Electrolyte and volume Metabolism Inflammation Increased capillary


regulation permeability

Kidney Adipose Muscle Liver Immune cells


TNF
IL-1
IL-6
Reduced sodium reabsorption
Increased potassium reabsorption Decreased Increased Glucocorticoid
gluconeogenesis inflammatory receptor insensitivity
Reduced cytokines
Hyponatraemia production of Reduced amino
Hyperkalaemia free fatty acids acid liberation Hypoglycaemia Glucocorticoid
resistance

Toxic effects of
uncontrolled Aggravation of
Hypotension Decreased energy reserves inflammation cortisol deficiency

Fig. 7 | Pathophysiological aspects of adrenal crisis. An imbalance between demand and availability of glucocorticoids
may result in adrenal crisis. This state is characterized by a modified immune response owing to the lack of glucocorticoid-​
mediated immunomodulation, leading to increased inflammatory cytokines further activating pro-​inflammatory pathways.
The cortisol deficiency is further aggravated by a cytokine-​induced glucocorticoid receptor resistance. Furthermore,
cortisol deficiency leads to reduced responsiveness to catecholamines in blood vessels, aggravating the effects of volume
depletion caused by aldosterone deficiency in primary adrenal insufficiency. Cortisol deficiency additionally results in an
impaired mobilization of energy resources owing to reduced hepatic gluconeogenesis, reduced muscular amino acid
liberation and reduced production of fatty acids.

mineralocorticoid deficiency and volume depletion Tnfr1a-​null and Tnfr1b-​null mice and mice treated with
might worsen hypotension, which is further aggravated anti-​TNF serum were resistant to the lethal effects of
by vomiting and diarrhoea148. lipopolysaccharide153. Thus, studies implicate TNF as a
The suppressive activity of glucocorticoids pre- key mediator of adrenal crisis. In healthy individuals,
vents the harmful effects of an overshooting immune pyrexia (fever), infection and other stressful condi-
defence 143,149 , potentially primarily mediated by tions induce an increase in circulating cortisol levels
pro-​inflammatory cytokines. Infections trigger the by increased adrenal production or reduced cortisol
release of cytokines such as IL-1, TNF and IL-6. These breakdown owing to diminished expression or activity of
cytokines physiologically stimulate the HPA axis and, cortisol-​metabolizing enzymes154. In addition, increases
therefore, increase systemic cortisol concentrations150. in systemic and free bioavailable cortisol are facilitated
High cortisol levels in turn inhibit cytokine release by neutrophil elastase cleavage of CBG and pyrexia90.
and action, thereby preventing their detrimental Thus, it is recommended that an increase in cortisol
effects151,152. In a murine model of adrenal crisis153, adre- levels caused by stressful conditions be mimicked by
nalectomy increased sensitivity to the lethal inflam- carefully adjusting the hydrocortisone dose in patients
matory effects of TNF, whereas adrenalectomized with AI155 (Fig. 7).


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 11

0123456789();:
Primer

Central nervous system Neuropsychiatric system


• Anorexiaa • Depressiona,c
• Weight loss a
• Fatiguea
• Nausea a
• Reduced libidoc
• Salt craving b

• Dizzinessb
Gastrointestinal system
• Diarrhoea
Cardiovascular system • Vomitinga
• Hypotension and/or • Abdominal paina
dehydrationb

Dermatological system
Haematological system • Dry skinc
• Anaemia a
• Hyperpigmentation (PAI)
• Lymphocytosis • Hypopigmentation (SAI)
• Eosinophiliaa • Loss of pubic/axillary hairc

Blood electrolytes Musculoskeletal system


• Hyponatraemiaa,b • Myalgiaa
• Hyperkalaemiab • Joint pain
• Hypercalcaemiaa • Weaknessa

Fig. 8 | Clinical manifestations of adrenal insufficiency. Deficiency of adrenocortical hormones affects all systems in
the human body leading to a wide range of symptoms. Frequent symptoms and their main associations with the respec-
tive deficiency of glucocorticoids, mineralocorticoids and adrenal androgens are demonstrated. PAI, primary adrenal
insufficiency; SAI, secondary adrenal insufficiency. aSymptoms specific to glucocorticoid deficiency; bSymptoms
specific to mineralocorticoid deficiency; cSymptoms specific to adrenal androgen deficiency. Adapted from ref.274,
Springer Nature Limited.

Diagnosis, screening and prevention Besides the above-​mentioned symptoms of glucocor-


Presentation ticoid deficiency and androgen deficiency, in patients
Adrenal crisis may be the first presentation of AI as the with SAI, there is often complete hypopituitarism with
signs and symptoms of mild and advancing chronic glu- growth impairment, and gonadal and thyroid hormone
cocorticoid deficiency are often non-​specific. Hence, dysfunction, depending on the underlying aetiology. The
these non-​specific signs are typically not recognized onset of SAI can be acute, although chronic development
as AI by health-​care personnel, leading to diagnostic might happen in certain individuals. As pituitary adeno-
delays or misdiagnosis. The symptoms of chronic corti- mas are one of the most common causes of SAI, SAI can
sol deficiency include fatigue, reduced energy and appe- be the initial clinical presentation of a pituitary tumour.
tite, stomach ache or diffuse abdominal pain, pain in
joints and muscles, weight loss and increased cold sen- Diagnosis
sitivity (Fig. 8). Skin hyperpigmentation is a specific sign Primary adrenal insufficiency. The awareness of PAI is
indicative of PAI, but the severity of hyperpigmentation crucial, especially owing to the fact that most patients
differs widely between individuals. As a consequence of are diagnosed only after years of non-​specific symptoms
the associated autoimmune disorders in PAI, additional such as fatigue, poor well-​being, postural dizziness, nau-
symptoms and signs specific to the comorbidities may be sea and weight loss, which are frequently attributed to
present in patients15. Low blood pressure with increased other causes9. Treatment of suspected acute manifesta-
postural drop and salt craving might be signs of aldos- tion of AI should never be delayed by performing (or
terone deficiency in patients with PAI. DHEA deficiency awaiting the results of ) diagnostic procedures. The
is usually associated with loss (or lack of development) diagnostic test for suspected PAI is a paired measure-
of axillary and pubic hair, dry and itchy skin, reduced ment of serum cortisol and plasma ACTH. A morning
energy and loss of libido in women. serum cortisol of <140 nmol/l (5 µg/dl) in combination
Clinical manifestations of AI in childhood include with increased ACTH levels (twice the upper normal
failure to thrive, recurrent infections, family history of limit) is confirmative of PAI. Cortisol concentrations
neonatal deaths or early postnatal deaths, ambiguous vary according to the assay used and, therefore, check-
genitalia at birth and hepatitis156. ing the reference ranges with the relevant laboratory
In routine laboratory testing, hyponatraemia, hyper- is recommended158. Mass spectrometry analyses have
kalaemia and hypoglycaemia specifically can increase been shown to detect lower cortisol concentrations than
clinical suspicion for PAI; hypercalcaemia is rarely immunoassays owing to enhanced assay specificity159.
observed in patients1,157. In individuals whose morning cortisol levels are

12 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

ACTH 1–24 stimulation test ≥140 nmol/l, an ACTH 1–24 stimulation test (also known standard test for confirming the diagnosis of SAI. The
Also known as the Synacthen as the short Synacthen test or corticotropin-​stimulation insulin tolerance test can determine the integrity of
test or corticotropin-​ test) using a 250-​μg dose of synthetic ACTH 1–24 can the HPA axis by inducing a severe hypoglycaemic state,
stimulation test. This test be useful to assess the proper functioning of the adre- which activates the stress hormone axis and all insulin
is commonly used for the
assessment of adrenal
nal glands. A peak serum cortisol concentration of counter-​regulating hormones (for example, cortisol and
cortisol production. Cortisol <450 nmol/l 30 min after ACTH stimulation or a peak growth hormone)168. The insulin tolerance test involves
serum levels are measured concentration of <500 nmol/l 60 min after ACTH stim- the administration of an acute bolus dose of intravenous
before and 30–60 min after ulation establishes a positive diagnosis of AI160–162. These insulin to induce symptomatic and biochemical hypo-
supraphysiological stimulation
cut-​off values were developed using immunoassays glycaemia (blood glucose ≤40 mg/dl or 2.2 mmol/l), with
with synthetic 1–24 ACTH.
having high cross-​reactivity with non-​cortisol steroids, serum cortisol and plasma ACTH measured at frequent
leading to a higher response than assays with less cross-​ intervals for 120 min. Owing to hypoglycaemia, the test
reactivity or with mass spectrometry. Therefore, future is usually uncomfortable for the patient and requires
research establishing criteria and cut-​off values with cur- a high degree of supervision, especially as the induc-
rent new assays and methods is warranted. Combined tion of hypoglycaemia is unsafe in patients with or at
plasma renin and aldosterone levels should be measured risk of epilepsy, or cerebrovascular or cardiovascular
to determine the presence of mineralocorticoid defi- disorders. The diagnosis of SAI is ruled out if serum
ciency, indicated by low or low–normal aldosterone in cortisol peak values in the insulin tolerance test reach
the presence of increased levels of renin. at least 550 nmol/l (ref.1) or, as stated by some authors,
Once a diagnosis of PAI is established, the aetiol- >500 nmol/l (refs56,165,169,170).
ogy should be investigated. In geographical regions Owing to the low risks and the ease of testing, the
with a high prevalence of Addison disease, patients ACTH 1–24 stimulation test described above is the most
should be evaluated for the presence of 21-​hydroxylase widely performed test for SAI165,167. A peak serum cor-
autoantibodies in the serum. If patients test positive tisol of >550 nmol/l is regarded as a normal response
for autoantibodies, they should be screened for con- (although assay-​dependent). The loss of adrenal cor-
comitant autoimmune comorbidity. Negative testing tisol response subsequent to manifestation of ACTH
for 21-​hydroxylase autoantibodies does, however, not deficiency develops over time and, therefore, the short
exclude autoimmune PAI. In patients with a negative ACTH 1–24 test should not be utilized in the early
autoantibody test, CT imaging of the adrenal region postoperative period, that is <6 weeks after pituitary
is recommended to rule out inflammatory processes surgery171,172. However, the ACTH 1–24 test can be
or destruction of the adrenal glands by haemorrhage or useful for predicting HPA axis recovery56. The stimu-
infiltration, for example, by bilateral metastases of lation of the pituitary–adrenal axis with synthetic CRH
an extra-​adrenal cancer28,163,164. In men with negative excludes the direct assessment of hypothalamic function.
21-​hydroxylase autoantibodies, adrenoleukodystrophy Although CRH testing is used in some centres, it is not
should be ruled out by measuring the levels of very long known to be superior to the ACTH stimulation test165,167.
chain fatty acids in serum. In sub-​Saharan Africa, the The metyrapone test is based on metyrapone-​
most common aetiologies of PAI, such as tuberculosis mediated inhibition of the 11β-​hydroxylase enzyme,
and HIV infection, should be investigated to enable which catalyses the final step of cortisol synthesis,
treatment of the underlying condition5. thereby evoking an increased ACTH response by an
intact pituitary. Patients are usually given metyrapone
Secondary adrenal insufficiency. The diagnostic tools at midnight with a snack, and blood samples for analys-
used to confirm the presence of SAI vary widely among ing ACTH, 11-​deoxycortisol and cortisol are obtained
countries and even among different endocrine centres after 8 h. An increase in plasma 11-​deoxycortisol to
in the same country165. In general, any study evaluat- >200 nmol/l suggests an intact HPA axis173. In search
ing assay-​specific cut-​off values is limited by the lack of increased sensitivity and specificity of the test, other
of a well-​established gold standard to define SAI1,166. studies have suggested the use of the sum of serum
Assessing the stress responsivity of the HPA axis in 11-​deoxycortisol and cortisol with a cut-​off value of
patients with known or suspected pituitary disease is 450 nmol/l owing to assay cross-​reactivity174 or a com-
extremely important167. In general, besides the thyreo- bination of ACTH response and increase in plasma
tropic axis, the corticotropic axis is one of the last ana- 11-​deoxycortisol levels175.
tomical units to be lost in structural pituitary disease The glucagon stimulation test is rarely used but it
including tumours or trauma, although autoimmune may also be an acceptable alternative to the insulin tol-
hypophysitis may cause isolated ACTH deficiency A erance test for evaluating the HPA axis176. In the future,
history of traumatic brain injury or stroke and hypon- measurement of basal salivary cortisol and salivary
atraemia with the presence of symptoms, such as con- cortisone might improve the screening of SAI, owing
centration and cognitive deficits, nausea, vomiting, to its simplicity and cost-​effectiveness and because it
confusion, headache, somnolence and seizures, should is independent of binding proteins such as albumin or
prompt further investigation for potential SAI. CBG177,178.
A morning serum cortisol of <100 nmol/l in com-
bination with a low or low–normal ACTH level is con- Screening
firmative of SAI, whereas a morning serum cortisol Universal newborn screening for CAH has already been
of >450 nmol/l excludes SAI167. The insulin tolerance implemented in most developed and many developing
test is regarded by most clinicians as the reference countries. Timely screening is an important diagnostic


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 13

0123456789();:
Primer

tool, guaranteeing timely initiation of replacement specifically metabolic syndrome, cardiovascular disease
therapy179,180. As is the case for many rare diseases, it is and osteoporosis; and preventing mortality, particularly
important to have AI as a potential differential diag- due to adrenal crisis183.
nosis in mind; for example, patients with other auto-
immune diseases such as hypothyroidism or type 1 Glucocorticoid replacement
diabetes mellitus should be screened for PAI. Similarly, The standard tenet in endocrinology is that hormone
patients with other pituitary hormonal insufficiencies replacement should aim to restore normal physiology
should be screened for SAI. Screening for AI should be and health. In healthy individuals with a regular diur-
considered in all patients presenting with unexplained nal sleep pattern, cortisol levels rise between 2 a.m. and
hyponatraemia, hypotension, vomiting or diarrhoea157. 4 a.m., reaching a peak shortly after waking, and decline
The optimal schedule for screening for associated over the day to low levels from approximately 6 p.m.,
immune disorders in patients with Addison disease is with slight increases at meal times184. Attaining phys-
not established. Annual testing for thyroid disease, dia- iological cortisol profiles with existing treatments has
betes mellitus, premature ovarian failure, coeliac disease been unachievable so far, owing to the inability of the
and autoimmune gastritis with vitamin B12 deficiency medications to replicate the native circadian rhythm of
may be regarded as appropriate15. cortisol. Consequently, no markers of biological activ-
ity that predict morbidity and mortality are available to
Management date. Cortisol measurements are usually not useful in
Different guidelines or consensus statements for the monitoring the quality of replacement therapy. If hydro-
management of AI exist. These guidelines includ- cortisone or cortisone acetate is used for replacement,
ing those published by the US Endocrine Society, the cortisol measurements are just reflective of the medica-
European Society of Endocrinology, and specialist soci- tion or are inaccurate in the case of prednisolone doses
eties in the UK and Japan and other countries. Although owing to cross-​reactivity in immunoassays. However, in
the dosage detail might slightly vary, the goals of the some cases, for example, when a discrepancy between
treatment remain similar across all guidelines15,155,181,182. daily doses and patient symptoms exists, measurement
The most important goals of AI treatment include: of a cortisol profile might be helpful for examining the
achieving the optimal glucocorticoid dosing regi- gastrointestinal absorption of hydrocortisone. Thus, it
men; improving patient quality of life and daily life is suggested that the prescribing physician should titrate
performance; avoiding the risks of over-​replacement, the glucocorticoid dose based on patient symptoms of
cortisol deficiency and excess. Insufficient hydrocorti-
sone doses result in fatigue, weight loss and nausea, and
Box 1 | Pregnancy and adrenal insufficiency might lead to adrenal crisis. On the other hand, high
Pregnancy is physiologically associated with increased levels of corticotropin-​releasing
hydrocortisone doses can cause cortisol excess with
hormone, adrenocorticotropic hormone, and free and total cortisol275. Studies have found features of Cushing syndrome including weight gain,
reduced fertility rates, increased risk of preterm birth and postpartum complications increased abdominal fat, buffalo hump, thin skin, easy
in patients with adrenal insufficiency (AI)275–279, emphasizing the need for optimal bruising, hypertension and type 2 diabetes mellitus.
surveillance in planned or ongoing pregnancy. Risk of premature ovarian insufficiency Many patients with AI complain of fatigue despite treat-
has been found in 5–16% of premenopausal women with Addison disease15,67. The rate ment and evidence suggests that simply increasing the
of delivery via caesarean section is increased, partly owing to a lower threshold for glucocorticoid dose does not resolve the symptoms185.
recommending caesarean section276,278,280. In women with congenital adrenal In children with AI, monitoring of height and weight
hyperplasia (CAH), the birth rate is lower than in controls, gestational diabetes and is essential practice. In children, replacement doses are
caesarean section are more common than in controls, but perinatal outcomes are
prescribed as dose per body surface area, whereas in
comparable to those in controls281. Women with secondary adrenal insufficiency (SAI)
may have infertility due to concomitant gonadotropic insufficiency and need additional
adult patients treatment-​initiating doses are prescribed
therapies including ovulation induction. In general, the majority of women with AI according to guidelines for the specific glucocorti-
experience a pregnancy with good fetal and maternal outcomes. coid and adapted based on clinical follow up15,155,181,182.
In pregnant women with AI, adjustment of the hydrocortisone dose is crucial15,280. In general, in patients with PAI a complete replacement
The adjustment is usually implemented in weeks 20–24 of pregnancy with further of the daily cortisol production is necessary, whereas
adjustment based on clinical assessment. Given the anti-​mineralocorticoid effect in SAI a near-​complete replacement dose is often suf-
of progesterone282,283 and because its circulating concentrations increase during ficient as patients with SAI might have residual ACTH
pregnancy, fludrocortisone dose may need adjustment. Physicians should rely on secretion. In pregnant patients with AI the hydrocor-
blood pressure and serum electrolytes to judge fludrocortisone dosing as pregnancy tisone doses need to be adjusted during the course of
increases renin concentrations284,285. Hydrocortisone dose should be increased during
pregnancy (Box 1).
stressful events in a similar manner to its adjustment in non-​pregnant patients with AI.
Placental expression of 11β-​HSD2 results in rapid cortisol inactivation and prevents
Hydrocortisone (the pharmaceutical name for cor-
glucocorticoid over-​exposure of the fetus286. However, this inactivation does not occur tisol) is the first-​line glucocorticoid replacement for
with synthetic corticosteroids (for example dexamethasone) and, therefore, synthetic patients with AI. The plasma half-​life of hydrocortisone
glucocorticoids should be avoided unless glucocorticoid effects on the fetus are desired. is ~90 min and is, therefore, dosed twice or thrice daily
Spontaneous delivery is the preferred way of delivery. However, in women with CAH, with the highest dose given in the morning on awaken-
vaginal delivery may be impossible as a consequence of genital corrective surgery and, ing, the next dose given early afternoon (two-​dose reg-
therefore, should be assessed individually by a gynaecologist. Major hydrocortisone imen) or at lunch and afternoon (three-​dose regimen),
stress dose cover should be initiated upon entering the active stage of labour. Clear to replicate the circadian rhythm15. Plenadren is a dual-
communication with the obstetric department is necessary and should include written release hydrocortisone drug that enables once-daily
instructions on hydrocortisone replacement.
dosing of hydrocortisone. This formulation consists of

14 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

a coated outer layer that provides immediate release Mineralocorticoid replacement


of the drug and an extended-​release inner core186. Patients with PAI, in particular those with Addison
However, Plenadren has ~20% less bioavailability than disease or the salt-​wasting form of CAH, and patients
immediate release hydrocortisone187. Open-​label studies after bilateral adrenalectomy require mineralocorti-
of Plenadren have shown reductions in BMI in adults coid replacement if aldosterone production is impaired.
with AI after changing from hydrocortisone and these However, residual aldosterone production was found
changes were sustained after 12 months of treatment in ~13% of patients with Addison disease199. The goal
with improvements in lipid profile. However, whether of therapy is to achieve general well-​being, prevent salt
this finding was a consequence of lower overall expo- craving, and restore normal blood pressure and normal
sure to hydrocortisone owing to reduced bioavailabi­ serum electrolyte levels15,200.
lity or related to the pattern of hydrocortisone release Aldosterone by itself is not used in replacement
is unclear188,189. One new study demonstrated improved therapy owing to its short half-​life and rapid hepatic
immune cell profiles and reduced susceptibility to metabolism after oral ingestion. The preferred min-
infections in patients after switching to Plenadren190. eralocorticoid preparation for replacement therapy is
Prednisolone, a synthetic glucocorticoid, is more 9α-​fluorocortisol (also known as fludrocortisone) which
potent with a longer duration of action with an elim­ has a 200–400-​fold higher mineralocorticoid potency
ination half-​life of 150 min. Prednisolone has less than hydrocortisone201. Its introduction into clinical
mineralocorticoid activity than hydrocortisone and is practice ended the use of deoxycorticosterone, which
reco­mmended in patients with poor compliance15. In some had been used since the 1960s. Fludrocortisone is more
countries, cortisone acetate is used for replace­ment stable, available in tablet form and can be better titrated
therapy. Prednisone and cortisone acetate are precur­sors to regulate its biological effects. Fludrocortisone also
of prednisolone and cortisol, respectively, and require balances the other well-​known symptoms of aldosterone
activation by hepatic 11β-​hydroxysteroid dehydro­genase deficiency, such as electrolyte imbalance (hyperkalaemia
type 1 to show biological activity. Dexamethasone, a and hyponatraemia), hypovolaemia and hypotension.
potent, albeit difficult to titrate glucocorticoid is gen- However, the fact that the mineralocorticoid receptor
erally not recommended for replacement therapy in is also expressed in several brain regions might explain
AI owing to its complete lack of mineralocorticoid the effect of fludrocortisone on cognitive function
activity and its association with poor health outcomes and mood, with better performance in patients with
in patients with CAH105,191. In addition, dexametha- high mineralocorticoid receptor occupancy following
sone does not exert any mineralocorticoid activity, as fludrocortisone uptake202.
opposed to cortisol, where a 40-​mg dose of hydrocor- Children and young adults may require higher doses
tisone exerts mineralocorticoid activity equivalent to of fludrocortisone than most adult patients with PAI201
100 µg of fludrocortisone. Findings from studies using as the mineralocorticoid receptor is less sensitive in chil-
continuous subcutaneous infusions of hydrocortisone in dren and, therefore, mineralocorticoid requirements
small subsets of patients with PAI are inconsistent, with a should be re-​assessed during late adolescence or early
few studies showing improved quality of life192 and a few adulthood. Additionally, patients on prednisolone or
reporting otherwise193. In addition, continuous subcuta- dexamethasone might require higher fludrocortisone
neous infusions of hydrocortisone have been shown to doses, owing to their lower mineralocorticoid activity
improve symptoms of CAH194, although skin reactions than hydrocortisone. In patients who develop essential
are common. The treatment of CAH has the additional hypertension during their lifetime, hydrocortisone dose
challenge of controlling excess androgens in addition to should be re-​e valuated and the fludrocortisone
replacing cortisol. Current oral therapies with hydro- dose may be reduced depending on plasma renin and
cortisone, prednisolone and even dexamethasone can- electrolyte levels but should not be discontinued. A direct
not control androgen excess despite following dosing vasodilator (for example, calcium antagonist) is prob-
in a reverse circadian fashion with a dose just before ably the best choice of anti-​hypertensive treatment in
bedtime105,191. Chronocort, a modified-​release hydro- patients with PAI. Diuretics, especially aldosterone anta­
cortisone under development, differs from Plenadren gonists, such as spironolactone or eplerenone, should
by having a delayed and sustained absorption profile be avoided203. In patients with PAI and heart failure,
rather than an immediate-​release and sustained-​release angiotensin-​converting enzyme inhibitors, angiotensin II
profile195. Chronocort aims to mimic the physiological antagonists or β-​blockers are recommended, and the use
cortisol levels such that the night-​time dose releases of mineralocorticoid receptor antagonists and/or dis-
hydrocortisone in the early hours of the morning, continuation of fludrocortisone should be decided on an
providing physiological waking hour cortisol levels195. individual basis203. The effects of fludrocortisone might
A phase II open-​label study of Chronocort in 16 adults not last for 24 hours because of faster elimination rates in
with CAH showed improved biochemical control of some patients; in these patients, twice-​daily fludrocorti-
CAH196. Furthermore, a phase III study of Chronocort sone is recommended. Patients with mineralocorticoid
showed improved morning control of adrenal androgens deficiency are allowed to consume sodium salt and salty
compared with standard therapy197. A newly developed foods without restriction.
CRFR1 antagonist has been shown to offer an additional Monitoring of mineralocorticoid replacement ther-
strategy to reduce ACTH production without increasing apy is performed by questioning the patient about salt
glucocorticoid therapy above physiological replacement craving, light-​headedness or orthostatic dysregula-
doses in patients with CAH198. tion, by measuring blood pressure, by examining for


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 15

0123456789();:
Primer

the presence of peripheral oedema148 and by measur- body composition, bone composition or turnover
ing serum potassium and sodium levels. The meas- markers, metabolic or cardiovascular parameters were
urement of plasma renin levels might potentially be observed in patients with AI receiving DHEA replace-
useful200,204, especially in determining mineralocorticoid ment, indicating no relevant anabolic activity212,214–217.
under-​replacement205. However, plasma renin levels A meta-​analysis of DHEA trials in women with PAI or
might be high in women using hormonal contraceptives, SAI focusing on quality of life, depression, anxiety and
owing to the stimulation of plasma renin substrate by sexual well-​being revealed only minor effects of DHEA
oestrogen and, therefore, should be used with caution and the data were not sufficiently compelling to support
for monitoring purposes. Fludrocortisone-​mediated routine use of DHEA218. The decision to initiate DHEA
regulation of the extracellular fluid volume and blood replacement is made on an individual basis, with a focus
pressure changes following dose changes are only seen on women with impaired well-​being associated with
after several days or weeks. Therefore, fludrocortisone signs and symptoms of androgen deficiency, such as
dose changes should not be monitored earlier than dry skin and reduced libido219. A single oral morning
2–4 weeks after changing the dose201,206,207. Rapid weight dose of 25–50 mg DHEA, with evaluation of beneficial
gain, high blood pressure, oedema and hypokalaemia effects and adverse effects after 6 months of replacement,
indicate an excessive dose of fludrocortisone148. is recommended15,139. Patients need to be informed that
Phenytoin, phenobarbital, rifampicin and mitotane it may take several months for beneficial effects to occur.
induce hepatic 6β-​hydroxylation via CYP3A4, one of Monitoring of treatment includes clinical evaluation of
the major 9α-​fluorocortisol metabolic pathways. Patients androgenic effects and the measurement of morning
with PAI on these drugs need increased doses of fludro- serum DHEAS, androstenedione, testosterone and sex
cortisone and also of hydrocortisone as drug-​mediated hormone binding globulin prior to the intake of the daily
CYP3A4 induction results in more rapid inactivation DHEA dose, aiming at a mid-​normal reference range.
of cortisol208. In situations of increased salt loss such
as sweating in a hot climate, transiently increasing the Adverse effects
fludrocortisone dose and/or the salt intake might be Osteoporosis. Until 2009, studies investigating bone min-
useful148. This dosing is often routinely followed during eral density (BMD) in patients with AI included only a
the summer months in countries with a hot climate. small number of patients (<30) and found contradictory
Patients with PAI receiving high doses of hydrocortisone results. A two-​cohort cross-​sectional study in Norway, UK
(>50 mg per day) do not need an additional fludrocor- and New Zealand (in 292 patients), showed that patients
tisone dose as such high hydrocortisone doses provide on an average daily hydrocortisone dose of 30 mg per day
sufficient mineralocorticoid activity. had a significantly lower BMD than population-​based
controls, and BMD decreased further with increasing daily
DHEA replacement glucocorticoid doses220. By contrast, patients with AI who
Most, but not all trials investigating the effect of DHEA received lower daily doses of hydrocortisone than typi-
replacement in patients with AI have demonstrated a cally used in replacement therapy (mean 12.0 ± 2.7 mg/m2)
positive influence on mood and subjective health status had BMD values within the normal reference range221.
and on libido (particularly in women)142,209–212. These In addition, a prospective, longitudinal study demon-
effects were generally moderate and showed marked strated that cautious reduction of glucocorticoid doses
inter-​individual variability, with the most significant led to increases in BMD, whereas dose increments
effects observed in women. However, large-​scale out- reduced BMD in patients with PAI222. A population-​based
come studies of chronic DHEA supplementation are cohort study found a nearly doubled risk of hip fracture
lacking and, therefore, DHEA replacement is not a in patients with PAI compared with controls (HR 1.8,
mandatory part of the standard replacement regimen in 95% CI 1.6–2.1; P < 0.001)223. A cross-​sectional study in
patients with AI. Nevertheless, clinical signs of andro- 365 patients with SAI showed an association between
gen deficiency in women with AI may be successfully glucocorticoid replacement and reduced BMD, inde-
treated by DHEA replacement. Evidence shows a role for pendent of other known risk factors such as patient age224.
DHEA in immunomodulation; for instance, oral DHEA However, evidence shows that growth hormone deficiency
replacement has been shown to restore normal levels of is the main determinant of skeletal fragility in patients
regulatory T cells in patients with Addison disease213. with hypopituitarism, and growth hormone replacement
In addition, rodent-​based studies have suggested that therapy normalized fracture risk in these patients225,226.
DHEA enhances natural killer cell function213. However, Among different glucocorticoids used for replacement,
another study that documented highly decreased natu- prednisolone use was associated with the worst BMD in
ral killer cell cytotoxicity in patients with PAI did not patients with PAI221,222 and in patients with CAH227.
find any improvement of natural killer cell cytotoxicity
in patients with established DHEA replacement. This Cardiovascular morbidity and metabolic syndrome.
finding implies that the decreased natural killer cell cyto- Although glucocorticoid excess is a well-​known risk fac-
toxicity might rather be due to glucocorticoid-​induced tor for adverse cardiovascular and metabolic outcome,
alterations in clock gene regulation in natural killer cells. the question arises as to whether non-​physiological
Although increased IGF1 levels have been docu- cortisol profiles achieved by standard replacement
mented following initiation of DHEA replacement can also contribute to such adverse effects. In general,
(which may be of relevance for patients receiving growth cardiovascular mortality was found to be increased in
hormone replacement therapy), only minor effects on patients with AI compared with the general population65.

16 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

A study utilizing the US-​based national payer database mineralocorticoid used. Another study involving
including 10,383 patients with AI (1,014 patients with 588 patients with CAH and 58,800 matched controls
PAI, 8,818 patients with SAI and 551 patients with showed that morbidity associated with cardiovascular
CAH) revealed that the odds ratios for diabetes melli- disorders (hypertension, atrial fibrillation and venous
tus, hypertension and hyperlipidaemia were significantly thromboembolism) and metabolic disorders (dys-
higher in patients with AI than in matched controls lipidaemia, obesity, diabetes mellitus and obstructive
from the same database23. By contrast, the prevalence of sleep disorder) was increased in patients with CAH232,
these cardiovascular risk factors did not differ between with women being generally more affected than men.
549 Swedish patients with Addison disease and controls34. Furthermore, the specific glucocorticoid preparation
Several analyses have indicated that discrepancies in seems to play a part; for example, longer-​acting pre­
cardiovascular risk might be due to dose differences dnisolone was associated with a worse lipid profile than
in and the timing of glucocorticoid replacement. For hydrocortisone in patients with PAI and SAI233. Similarly,
example, hydrocortisone doses in the late afternoon or dexamethasone caused a more adverse metabolic
evening impaired insulin sensitivity more than morn- risk profile in patients with CAH than shorter-​acting
ing doses134,228,229. In addition, patients with SAI with glucocorticoids191,234,235.
daily hydrocortisone-​equivalent doses of >20 mg were Hypertension may occur in PAI and SAI, although it is
at increased risk of developing signs and symptoms of less frequently observed than in the general population34.
the metabolic syndrome230. Another Swedish analysis Patients with PAI with hypertension have to be assessed
involving 1,500 patients with PAI found an increased for signs of mineralocorticoid over-​replacement, such
risk of ischaemic heart disease but not cerebro­vascular as low renin levels; however, in most patients the min-
disease, particularly in women, compared with the eralocorticoid dose may be maintained. High doses of
general population231. This study showed a significant hydrocortisone can cause hypertension, but might also
positive correlation between the prevalence of ischae- promote the progression of atheromatous vascular dis-
mic heart disease and the doses of glucocorticoid and ease and may contribute to the increased number of pre-
mature cardiovascular deaths among patients with AI65.
An open crossover study in ten men with SAI who
Box 2 | Sick day rules for stress dosing with glucocorticoids were randomized to three hydrocortisone dose regi-
mens (each over 6 weeks), demonstrated that the lowest
Cortisol levels increase in response to stress to cover the body’s increased need for
protective stress hormones. Accordingly, extra doses of glucocorticoids must be dose regimen (10 mg–5 mg–0 mg) was associated with
given under respective circumstances in patients with AI. Sick day rules are a set of the greatest lowering of blood pressure, and this was
recommendations for glucocorticoid dose adjustment under specific circumstances supported by the findings of another study236,237. Thus,
to prevent adrenal crisis10,15. mineralocorticoid dose as well as glucocorticoid replace-
Conditions
ment dose needs to be carefully evaluated in patients
with hypertension.
• Management of illness with fever at home (sick day rule 1) should include doubling
(if fever >38 °C) or tripling (if fever >39 °C) of oral hydrocortisone replacement dosing
until recovery (usually after 2–3 days). Adrenal crisis
• If the patient is unable to tolerate oral medication owing to vomiting and/or
The management of adrenal crisis or acute AI is straight-
diarrhoea, trauma, high fever and clinical deterioration (sick day rule 2), 100 mg forward and, if delivered promptly, highly successful.
parenteral (intramuscular or subcutaneous) hydrocortisone should be given and An initial intravenous or intramuscular bolus injection
health professionals contacted in case of initial self-​treatment. of 100 mg hydrocortisone should be followed prefer-
ably by continuous intravenous infusion of 200 mg of
Medical procedures hydrocortisone per 24 hours, or alternatively, by intra-
• For minor or moderate surgical stress and major dental procedures, 100 mg venous or intramuscular bolus injections of 50 mg of
hydrocortisone as a subcutaneous bolus before anaesthesia or as an intravenous hydrocortisone every 6 hours. A pharmacokinetic study
infusion for the duration of surgery should be given.
demonstrated that continuous intravenous infusion can
• For major surgery with general anaesthesia, trauma, delivery or disease that requires prevent the intermittent troughs in circulating corti-
intensive care, 100 mg hydrocortisone as an intravenous injection followed by
sol observed with intramuscular or intravenous bolus
continuous intravenous infusion of 200 mg hydrocortisone over 24 h is recommended;
alternatively, if continuous administration is not possible or feasible, an intravenous or
injections146. After the initial treatment period and
intramuscular bolus of 50 mg hydrocortisone every 6 h should be administereda. with recovery of the patient, the hydrocortisone dose
should be tapered in line with the clinical response of
Acute adrenal crisis the patient, with close monitoring of blood pressure
• 100 mg hydrocortisone as an intravenous bolus injection followed by 200 mg and clinical symptoms. Intravenous glucose may be
hydrocortisone as a continuous infusion over 24 h should be administered; needed, in particular in children, in whom concomi-
subsequently, dosing should be tapered and switched to oral administration tant hypoglycaemia is much more frequently observed
according to the patient’s clinical condition.
than in adults238. Transfer to oral hydrocortisone within
• Fluid resuscitation should be tightly monitored and should involve administration of 24 hours is possible if there is no ongoing underlying
up to 1,000 ml isotonic saline within the first hour, followed by continuous intravenous
inflammatory precipitant. Intravenous fluids, gener-
isotonic saline guided by individual patient needs, particularly taking into consideration
ally isotonic saline, should be given generously, albeit
plasma sodium levels and urinary excretion.
under tight monitoring to avoid either exacerbation
a
Different glucocorticoid supplementation schemes for surgical procedures exist that are or too-​rapid correction of hyponatraemia10,239 (Box 2).
tailored to the severity of the surgery287; of note, one study showed that continuous intravenous Treatment of the precipitating cause of the crisis is essen-
infusion is the best scheme for surgical procedures146.
tial. After fluid resuscitation, it is advisable to consider


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 17

0123456789();:
Primer

Box 3 | Patient education and prevention of adrenal crisis of the origin of the AI or concomitant disease185,244,245.
Studies have found impairment of physical and emo-
Patient education pursues several goals, including: improving quality of life and daily tional health with reduced vitality and increased fatigue
life performance by providing a better understanding of the disease and knowledge on and anxiety3,246–250. In addition, affective and depressive
options for action by the patient; reducing morbidity from chronic over-​replacement or disorders are highly prevalent in patients with AI23,142,185.
under-​replacement; and reducing mortality by prevention or optimal management of
Multiple studies have found high inter-​individual vari-
emergency situations.
Education is key for preventing adrenal crisis. One study showed that only 30–66% of ation in the quality of life scores, indicating that some
patients were equipped with glucocorticoids for emergency use11,69,288. In patients with subgroups of patients are more vulnerable than others251.
AI, rapid situational dose adaptation is frequently required to adapt the corticosteroid This difference is also reflected by variable daily perfor-
supply to the body’s need. Glucocorticoid administration by health professionals in mance of patients. In general, 40–80% of patients with
case of emergencies can be delayed265; a well-​trained patient (or relative) is therefore AI report disruption of their daily life activities owing
crucial for optimal management. Education in dose adjustment and self-​injection of to the disease. Several studies have found occupational
glucocorticoids enables patients to manage daily challenges more independently267. changes in AI, with disability pensions received up to
All patients with AI have to be equipped with a steroid emergency card and four times more frequently among patients than among
prescription for an additional supply of oral glucocorticoids as well as a hydrocortisone the general population185,245,252. Interestingly, patients in
self-​injection kit for emergency management. Patients require education to recognize
whom the diagnosis of AI was delayed had a significantly
signs and symptoms of adrenal crisis and on the correct behaviour in the event of
an emergency, which includes explanation of sick day rules and training in hydrocortisone worse quality of life than those in whom the diagnosis
emergency self-​injection10,15. Patients need written instructions on glucocorticoid dose was not delayed9.
adaptation in case of medical interventions. Clinical data on correct stress dosing are The underlying causes of impaired quality of life in
limited and many recommendations are based on expert opinion and may show slight patients with AI remain to be elucidated. The effects of
variations. the chronic disease in combination with a lack of DHEA
Studies have demonstrated the benefit of structured education strategies, revealing and epinephrine, and the complexities associated with
a significant increase in knowledge on AI and its management as well as improved non-​physiological mineralocorticoid and glucocorti-
subjective self-​assurance of patients to deal with their disease265–267,289,290. Nevertheless, coid replacement discussed above, could all contribute
assurance in correct handling of emergencies declined after several months267, to the impaired quality of life in patients251. The lack of
indicating that at least bi-​annual repetition of education is necessary for patient
circadian and ultradian cortisol tissue exposure is par-
empowerment.
ticularly regarded as a factor contributing to the reduced
quality of life. A cross-​sectional study demonstrated no
causes and triggers of adrenal crises that may recur and differences in the quality of life based on the type of glu-
review preventative measures. Patients should be edu- cocorticoid prescribed252. However, high daily hydro-
cated about the potential seriousness of such events, with cortisone doses were associated with a worse quality of
the advice that adequate self-​management makes events life status185,244,253, although these studies do not provide
rare and adverse consequences unlikely so that excessive any information about causality. For instance, patients
anxiety is minimized. Delayed treatment of adrenal with hypercortisolism exhibit functional damage in
emergencies risks death. Delays may be owing to concern brain regions involved in stress reactivity and emotional
about hydrocortisone-​induced adverse effects. Physician processing. Patients reporting reduced quality of life are
training and awareness may be insufficient owing to the more likely to receive increased doses of hydrocortisone,
rare incidence of adrenal crisis240,241. Improved patient which might further impair quality of life244. Thus, a dose
self-​treatment, particularly with self-​directed delivery increase based on a patient-​reported impaired quality
of intramuscular hydrocortisone will help circumvent of life always needs re-​evaluation. Non-​blinded stud-
these delays. Subcutaneous hydrocortisone may be used ies have shown that hydrocortisone formulations that
as an alternative to intramuscular hydrocortisone for closely mimic the physiological cortisol profile seem to
prevention of adrenal crisis, as subcutaneous delivery improve quality of life186,254.
showed satisfactory and minimally delayed absorption
in volunteers with PAI242. However, its use is off-​label Outlook
and it is not clear whether absorption of hydrocortisone Mechanisms and diagnosis
is diminished in the presence of severe clinical deteri- AI is receiving increasing attention, particularly owing
oration with peripheral vasoconstriction in patients to its association with the use of novel drugs (Table 4).
with hypotension or shock. Parenteral hydrocortisone Timely diagnosis of AI is crucial to prevent negative
administration seems to be superior to rectal supposi- long-​term consequences and adrenal crisis. Bearing
tories (prednisone), which are used in some patients as in mind that AI is a potential differential diagnosis,
an alternative for crisis prevention15,243. However, the key particularly in patients with unexplained hyponatrae-
strategy for prevention of adrenal emergencies is patient mia, fatigue, hypotension and weight loss, is key for
education and early response to potential risks (Box 3). timely identification and treatment of patients with
AI. Diagnostic evaluation often remains challenging
Quality of life in patients with SAI and the optimum diagnostic tests
The general well-​being of an individual and the expecta- and cut-​off values remain to be established by large pro-
tions for a good quality of life are influenced by somatic spective studies. Several open questions remain to be
and mental symptoms. Health-​related quality of life, addressed including the mechanisms associated with
assessed by disease-​specific and general questionnaires, the onset of Addison disease. Future research facilitat-
is frequently impaired in patients with PAI and SAI ing the translation of genetic susceptibility into potential
according to data from several countries, irrespective pathogenetic mechanisms of autoimmune inflammation

18 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

Table 4 | Drugs inducing adrenal insufficiency


Compounds Mechanism
Supraphysiological treatment with Suppression of the HPA axis through negative feedback on the
glucocorticoid hormones, e.g. prednisone, hypothalamus and pituitary and induction of tertiary adrenal
prednisolone, dexamethasone, triamcinolone insufficiency in case of long-​term treatment
Opioids, e.g. morphine, fentanyl, tramadol, Inhibition of the HPA axis by blocking release of hypothalamic
methadone or mixed opioids corticotropin-​releasing hormone; also affect adrenal steroid
production
Mifepristone Glucocorticoid receptor antagonist; adrenal insufficiency occurs
in overdosing
Adrenostatic agents, e.g. etomidate, Inhibition of corticosteroid synthesis by inhibition of steroidogenic
metyrapone, ketoconazole, osilodrostat enzymes, mainly 11β-​hydroxylase (CYP11B1) and aldosterone
synthase (CYP11B2), side-​chain cleavage enzyme (CYP11A1).
Mitotane Destruction of adrenocortical tissue (adrenolytic effect); strong
induction of the cortisol-​metabolizing enzyme CYP3A4
Rifampicin, phenytoin, troglitazone, Increased cortisol metabolism by induction of CYP3A4
phenobarbital
IFNα Induction of hypophysitis
Immune checkpoint inhibitors, e.g. avelumab, Hypophysitis due to lymphocytic pituitary infiltration of lymphocytes
ipilimumab, nivolumab, pembrolizumab leading to ACTH deficiency; less common, adrenalitis and direct
impairment of adrenal cortex function (new immune checkpoint
inhibitors seem to cause hypophysitis less frequently)
ACTH, adrenocorticotropic hormone; HPA, hypothalamic–pituitary–adrenal; IFNα, interferon-​α. Table adapted from refs31,269.

is warranted. Understanding the as-​yet largely unclear Restored immune function might therefore help pre-
pathophysiology of adrenal crisis is of high importance vent adrenal crisis and associated mortality. Ongoing
for the prevention and management of such emergen- and future trials are expected to elucidate these aspects
cies. In addition, the precise mechanisms underlying in patients with AI.
ICI-​induced hypophysitis require further investigation.
Regenerative medicine. Regenerative medicine
Management approaches have attempted to restore endogenous
Given the persistent disease burden (for example, adrenocortical steroid production with the aim of mim-
reduced vitality) and the increased mortality observed icking the pulsatility of cortisol259. In preclinical studies,
in patients with established replacement therapy, ther- a bioartificial adrenal could be established by encapsu-
apeutic concepts are currently changing. In addition lation of bovine adrenocortical cells in alginate260. The
to standard hydrocortisone therapy, new formulations adrenal cortex generally shows high plasticity and regen-
and modes of administration designed to best mimic erative capacity. The adrenocortical progenitor cells are
the physiological cortisol circadian rhythm are being located in the subcapsular region of the adrenal cortex,
evaluated in clinical trials193,195,254,255. Preliminary results which is assumed to contribute to rapid regeneration.
indicate that easier dose reduction and optimization of Indeed, residual adrenal function has been observed in
glucocorticoid exposure over the day can be achieved, up to 30% of patients with Addison disease199, provid-
and may reduce adverse effects. The extent to which ing a basis for subsequent studies aiming at restoring
these new formulations can improve the long-​term endogenous steroid production. Long-​term treatment
outcomes in patients with AI will be revealed by future with 1–24 ACTH or B cell depletion therapy was able to
study results. The relevance of the pulsatile ultradian restore endogenous cortisol secretion, although only in
activity of the HPA axis in addition to the circadian few patients261–263. Nevertheless, stem cell-​based regen-
pattern of cortisol secretion is of additional scientific erative medicine and adeno-​associated virus-​mediated
interest and probably also of clinical importance. A pilot gene therapy (particularly for CAH) may be promising
study investigating the effects of pulsatility on cognition, approaches to the treatment of AI in the future.
emotion and sleep in young cortisol-​suppressed men The backbone of optimal therapy, especially in
demonstrated that pulsatility is important for intact neu- rare diseases such as AI, is a well-​informed patient
rological physiology, optimal quality of sleep and perfor- and environment, as emergency management is not
mance of working memory compared with continuous always known to health-​care professionals but often
cortisol infusions89. Existing glucocorticoid replace- requires timely interventions240,264,265. The establishment
ment regimens could furthermore adversely impact the and continuous improvement of structured training
CLOCK-​mediated regulation of the immune system programmes266,267 and the development and homogeni-
including natural killer cell function. A decreased natu- zation of international guidelines and emergency tools268
ral killer cell cytotoxicity was observed in patients with will make an important contribution to the long-​term
PAI256, which might underlie the observed increased rate outcomes in patients.
of infections21,257,258 and the reported increased mortality
owing to respiratory infections in patients with PAI65. Published online xx xx xxxx


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 19

0123456789();:
Primer

1. Arlt, W. & Allolio, B. Adrenal insufficiency. Lancet 361, 26. Betterle, C., Dal Pra, C., Mantero, F. & Zanchetta, R. 49. Chang, L. S. et al. Endocrine toxicity of cancer
1881–1893 (2003). Autoimmune adrenal insufficiency and autoimmune immunotherapy targeting immune checkpoints.
2. Ross, I. L. & Levitt, N. S. Addison’s disease polyendocrine syndromes: autoantibodies, Endocr. Rev. 40, 17–65 (2019).
symptoms–a cross sectional study in urban South autoantigens, and their applicability in diagnosis 50. Li, T. et al. Prevalence of opioid-​induced adrenal
Africa. PLoS ONE 8, e53526 (2013). and disease prediction. Endocr. Rev. 23, 327–364 insufficiency in patients taking chronic opioids.
3. Erichsen, M. M. et al. Clinical, immunological, and (2002). J. Clin. Endocrinol. Metab. 105, e3766–e3775
genetic features of autoimmune primary adrenal 27. Laureti, S. et al. Levels of adrenocortical (2020).
insufficiency: observations from a Norwegian registry. autoantibodies correlate with the degree of adrenal 51. de Vries, F. et al. Opioids and their endocrine effects:
J. Clin. Endocrinol. Metab. 94, 4882–4890 (2009). dysfunction in subjects with preclinical Addison’s a systematic review and meta-​analysis. J. Clin.
4. Afreen, B., Khan, K. A. & Riaz, A. Adrenal insufficiency disease. J. Clin. Endocrinol. Metab. 83, 3507–3511 Endocrinol. Metab. 105, 1020–1029 (2020).
in Pakistani HIV infected patients. J. Ayub Med. Coll. (1998). 52. Saeed, Z. I., Bancos, I. & Donegan, D. Current
Abbottabad 29, 428–431 (2017). 28. Herndon, J., Nadeau, A. M., Davidge-​Pitts, C. J., knowledge and practices of heath care professionals
5. Mofokeng, T. R. P., Beshyah, S. A., Mahomed, F., Young, W. F. & Bancos, I. Primary adrenal insufficiency on opioid-​induced adrenal insufficiency. Endocr. Pract.
Ndlovu, K. C. Z. & Ross, I. L. Significant barriers to due to bilateral infiltrative disease. Endocrine 62, 25, 1012–1021 (2019).
diagnosis and management of adrenal insufficiency 721–728 (2018). 53. Woods, C. P. et al. Adrenal suppression in patients
in Africa. Endocr. Connect. 9, 445–456 (2020). 29. Merke, D. P. & Auchus, R. J. Congenital adrenal taking inhaled glucocorticoids is highly prevalent
6. Odeniyi, I. A., Fasanmade, O. A., Ajala, M. O. & hyperplasia due to 21-hydroxylase deficiency. and management can be guided by morning cortisol.
Ohwovoriole, A. E. Adrenocortical function in N. Engl. J. Med. 383, 1248–1261 (2020). Eur. J. Endocrinol. 173, 633–642 (2015).
Nigerians with human immunodeficiency virus 30. White, P. C. & Speiser, P. W. Congenital adrenal 54. Schlaghecke, R., Kornely, E., Santen, R. T. &
infection. Ghana. Med. J. 47, 171–177 (2013). hyperplasia due to 21-hydroxylase deficiency. Ridderskamp, P. The effect of long-​term glucocorticoid
7. Tripathy, S. K., Agrawala, R. K. & Baliarsinha, A. K. Endocr. Rev. 21, 245–291 (2000). therapy on pituitary-​adrenal responses to exogenous
Endocrine alterations in HIV-​infected patients. 31. Bancos, I., Hahner, S., Tomlinson, J. & Arlt, W. corticotropin-​releasing hormone. N. Engl. J. Med.
Indian. J. Endocrinol. Metab. 19, 143–147 (2015). Diagnosis and management of adrenal insufficiency. 326, 226–230 (1992).
8. Broersen, L. H., Pereira, A. M., Jorgensen, J. O. & Lancet Diabetes Endocrinol. 3, 216–226 (2015). 55. Mebrahtu, T. F. et al. Dose dependency of iatrogenic
Dekkers, O. M. Adrenal insufficiency in corticosteroids 32. Michels, A. W. & Gottlieb, P. A. Autoimmune glucocorticoid excess and adrenal insufficiency and
use: systematic review and meta-​analysis. J. Clin. polyglandular syndromes. Nat. Rev. Endocrinol. 6, mortality: a cohort study in England. J. Clin.
Endocrinol. Metab. 100, 2171–2180 (2015). 270–277 (2010). Endocrinol. Metab. 104, 3757–3767 (2019).
9. Bleicken, B., Hahner, S., Ventz, M. & Quinkler, M. 33. Husebye, E. S., Anderson, M. S. & Kampe, O. 56. Pofi, R. et al. The short Synacthen (corticotropin) test
Delayed diagnosis of adrenal insufficiency is common: Autoimmune polyendocrine syndromes. N. Engl. can be used to predict recovery of hypothalamo-​
a cross-​sectional study in 216 patients. Am. J. Med. Sci. J. Med. 378, 2543–2544 (2018). pituitary-adrenal axis function. J. Clin. Endocrinol.
339, 525–531 (2010). 34. Dalin, F. et al. Clinical and immunological Metab. 103, 3050–3059 (2018).
10. Allolio, B. Extensive expertise in endocrinology characteristics of autoimmune Addison disease: A retrospective analysis of 776 patients with
adrenal crisis. Eur. J. Endocrinol. 172, R115–R124 a nationwide Swedish multicenter study. J. Clin. reversible causes of AI who had at least two short
(2015). Endocrinol. Metab. 102, 379–389 (2017). Synacthen tests performed to predict recovery of
11. Hahner, S. et al. High incidence of adrenal crisis in 35. Reato, G. et al. Premature ovarian failure in patients HPA axis function.
educated patients with chronic adrenal insufficiency: with autoimmune Addison’s disease: clinical, genetic, 57. Joseph, R. M., Hunter, A. L., Ray, D. W. & Dixon, W. G.
a prospective study. J. Clin. Endocrinol. Metab. 100, and immunological evaluation. J. Clin. Endocrinol. Systemic glucocorticoid therapy and adrenal
407–416 (2015). Metab. 96, E1255–E1261 (2011). insufficiency in adults: a systematic review.
12. Puar, T. H., Stikkelbroeck, N. M., Smans, L. C., 36. Charmandari, E., Nicolaides, N. C. & Chrousos, G. P. Semin. Arthritis Rheum. 46, 133–141 (2016).
Zelissen, P. M. & Hermus, A. R. Adrenal crisis: still a Adrenal insufficiency. Lancet 383, 2152–2167 58. Leong, S. H., Shander, S. & Ratnasingam, J. Predicting
deadly event in the 21st century. Am. J. Med. 129, (2014). recovery of the hypothalamic-​pituitary-adrenal axis
339 e331–339 e339 (2016). 37. Regal, M., Paramo, C., Sierra, S. M. & after prolonged glucocorticoid use. Endocr. Pract. 24,
13. Rushworth, R. L., Torpy, D. J. & Falhammar, H. Adrenal Garcia-​Mayor, R. V. Prevalence and incidence of 14–20 (2018).
crises: perspectives and research directions. Endocrine hypopituitarism in an adult Caucasian population in 59. Odenwald, B. et al. Children with classic congenital
55, 336–345 (2017). northwestern Spain. Clin. Endocrinol. 55, 735–740 adrenal hyperplasia experience salt loss and
14. Smans, L. C., Van der Valk, E. S., Hermus, A. R. & (2001). hypoglycemia: evaluation of adrenal crises during the
Zelissen, P. M. Incidence of adrenal crisis in patients 38. Bates, A. S., Van’t Hoff, W., Jones, P. J. & Clayton, R. N. first 6 years of life. Eur. J. Endocrinol. 174, 177–186
with adrenal insufficiency. Clin. Endocrinol. 84, 17–22 The effect of hypopituitarism on life expectancy. (2016).
(2016). J. Clin. Endocrinol. Metab. 81, 1169–1172 (1996). 60. Reisch, N. et al. Frequency and causes of adrenal
15. Bornstein, S. R. et al. Diagnosis and treatment of 39. Tomlinson, J. W. et al. Association between premature crises over lifetime in patients with 21-hydroxylase
primary adrenal insufficiency: an Endocrine Society mortality and hypopituitarism. West Midlands deficiency. Eur. J. Endocrinol. 167, 35–42
clinical practice guideline. J. Clin. Endocrinol. Metab. Prospective Hypopituitary Study Group. Lancet 357, (2012).
101, 364–389 (2016). 425–431 (2001). 61. Ritzel, K. et al. Clinical review: outcome of bilateral
Practice guidelines on the management of adrenal A prospective study including 1,014 patients adrenalectomy in Cushing’s syndrome: a systematic
insufficiency from an international expert panel. with hypopituitarism investigating the total review. J. Clin. Endocrinol. Metab. 98, 3939–3948
16. Dunlop, D. Eighty-​six cases of Addison’s disease. and specific-cause mortality compared with an (2013).
Br. Med. J. 2, 887–891 (1963). age-​matched and sex-matched UK population. 62. Meyer, G., Badenhoop, K. & Linder, R. Addison’s
17. Laureti, S., Vecchi, L., Santeusanio, F. & Falorni, A. 40. Carosi, G. et al. Hypothalamic-​pituitary axis in disease with polyglandular autoimmunity carries a
Is the prevalence of Addison’s disease underestimated? non-​functioning pituitary adenomas: focus on the more than 2.5-fold risk for adrenal crises: German
J. Clin. Endocrinol. Metab. 84, 1762 (1999). prevalence of isolated central hypoadrenalism. health insurance data 2010-2013. Clin. Endocrinol.
18. Hellesen, A., Bratland, E. & Husebye, E. S. Neuroendocrinology 102, 267–273 (2015). 85, 347–353 (2016).
Autoimmune Addison’s disease – an update on 41. Appelman-​Dijkstra, N. M. et al. Pituitary dysfunction 63. Papierska, L. & Rabijewski, M. Delay in diagnosis
pathogenesis. Ann. Endocrinol. 79, 157–163 (2018). in adult patients after cranial radiotherapy: systematic of adrenal insufficiency is a frequent cause of
19. Mason, A. S., Meade, T. W., Lee, J. A. & Morris, J. N. review and meta-​analysis. J. Clin. Endocrinol. Metab. adrenal crisis. Int. J. Endocrinol. 2013, 482370
Epidemiological and clinical picture of Addison’s 96, 2330–2340 (2011). (2013).
disease. Lancet 2, 744–747 (1968). 42. Jenkins, P. J., Bates, P., Carson, M. N., Stewart, P. M. 64. Gidlof, S. et al. One hundred years of congenital
20. Olafsson, A. S. & Sigurjonsdottir, H. A. Increasing & Wass, J. A. Conventional pituitary irradiation is adrenal hyperplasia in Sweden: a retrospective,
prevalence of Addison disease: results from a effective in lowering serum growth hormone and population-​based cohort study. Lancet Diabetes
nationwide study. Endocr. Pract. 22, 30–35 (2016). insulin-​like growth factor-​I in patients with acromegaly. Endocrinol. 1, 35–42 (2013).
21. Bjornsdottir, S. et al. Drug prescription patterns in J. Clin. Endocrinol. Metab. 91, 1239–1245 (2006). 65. Bergthorsdottir, R., Leonsson-​Zachrisson, M., Oden, A.
patients with Addison’s disease: a Swedish population-​ 43. Knappe, U. J. et al. Fractionated radiotherapy & Johannsson, G. Premature mortality in patients with
based cohort study. J. Clin. Endocrinol. Metab. 98, and radiosurgery in acromegaly: analysis of 352 Addison’s disease: a population-​based study.
2009–2018 (2013). patients from the German Acromegaly Registry. J. Clin. Endocrinol. Metab. 91, 4849–4853
22. Meyer, G., Neumann, K., Badenhoop, K. & Linder, R. Eur. J. Endocrinol. 182, 275–284 (2020). (2006).
Increasing prevalence of Addison’s disease in German 44. Imber, B. S., Lee, H. S., Kunwar, S., Blevins, L. S. & A population-​based, retrospective, observational
females: health insurance data 2008-2012. Eur. J. Aghi, M. K. Hypophysitis: a single-​center case series. study from Sweden showing that the risk ratio
Endocrinol. 170, 367–373 (2014). Pituitary 18, 630–641 (2015). for death is more than twofold higher in patients
23. Stewart, P. M. et al. Exploring inpatient 45. Wang, S. et al. Primary lymphocytic hypophysitis: with Addison disease than in the background
hospitalizations and morbidity in patients with clinical characteristics and treatment of 50 cases in a population.
adrenal insufficiency. J. Clin. Endocrinol. Metab. 101, single centre in China over 18 years. Clin. Endocrinol. 66. Bensing, S. et al. Increased death risk and altered
4843–4850 (2016). 87, 177–184 (2017). cancer incidence pattern in patients with isolated
24. Willis, A. C. & Vince, F. P. The prevalence of Addison’s 46. Turcu, A. F. et al. Pituitary stalk lesions: the Mayo or combined autoimmune primary adrenocortical
disease in Coventry, UK. Postgrad. Med. J. 73, Clinic experience. J. Clin. Endocrinol. Metab. 98, insufficiency. Clin. Endocrinol. 69, 697–704
286–288 (1997). 1812–1818 (2013). (2008).
25. Chantzichristos, D. et al. Incidence, prevalence and 47. Lu, J., Li, L., Lan, Y., Liang, Y. & Meng, H. Immune 67. Erichsen, M. M. et al. Normal overall mortality rate
seasonal onset variation of Addison’s disease among checkpoint inhibitor-​associated pituitary-​adrenal in Addison’s disease, but young patients are at risk of
persons with type 1 diabetes mellitus: nationwide, dysfunction: a systematic review and meta-​analysis. premature death. Eur. J. Endocrinol. 160, 233–237
matched cohort studies. Eur. J. Endocrinol. 178, Cancer Med. 8, 7503–7515 (2019). (2009).
113–120 (2018). 48. Barroso-​Sousa, R. et al. Incidence of endocrine 68. Falhammar, H. et al. Increased mortality in patients
A nationwide, matched, observational cohort study dysfunction following the use of different immune with congenital adrenal hyperplasia due to
investigating the incidence and prevalence of checkpoint inhibitor regimens: a systematic review 21-hydroxylase deficiency. J. Clin. Endocrinol. Metab.
primary adrenal insufficiency in Sweden. and meta-​analysis. JAMA Oncol. 4, 173–182 (2018). 99, E2715–E2721 (2014).

20 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

69. Hahner, S. et al. Epidemiology of adrenal crisis in 91. Henley, D., Lightman, S. & Carrell, R. Cortisol and 116. Lamprecht, A., Sorbello, J., Jang, C., Torpy, D. J.
chronic adrenal insufficiency: the need for new CBG – Getting cortisol to the right place at the right & Inder, W. J. Secondary adrenal insufficiency and
prevention strategies. Eur. J. Endocrinol. 162, time. Pharmacol. Ther. 166, 128–135 (2016). pituitary dysfunction in oral/transdermal opioid
597–602 (2010). 92. Stowasser, M., Ahmed, A. H., Pimenta, E., Taylor, P. J. users with non-​cancer pain. Eur. J. Endocrinol. 179,
70. El-​Maouche, D. et al. Longitudinal assessment & Gordon, R. D. Factors affecting the aldosterone/ 353–362 (2018).
of illnesses, stress dosing, and illness sequelae in renin ratio. Horm. Metab. Res. 44, 170–176 117. Fraser, L. A. et al. Oral opioids for chronic non-​cancer
patients with congenital adrenal hyperplasia. J. Clin. (2012). pain: higher prevalence of hypogonadism in men than
Endocrinol. Metab. 103, 2336–2345 (2018). 93. Atlas, S. A. The renin-​angiotensin aldosterone system: in women. Exp. Clin. Endocrinol. Diabetes 117, 38–43
71. Rushworth, R. L., Chrisp, G. L., Dean, B., Falhammar, H. pathophysiological role and pharmacologic inhibition. (2009).
& Torpy, D. J. Hospitalisation in children with adrenal J. Manag. Care Pharm. 13, 9–20 (2007). 118. Fountas, A., Van Uum, S. & Karavitaki, N. Opioid-​
insufficiency and hypopituitarism: is there a differential 94. Tucci, J. R., Espiner, E. A., Jagger, P. I., Pauk, G. L. & induced endocrinopathies. Lancet Diabetes
burden between boys and girls and between age Lauler, D. P. ACTH stimulation of aldosterone secretion Endocrinol. 8, 68–80 (2020).
groups? Horm. Res. Paediatr. 88, 339–346 (2017). in normal subjects and in patients with chronic 119. Pekic, S. & Popovic, V. Diagnosis of endocrine disease:
72. Rushworth, R. L., Falhammar, H., Munns, C. F., adrenocortical insufficiency. J. Clin. Endocrinol. Metab. expanding the cause of hypopituitarism. Eur. J.
Maguire, A. M. & Torpy, D. J. Hospital admission 27, 568–575 (1967). Endocrinol. 176, R269–R282 (2017).
patterns in children with CAH: admission rates and 95. Kem, D. C. et al. Plasma aldosterone response to 120. Higham, C. E., Johannsson, G. & Shalet, S. M.
adrenal crises decline with age. Int. J. Endocrinol. ACTH in primary aldosteronism and in patients with Hypopituitarism. Lancet 388, 2403–2415 (2016).
2016, 5748264 (2016). low renin hypertension. J. Clin. Endocrinol. Metab. 46, 121. Wu, W. et al. Mutations in PROP1 cause familial
73. Rushworth, R. L. & Torpy, D. J. A descriptive study 552–560 (1978). combined pituitary hormone deficiency. Nat. Genet.
of adrenal crises in adults with adrenal insufficiency: 96. Bollag, W. B. Regulation of aldosterone synthesis 18, 147–149 (1998).
increased risk with age and in those with bacterial and secretion. Compr. Physiol. 4, 1017–1055 122. Jullien, N. et al. Clinical lessons learned in
infections. BMC Endocr. Disord. 14, 79 (2014). (2014). constitutional hypopituitarism from two decades
74. Rushworth, R. L. & Torpy, D. J. Modern hydrocortisone 97. Mitchell, A. L. & Pearce, S. H. Autoimmune Addison of experience in a large international cohort.
replacement regimens in adrenal insufficiency patients disease: pathophysiology and genetic complexity. Clin. Endocrinol. 94, 277–289 (2021).
and the risk of adrenal crisis. Horm. Metab. Res. 47, Nat. Rev. Endocrinol. 8, 306–316 (2012). 123. Tutunculer, F., Saka, N., Arkaya, S. C., Abbasoglu, S.
637–642 (2015). 98. Winqvist, O., Karlsson, F. A. & Kampe, O. & Bas, F. Evaluation of adrenomedullary function in
75. Rushworth, R. L. & Torpy, D. J. Adrenal insufficiency 21-Hydroxylase, a major autoantigen in idiopathic patients with congenital adrenal hyperplasia.
in Australia: is it possible that the use of lower dose, Addison’s disease. Lancet 339, 1559–1562 (1992). Horm. Res. 72, 331–336 (2009).
short-​acting glucocorticoids has increased the risk 99. Naletto, L. et al. The natural history of autoimmune 124. Morita, S. et al. Reduced epinephrine reserve in
of adrenal crises? Horm. Metab. Res. 47, 427–432 Addison’s disease from the detection of autoantibodies response to insulin-​induced hypoglycemia in patients
(2015). to development of the disease: a long follow-​up study with pituitary adenoma. Eur. J. Endocrinol. 157,
76. Okamoto, M. The metabolism of cortisol in on 143 patients. Eur. J. Endocrinol. 180, 223–234 265–270 (2007).
hyperthyroidism and Cushing’s syndrome. Endocrinol. (2019). 125. Merke, D. P. et al. Adrenomedullary dysplasia and
Jpn. 10, 159–168 (1963). 100. Dawoodji, A. et al. High frequency of cytolytic hypofunction in patients with classic 21-hydroxylase
77. Fonseca, V., Brown, R., Hochhauser, D., Ginsburg, J. 21-hydroxylase-​specific CD8+ T cells in autoimmune deficiency. N. Engl. J. Med. 343, 1362–1368
& Havard, C. W. Acute adrenal crisis precipitated by Addison’s disease patients. J. Immunol. 193, (2000).
thyroxine. Br. Med. J. 292, 1185–1186 (1986). 2118–2126 (2014). 126. Bornstein, S. R., Breidert, M., Ehrhart-​Bornstein, M.,
78. Rushworth, R. L., Chrisp, G. L. & Torpy, D. J. 101. Bratland, E., Skinningsrud, B., Undlien, D. E., Kloos, B. & Scherbaum, W. A. Plasma catecholamines
Glucocorticoid-​induced adrenal insufficiency: a study Mozes, E. & Husebye, E. S. T cell responses to steroid in patients with Addison’s disease. Clin. Endocrinol.
of the incidence in hospital patients and a review cytochrome P450 21-hydroxylase in patients with 42, 215–218 (1995).
of peri-​operative management. Endocr. Pract. 24, autoimmune primary adrenal insufficiency. J. Clin. 127. Mpoy, M. & Kolanowski, J. Urinary catecholamine
437–445 (2018). Endocrinol. Metab. 94, 5117–5124 (2009). excretion in patients with secondary adrenocortical
79. Rushworth, R. L., Slobodian, P. & Torpy, D. J. 102. Eriksson, D. et al. Common genetic variation in the insufficiency. J. Endocrinol. Invest. 9, 253–255
Interruptions to supply of high-​dose hydrocortisone autoimmune regulator (AIRE) locus is associated with (1986).
tablets and the incidence of adrenal crises. Clin. autoimmune Addison’s disease in Sweden. Sci. Rep. 8, 128. Kim, M. S. et al. Decreased adrenomedullary function
Endocrinol. 83, 999–1000 (2015). 8395 (2018). in infants with classical congenital adrenal hyperplasia.
80. Gjerstad, J. K., Lightman, S. L. & Spiga, F. Role of 103. Skov, J. et al. Heritability of Addison’s disease and J. Clin. Endocrinol. Metab. 99, E1597–E1601 (2014).
glucocorticoid negative feedback in the regulation prevalence of associated autoimmunity in a cohort 129. Geiger, A. M., Pitts, K. P., Feldkamp, J., Kirschbaum, C.
of HPA axis pulsatility. Stress 21, 403–416 of 112,100 Swedish twins. Endocrine 58, 521–527 & Wolf, J. M. Cortisol-​dependent stress effects on cell
(2018). (2017). distribution in healthy individuals and individuals
81. Lightman, S. L. & Conway-​Campbell, B. L. The crucial 104. Loeb, R. F. Chemical changes in the blood in Addison’s suffering from chronic adrenal insufficiency. Brain
role of pulsatile activity of the HPA axis for continuous disease. Science 76, 420–421 (1932). Behav. Immun. 50, 241–248 (2015).
dynamic equilibration. Nat. Rev. Neurosci. 11, 105. Han, T. S., Walker, B. R., Arlt, W. & Ross, R. J. 130. Wurtman, R. J. & Axelrod, J. Control of enzymatic
710–718 (2010). Treatment and health outcomes in adults with synthesis of adrenaline in the adrenal medulla by
82. Roelfsema, F., Aoun, P. & Veldhuis, J. D. Pulsatile congenital adrenal hyperplasia. Nat. Rev. Endocrinol. adrenal cortical steroids. J. Biol. Chem. 241,
cortisol feedback on ACTH secretion is mediated 10, 115–124 (2014). 2301–2305 (1966).
by the glucocorticoid receptor and modulated by 106. Kemp, S., Huffnagel, I. C., Linthorst, G. E., 131. Ehrhart-​Bornstein, M., Hinson, J. P., Bornstein, S. R.,
gender. J. Clin. Endocrinol. Metab. 101, 4094–4102 Wanders, R. J. & Engelen, M. Adrenoleukodystrophy – Scherbaum, W. A. & Vinson, G. P. Intraadrenal
(2016). neuroendocrine pathogenesis and redefinition of interactions in the regulation of adrenocortical
83. Walker, J. J., Terry, J. R. & Lightman, S. L. Origin of natural history. Nat. Rev. Endocrinol. 12, 606–615 steroidogenesis. Endocr. Rev. 19, 101–143 (1998).
ultradian pulsatility in the hypothalamic-​pituitary- (2016). 132. To, T. T. et al. Pituitary-​interrenal interaction in
adrenal axis. Proc. Biol. Sci. 277, 1627–1633 107. Laureti, S. et al. X-​linked adrenoleukodystrophy is zebrafish interrenal organ development.
(2010). a frequent cause of idiopathic Addison’s disease in Mol. Endocrinol. 21, 472–485 (2007).
84. Lightman, S. L., Birnie, M. T. & Conway-​Campbell, B. L. young adult male patients. J. Clin. Endocrinol. Metab. 133. Weise, M. et al. Patients with classic congenital
Dynamics of ACTH and cortisol secretion and 81, 470–474 (1996). adrenal hyperplasia have decreased epinephrine
implications for disease. Endocr. Rev. 41, 470–490 108. Bezman, L. & Moser, H. W. Incidence of X-​linked reserve and defective glucose elevation in response to
(2020). adrenoleukodystrophy and the relative frequency of high-​intensity exercise. J. Clin. Endocrinol. Metab. 89,
85. Oster, H. et al. The functional and clinical significance its phenotypes. Am. J. Med. Genet. 76, 415–419 591–597 (2004).
of the 24-hour rhythm of circulating glucocorticoids. (1998). 134. Elbelt, U., Hahner, S. & Allolio, B. Altered insulin
Endocr. Rev. 38, 3–45 (2017). 109. Hannah-​Shmouni, F. & Stratakis, C. A. An overview of requirement in patients with type 1 diabetes and
86. Caratti, G. et al. Glucocorticoid receptor function in inborn errors of metabolism manifesting with primary primary adrenal insufficiency receiving standard
health and disease. Clin. Endocrinol. 83, 441–448 adrenal insufficiency. Rev. Endocr. Metab. Disord. 19, glucocorticoid replacement therapy. Eur. J. Endocrinol.
(2015). 53–67 (2018). 160, 919–924 (2009).
87. Chrousos, G. P. in Endocrinology 7th edition 110. Narumi, S. Rare monogenic causes of primary adrenal 135. Orentreich, N., Brind, J. L., Rizer, R. L. &
(eds Jameson, J. L. & De Groot, L.) 1727–1740.e5 insufficiency. Curr. Opin. Endocrinol. Diabetes Obes. Vogelman, J. H. Age changes and sex differences in
(Elsevier, 2016). 25, 172–177 (2018). serum dehydroepiandrosterone sulfate concentrations
88. Stavreva, D. A. et al. Ultradian hormone stimulation 111. Tanriverdi, F. et al. Pituitary dysfunction after throughout adulthood. J. Clin. Endocrinol. Metab. 59,
induces glucocorticoid receptor-​mediated pulses of traumatic brain injury: a clinical and pathophysiological 551–555 (1984).
gene transcription. Nat. Cell Biol. 11, 1093–1102 approach. Endocr. Rev. 36, 305–342 (2015). 136. Korth-​Schutz, S., Levine, L. S. & New, M. I. Serum
(2009). 112. Gubbi, S., Hannah-​Shmouni, F., Stratakis, C. A. androgens in normal prepubertal and pubertal
This study showed that an ultradian administration & Koch, C. A. Primary hypophysitis and other children and in children with precocious adrenarche.
of glucocorticoids induces cyclic GR-​mediated autoimmune disorders of the sellar and suprasellar J. Clin. Endocrinol. Metab. 42, 117–124 (1976).
transcriptional regulation in cultured cells and in regions. Rev. Endocr. Metab. Disord. 19, 335–347 137. Lebbe, M. et al. The steroid metabolome in the
animal models. (2018). isolated ovarian follicle and its response to androgen
89. Kalafatakis, K. et al. Ultradian rhythmicity of plasma 113. Faje, A. Immunotherapy and hypophysitis: clinical exposure and antagonism. Endocrinology 158,
cortisol is necessary for normal emotional and presentation, treatment, and biologic insights. 1474–1485 (2017).
cognitive responses in man. Proc. Natl Acad. Sci. USA Pituitary 19, 82–92 (2016). 138. Gleicher, N., Weghofer, A. & Barad, D. H. The role
115, E4091–E4100 (2018). 114. Caturegli, P. et al. Autoimmune hypophysitis. of androgens in follicle maturation and ovulation
90. Chan, W. L., Carrell, R. W., Zhou, A. & Read, R. J. Endocr. Rev. 26, 599–614 (2005). induction: friend or foe of infertility treatment?
How changes in affinity of corticosteroid-​binding 115. AbdelRazek, M. A., Venna, N. & Stone, J. H. Reprod. Biol. Endocrinol. 9, 116 (2011).
globulin modulate free cortisol concentration. IgG4-related disease of the central and peripheral 139. Lang, K., Burger-​Stritt, S. & Hahner, S. Is DHEA
J. Clin. Endocrinol. Metab. 98, 3315–3322 (2013). nervous systems. Lancet Neurol. 17, 183–192 (2018). replacement beneficial in chronic adrenal failure?


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 21

0123456789();:
Primer

Best Pract. Res. Clin. Endocrinol. Metab. 29, 25–32 162. Oelkers, W. The role of high- and low-​dose Association of Anaesthetists, the Royal College of
(2015). corticotropin tests in the diagnosis of secondary Physicians and the Society for Endocrinology UK.
140. Labrie, F. et al. Structure, regulation and role of adrenal insufficiency. Eur. J. Endocrinol. 139, Anaesthesia 75, 654–663 (2020).
3β-​hydroxysteroid dehydrogenase, 17β-​hydroxysteroid 567–570 (1998). 183. Grossman, A., Johannsson, G., Quinkler, M. &
dehydrogenase and aromatase enzymes in the 163. Lutz, A. et al. Adrenocortical function in patients Zelissen, P. Therapy of endocrine disease: perspectives
formation of sex steroids in classical and peripheral with macrometastases of the adrenal gland. on the management of adrenal insufficiency: clinical
intracrine tissues. Baillieres Clin. Endocrinol. Metab. Eur. J. Endocrinol. 143, 91–97 (2000). insights from across Europe. Eur. J. Endocrinol. 169,
8, 451–474 (1994). 164. Mao, J. J., Dages, K. N., Suresh, M. & Bancos, I. R165–R175 (2013).
141. Pluchino, N. et al. Neurobiology of DHEA and effects Presentation, disease progression and outcomes 184. Debono, M. et al. Modified-​release hydrocortisone to
on sexuality, mood and cognition. J. Steroid Biochem. of adrenal gland metastases. Clin. Endocrinol. 93, provide circadian cortisol profiles. J. Clin. Endocrinol.
Mol. Biol. 145, 273–280 (2015). 546–554 (2020). Metab. 94, 1548–1554 (2009).
142. Arlt, W. et al. Dehydroepiandrosterone replacement 165. Petersenn, S., Honegger, J. & Quinkler, M. National 185. Hahner, S. et al. Impaired subjective health status in
in women with adrenal insufficiency. N. Engl. J. Med. German audit of diagnosis, treatment, and teaching 256 patients with adrenal insufficiency on standard
341, 1013–1020 (1999). in secondary adrenal insufficiency. Horm. Metab. Res. therapy based on cross-​sectional analysis. J. Clin.
A double-​blind clinical study investigating DHEA in 49, 580–588 (2017). Endocrinol. Metab. 92, 3912–3922 (2007).
women with AI demonstrating improved well-​being 166. Schmidt, I. L., Lahner, H., Mann, K. & Petersenn, S. A cross-​sectional study showing that patients with
and sexuality. Diagnosis of adrenal insufficiency: evaluation of the AI on current standard replacement suffer from
143. Sapolsky, R. M., Romero, L. M. & Munck, A. U. corticotropin-​releasing hormone test and basal serum significantly impaired health-​related subjective
How do glucocorticoids influence stress responses? cortisol in comparison to the insulin tolerance test health status, irrespective of origin of disease or
Integrating permissive, suppressive, stimulatory, and in patients with hypothalamic-​pituitary-adrenal concomitant disease.
preparative actions. Endocr. Rev. 21, 55–89 (2000). disease. J. Clin. Endocrinol. Metab. 88, 4193–4198 186. Johannsson, G. et al. Improved cortisol exposure-​time
144. Besse, J. C. & Bass, A. D. Potentiation by (2003). profile and outcome in patients with adrenal
hydrocortisone of responses to catecholamines in 167. Grossman, A. B. The diagnosis and management of insufficiency: a prospective randomized trial of a
vascular smooth muscle. J. Pharmacol. Exp. Ther. central hypoadrenalism. J. Clin. Endocrinol. Metab. novel hydrocortisone dual-​release formulation.
154, 224–238 (1966). 95, 4855–4863 (2010). J. Clin. Endocrinol. Metab. 97, 473–481 (2012).
145. Kalsner, S. Mechanism of hydrocortisone potentiation 168. Erturk, E., Jaffe, C. A. & Barkan, A. L. Evaluation of A randomized trial comparing thrice-​daily
of responses to epinephrine and norepinephrine in the integrity of the hypothalamic-​pituitary-adrenal conventional hydrocortisone replacement with
rabbit aorta. Circ. Res. 24, 383–395 (1969). axis by insulin hypoglycemia test. J. Clin. Endocrinol. once-​daily dual-​release hydrocortisone.
146. Prete, A. et al. Prevention of adrenal crisis: cortisol Metab. 83, 2350–2354 (1998). 187. European Medicines Agency. Summary of Product
responses to major stress compared to stress dose 169. Hurel, S. J. et al. The short Synacthen and insulin Characteristics: Plenadren. https://www.ema.europa.
hydrocortisone delivery. J. Clin. Endocrinol. Metab. stress tests in the assessment of the hypothalamic-​ eu/en/medicines/human/EPAR/plenadren (2018).
105, 2262–2274 (2020). pituitary-adrenal axis. Clin. Endocrinol. 44, 141–146 188. Giordano, R. et al. Improvement of anthropometric
This study provides an evidence base on the (1996). and metabolic parameters, and quality of life following
steroidogenic response of the adrenal cortex in 170. Reynolds, R. M., Stewart, P. M., Seckl, J. R. & treatment with dual-​release hydrocortisone in patients
case of major stress (major trauma, sepsis and Padfield, P. L. Assessing the HPA axis in patients with with Addison’s disease. Endocrine 51, 360–368
combat stress). pituitary disease: a UK survey. Clin. Endocrinol. 64, (2016).
147. Udelsman, R. et al. Adaptation during surgical stress. 82–85 (2006). 189. Quinkler, M., Miodini Nilsen, R., Zopf, K., Ventz, M.
A reevaluation of the role of glucocorticoids. J. Clin. 171. Mukherjee, J. J. et al. A comparison of the insulin & Oksnes, M. Modified-​release hydrocortisone
Invest. 77, 1377–1381 (1986). tolerance/glucagon test with the short ACTH decreases BMI and HbA1c in patients with primary
148. Quinkler, M., Oelkers, W., Remde, H. & Allolio, B. stimulation test in the assessment of the and secondary adrenal insufficiency. Eur. J. Endocrinol.
Mineralocorticoid substitution and monitoring in hypothalamo-​pituitary-adrenal axis in the early 172, 619–626 (2015).
primary adrenal insufficiency. Best Pract. Res. Clin. post-​operative period after hypophysectomy. 190. Isidori, A. M. et al. Effect of once-​daily, modified-​
Endocrinol. Metab. 29, 17–24 (2015). Clin. Endocrinol. 47, 51–60 (1997). release hydrocortisone versus standard glucocorticoid
149. Munck, A., Guyre, P. M. & Holbrook, N. J. 172. Ospina, N. S. et al. ACTH stimulation tests for the therapy on metabolism and innate immunity in
Physiological functions of glucocorticoids in stress and diagnosis of adrenal insufficiency: systematic review patients with adrenal insufficiency (DREAM): a single-​
their relation to pharmacological actions. Endocr. Rev. and meta-​analysis. J. Clin. Endocrinol. Metab. 101, blind, randomised controlled trial. Lancet Diabetes
5, 25–44 (1984). 427–434 (2016). Endocrinol. 6, 173–185 (2018).
150. Besedovsky, H., del Rey, A., Sorkin, E. & Dinarello, C. A. 173. Steiner, H., Bahr, V., Exner, P. & Oelkers, P. W. 191. Whittle, E. & Falhammar, H. Glucocorticoid regimens
Immunoregulatory feedback between interleukin-1 Pituitary function tests: comparison of ACTH and in the treatment of congenital adrenal hyperplasia:
and glucocorticoid hormones. Science 233, 652–654 11-deoxy-​cortisol responses in the metyrapone test a systematic review and meta-​analysis. J. Endocr. Soc.
(1986). and with the insulin hypoglycemia test. Exp. Clin. 3, 1227–1245 (2019).
151. Barber, A. E. et al. Glucocorticoid therapy alters Endocrinol. 102, 33–38 (1994). 192. Lovas, K. & Husebye, E. S. Continuous subcutaneous
hormonal and cytokine responses to endotoxin in 174. Berneis, K. et al. Combined stimulation of hydrocortisone infusion in Addison’s disease.
man. J. Immunol. 150, 1999–2006 (1993). adrenocorticotropin and compound-​S by single Eur. J. Endocrinol. 157, 109–112 (2007).
152. Morrow, L. E., McClellan, J. L., Conn, C. A. & dose metyrapone test as an outpatient procedure 193. Gagliardi, L. et al. Continuous subcutaneous
Kluger, M. J. Glucocorticoids alter fever and to assess hypothalamic-​pituitary-adrenal function. hydrocortisone infusion therapy in Addison’s disease:
IL-6 responses to psychological stress and to J. Clin. Endocrinol. Metab. 87, 5470–5475 a randomized, placebo-​controlled clinical trial.
lipopolysaccharide. Am. J. Physiol. 264, (2002). J. Clin. Endocrinol. Metab. 99, 4149–4157 (2014).
R1010–R1016 (1993). 175. Giordano, R. et al. Hypothalamus-​pituitary-adrenal 194. Nella, A. A. et al. A phase 2 study of continuous
153. Koniaris, L. G., Wand, G. & Wright, T. M. TNF mediates axis evaluation in patients with hypothalamo-​pituitary subcutaneous hydrocortisone infusion in adults with
a murine model of Addison’s crisis. Shock 15, 29–34 disorders: comparison of different provocative tests. congenital adrenal hyperplasia. J. Clin. Endocrinol.
(2001). Clin. Endocrinol. 68, 935–941 (2008). Metab. 101, 4690–4698 (2016).
154. Boonen, E. et al. Reduced cortisol metabolism during 176. Hamrahian, A. H. et al. Revised GH and cortisol 195. Whitaker, M. et al. An oral multiparticulate, modified-​
critical illness. N. Engl. J. Med. 368, 1477–1488 cut-​points for the glucagon stimulation test in the release, hydrocortisone replacement therapy that
(2013). evaluation of GH and hypothalamic-​pituitary-adrenal provides physiological cortisol exposure.
155. Husebye, E. S. et al. Consensus statement on the axes in adults: results from a prospective randomized Clin. Endocrinol. 80, 554–561 (2014).
diagnosis, treatment and follow-​up of patients with multicenter study. Pituitary 19, 332–341 (2016). 196. Mallappa, A. et al. A phase 2 study of Chronocort, a
primary adrenal insufficiency. J. Intern. Med. 275, 177. Deutschbein, T., Unger, N., Mann, K. & Petersenn, S. modified-​release formulation of hydrocortisone, in the
104–115 (2014). Diagnosis of secondary adrenal insufficiency in treatment of adults with classic congenital adrenal
156. Buonocore, F., McGlacken-​Byrne, S. M., Del Valle, I. patients with hypothalamic-​pituitary disease: hyperplasia. J. Clin. Endocrinol. Metab. 100,
& Achermann, J. C. Current insights into adrenal comparison between serum and salivary cortisol 1137–1145 (2015).
insufficiency in the newborn and young infant. during the high-​dose short synacthen test. Eur. J. Phase II study investigating the modified-​release
Front. Pediatr. 8, 619041 (2020). Endocrinol. 160, 9–16 (2009). hydrocortisone formulation Chronocort in 16
157. Saevik, A. B. et al. Clues for early detection of 178. Debono, M. et al. Salivary cortisone reflects cortisol adults with classic congenital adrenal hyperplasia.
autoimmune Addison’s disease – myths and realities. exposure under physiological conditions and after Twice-​daily Chronocort induced cortisol profiles
J. Intern. Med. 283, 190–199 (2018). hydrocortisone. J. Clin. Endocrinol. Metab. 101, similar to physiological cortisol secretion.
158. El-​Farhan, N. et al. Method-​specific serum cortisol 1469–1477 (2016). 197. Merke, D. P. et al. A phase 3 study of a modified-​
responses to the adrenocorticotrophin test: 179. Thil’en, A. et al. Benefits of neonatal screening for release hydrocortisone in the treatment of congenital
comparison of gas chromatography-​mass congenital adrenal hyperplasia (21-hydroxylase adrenal hyperplasia [abstract OR25-02]. J. Endocr.
spectrometry and five automated immunoassays. deficiency) in Sweden. Pediatrics 101, E11 Soc. 4 (Suppl. 1), A107–A108 (2020).
Clin. Endocrinol. 78, 673–680 (2013). (1998). 198. Turcu, A. F. et al. Single-​dose study of a corticotropin-​
159. Ueland, G. A. et al. The short cosyntropin test 180. Kishore Kumar, R., Das, H. & Kini, P. Newborn releasing factor receptor-1 antagonist in women with
revisited: new normal reference range using screening for congenital adrenal hyperplasia in India: 21-hydroxylase deficiency. J. Clin. Endocrinol. Metab.
LC-​MS/MS. J. Clin. Endocrinol. Metab. 103, what do we need to watch out for? J. Obstet. 101, 1174–1180 (2016).
1696–1703 (2018). Gynaecol. India 66, 415–419 (2016). 199. Saevik, A. B. et al. Residual corticosteroid production
160. Dickstein, G. et al. Adrenocorticotropin stimulation 181. Yanase, T. et al. Diagnosis and treatment of adrenal in autoimmune Addison disease. J. Clin. Endocrinol.
test: effects of basal cortisol level, time of day, and insufficiency including adrenal crisis: a Japan Metab. 105, 2430–2441 (2020).
suggested new sensitive low dose test. J. Clin. Endocrine Society clinical practice guideline [Opinion]. 200. Pofi, R. et al. Plasma renin measurements are
Endocrinol. Metab. 72, 773–778 (1991). Endocr. J. 63, 765–784 (2016). unrelated to mineralocorticoid replacement dose
161. May, M. E. & Carey, R. M. Rapid adrenocorticotropic 182. Woodcock, T. et al. Guidelines for the management of in patients with primary adrenal insufficiency.
hormone test in practice. retrospective review. glucocorticoids during the peri-​operative period for J. Clin. Endocrinol. Metab. 105, 314–326
Am. J. Med. 79, 679–684 (1985). patients with adrenal insufficiency: guidelines from the (2020).

22 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:
Primer

201. Oelkers, W., Diederich, S. & Bahr, V. Diagnosis and 220. Lovas, K. et al. Glucocorticoid replacement 238. Ventura, M. et al. The spectrum of pediatric adrenal
therapy surveillance in Addison’s disease: rapid therapy and pharmacogenetics in Addison’s disease: insufficiency: insights from 34 years of experience.
adrenocorticotropin (ACTH) test and measurement effects on bone. Eur. J. Endocrinol. 160, 993–1002 J. Pediatr. Endocrinol. Metab. 32, 721–726 (2019).
of plasma ACTH, renin activity, and aldosterone. (2009). 239. Sterns, R. H. Disorders of plasma sodium–causes,
J. Clin. Endocrinol. Metab. 75, 259–264 (1992). 221. Koetz, K. R., Ventz, M., Diederich, S. & Quinkler, M. consequences, and correction. N. Engl. J. Med. 372,
202. Schultebraucks, K., Wingenfeld, K., Otte, C. & Bone mineral density is not significantly reduced in 55–65 (2015).
Quinkler, M. The role of fludrocortisone in cognition adult patients on low-​dose glucocorticoid replacement 240. Kampmeyer, D., Lehnert, H., Moenig, H., Haas, C. S. &
and mood in patients with primary adrenal therapy. J. Clin. Endocrinol. Metab. 97, 85–92 Harbeck, B. A strong need for improving the education
insufficiency (Addison’s disease). Neuroendocrinology (2012). of physicians on glucocorticoid replacement treatment
103, 315–320 (2016). Systematic assessment of BMD in 122 patients in adrenal insufficiency: an interdisciplinary and
203. Esposito, D., Pasquali, D. & Johannsson, G. Primary with AI demonstrating normal BMD values on low multicentre evaluation. Eur. J. Intern. Med. 33,
adrenal insufficiency: managing mineralocorticoid glucocorticoid replacement doses. e13–e15 (2016).
replacement therapy. J. Clin. Endocrinol. Metab. 103, 222. Schulz, J. et al. Reduction in daily hydrocortisone dose 241. Harbeck, B. et al. Glucocorticoid replacement therapy
376–387 (2018). improves bone health in primary adrenal insufficiency. in adrenal insufficiency–a challenge to physicians?
204. Flad, T. M., Conway, J. D., Cunningham, S. K. & Eur. J. Endocrinol. 174, 531–538 (2016). Endocr. J. 62, 463–468 (2015).
McKenna, T. J. The role of plasma renin activity 223. Bjornsdottir, S. et al. Risk of hip fracture in Addison’s 242. Hahner, S., Burger-​Stritt, S. & Allolio, B. Subcutaneous
in evaluating the adequacy of mineralocorticoid disease: a population-​based cohort study. J. Intern. hydrocortisone administration for emergency use
replacement in primary adrenal insufficiency. Med. 270, 187–195 (2011). in adrenal insufficiency. Eur. J. Endocrinol. 169,
Clin. Endocrinol. 45, 529–534 (1996). 224. Ragnarsson, O., Nystrom, H. F. & Johannsson, G. 147–154 (2013).
205. Cohen, N., Gilbert, R., Wirth, A., Casley, D. & Glucocorticoid replacement therapy is independently 243. Burger-​Stritt, S., Bachmann, L., Kurlbaum, M. &
Jerums, G. Atrial natriuretic peptide and plasma renin associated with reduced bone mineral density in Hahner, S. Emergency treatment of adrenal crisis with
levels in assessment of mineralocorticoid replacement women with hypopituitarism. Clin. Endocrinol. 76, prednisone suppositories: a bioequivalence study in
in Addison’s disease. J. Clin. Endocrinol. Metab. 81, 246–252 (2012). female patients with Addison’s disease. Endocr. Connect.
1411–1415 (1996). 225. Mo, D. et al. Fracture risk in adult patients treated 8, 425–434 (2019).
206. Oelkers, W. & L’Age, M. Control of mineralocorticoid with growth hormone replacement therapy for growth 244. Andela, C. D. et al. Quality of life in patients
substitution in Addison’s disease by plasma renin hormone deficiency: a prospective observational with adrenal insufficiency correlates stronger with
measurement. Klin. Wochenschr. 54, 607–612 cohort study. Lancet Diabetes Endocrinol. 3, hydrocortisone dosage, than with long-​term systemic
(1976). 331–338 (2015). cortisol levels. Psychoneuroendocrinology 72, 80–86
207. Smith, S. J. et al. Evidence that patients with Addison’s 226. Mazziotti, G. et al. Management of endocrine (2016).
disease are undertreated with fludrocortisone. Lancet disease: risk of overtreatment in patients with 245. Lovas, K., Loge, J. H. & Husebye, E. S. Subjective
1, 11–14 (1984). adrenal insufficiency: current and emerging aspects. health status in Norwegian patients with Addison’s
208. Chortis, V. et al. Mitotane therapy in adrenocortical Eur. J. Endocrinol. 177, R231–R248 (2017). disease. Clin. Endocrinol. 56, 581–588 (2002).
cancer induces CYP3A4 and inhibits 5α-​reductase, 227. Falhammar, H., Filipsson Nystrom, H., Wedell, A., This is the first systematic assessment of subjective
explaining the need for personalized glucocorticoid Brismar, K. & Thoren, M. Bone mineral density, bone health status in a large cohort of patients with
and androgen replacement. J. Clin. Endocrinol. Metab. markers, and fractures in adult males with congenital primary adrenal insufficiency.
98, 161–171 (2013). adrenal hyperplasia. Eur. J. Endocrinol. 168, 246. Forss, M., Batcheller, G., Skrtic, S. & Johannsson, G.
209. Johannsson, G. et al. Low dose dehydroepiandrosterone 331–341 (2013). Current practice of glucocorticoid replacement therapy
affects behavior in hypopituitary androgen-​deficient 228. Guarnotta, V., Ciresi, A., Pillitteri, G. & Giordano, C. and patient-​perceived health outcomes in adrenal
women: a placebo-​controlled trial. J. Clin. Endocrinol. Improved insulin sensitivity and secretion in insufficiency – a worldwide patient survey. BMC
Metab. 87, 2046–2052 (2002). prediabetic patients with adrenal insufficiency on Endocr. Disord. 12, 8 (2012).
210. Lovas, K. et al. Replacement of dual-​release hydrocortisone treatment: a 36-month 247. Kluger, N. et al. Impaired health-​related quality of life
dehydroepiandrosterone in adrenal failure: no retrospective analysis. Clin. Endocrinol. 88, 665–672 in Addison’s disease–impact of replacement therapy,
benefit for subjective health status and sexuality (2018). comorbidities and socio-​economic factors.
in a 9-month, randomized, parallel group clinical trial. 229. Plat, L. et al. Metabolic effects of short-​term Clin. Endocrinol. 81, 511–518 (2014).
J. Clin. Endocrinol. Metab. 88, 1112–1118 (2003). elevations of plasma cortisol are more pronounced in 248. Tiemensma, J. et al. Psychological morbidity and
211. van Thiel, S. W. et al. Effects of the evening than in the morning. J. Clin. Endocrinol. impaired quality of life in patients with stable
dehydroepiandrostenedione, superimposed on Metab. 84, 3082–3092 (1999). treatment for primary adrenal insufficiency:
growth hormone substitution, on quality of life and This study showed that elevations of evening cross-​sectional study and review of the literature.
insulin-​like growth factor I in patients with secondary cortisol levels could contribute to alterations in Eur. J. Endocrinol. 171, 171–182 (2014).
adrenal insufficiency: a randomized, placebo-​ glucose tolerance, insulin sensitivity and insulin 249. Ragnarsson, O. et al. The relationship between
controlled, cross-​over trial. J. Clin. Endocrinol. Metab. secretion. glucocorticoid replacement and quality of life in
90, 3295–3303 (2005). 230. Filipsson, H., Monson, J. P., Koltowska-​Haggstrom, M., 2737 hypopituitary patients. Eur. J. Endocrinol. 171,
212. Gurnell, E. M. et al. Long-​term DHEA replacement Mattsson, A. & Johannsson, G. The impact of 571–579 (2014).
in primary adrenal insufficiency: a randomized, glucocorticoid replacement regimens on metabolic 250. Oksnes, M. et al. Quality of life in European patients
controlled trial. J. Clin. Endocrinol. Metab. 93, outcome and comorbidity in hypopituitary patients. with Addison’s disease: validity of the disease-​specific
400–409 (2008). J. Clin. Endocrinol. Metab. 91, 3954–3961 (2006). questionnaire AddiQoL. J. Clin. Endocrinol. Metab.
213. Coles, A. J. et al. Dehydroepiandrosterone Large Swedish cohort–control study revealing 97, 568–576 (2012).
replacement in patients with Addison’s disease has increased risk of ischaemic heart disease with 251. Burger-​Stritt, S., Pulzer, A. & Hahner, S. Quality
a bimodal effect on regulatory (CD4+CD25hi increasing steroid replacement dose in women with of life and life expectancy in patients with adrenal
and CD4+FoxP3+) T cells. Eur. J. Immunol. 35, autoimmune Addison disease. No increase in insufficiency: what is true and what is urban myth?
3694–3703 (2005). cerebrovascular disease risk was detected. Front. Horm. Res. 46, 171–183 (2016).
214. Dhatariya, K., Bigelow, M. L. & Nair, K. S. Effect 231. Skov, J., Sundstrom, A., Ludvigsson, J. F., Kampe, O. & 252. Bleicken, B. et al. Impaired subjective health status
of dehydroepiandrosterone replacement on insulin Bensing, S. Sex-​specific risk of cardiovascular disease in chronic adrenal insufficiency: impact of different
sensitivity and lipids in hypoadrenal women. Diabetes in autoimmune Addison disease–a population-​based glucocorticoid replacement regimens. Eur. J.
54, 765–769 (2005). cohort study. J. Clin. Endocrinol. Metab. 104, Endocrinol. 159, 811–817 (2008).
215. Srinivasan, M. et al. Effect of dehydroepiandrosterone 2031–2040 (2019). 253. Bleicken, B. et al. Influence of hydrocortisone dosage
replacement on lipoprotein profile in hypoadrenal 232. Falhammar, H. et al. Increased cardiovascular and scheme on health-​related quality of life in patients with
women. J. Clin. Endocrinol. Metab. 94, 761–764 metabolic morbidity in patients with 21-hydroxylase adrenal insufficiency. Clin. Endocrinol. 72, 297–304
(2009). deficiency: a Swedish population-​based national (2010).
216. Callies, F. et al. Dehydroepiandrosterone replacement cohort study. J. Clin. Endocrinol. Metab. 100, 254. Oksnes, M. et al. Continuous subcutaneous
in women with adrenal insufficiency: effects on body 3520–3528 (2015). hydrocortisone infusion versus oral hydrocortisone
composition, serum leptin, bone turnover, and 233. Quinkler, M. et al. Prednisolone is associated with a replacement for treatment of Addison’s disease:
exercise capacity. J. Clin. Endocrinol. Metab. 86, worse lipid profile than hydrocortisone in patients with a randomized clinical trial. J. Clin. Endocrinol. Metab.
1968–1972 (2001). adrenal insufficiency. Endocr. Connect. 6, 1–8 (2017). 99, 1665–1674 (2014).
217. Christiansen, J. J. et al. Very short term 234. Han, T. S. et al. Quality of life in adults with congenital Randomized clinical trial comparing continuous
dehydroepiandrosterone treatment in female adrenal hyperplasia relates to glucocorticoid treatment, subcutaneous hydrocortisone infusion with
adrenal failure: impact on carbohydrate, lipid and adiposity and insulin resistance: United Kingdom conventional oral hydrocortisone in 33 patients
protein metabolism. Eur. J. Endocrinol. 152, 77–85 Congenital adrenal Hyperplasia Adult Study Executive with Addison disease showing more improvement
(2005). (CaHASE). Eur. J. Endocrinol. 168, 887–893 (2013). in some quality of life parameters under more
218. Alkatib, A. A. et al. A systematic review and meta-​ 235. Han, T. S. et al. Glucocorticoid treatment regimen physiological cortisol profiles.
analysis of randomized placebo-​controlled trials of and health outcomes in adults with congenital adrenal 255. Nilsson, A. G. et al. Long-​term safety of once-​daily,
DHEA treatment effects on quality of life in women hyperplasia. Clin. Endocrinol. 78, 197–203 (2013). dual-​release hydrocortisone in patients with adrenal
with adrenal insufficiency. J. Clin. Endocrinol. Metab. 236. Behan, L. A. et al. Low-​dose hydrocortisone insufficiency: a phase 3b, open-​label, extension study.
94, 3676–3681 (2009). replacement is associated with improved arterial Eur. J. Endocrinol. 176, 715–725 (2017).
Meta-​analysis of randomized trials of DHEA in AI stiffness index and blood pressure dynamics in 256. Bancos, I. et al. Primary adrenal insufficiency is
showing overall small effects of DHEA on quality severely adrenocorticotrophin-​deficient hypopituitary associated with impaired natural killer cell function:
of life parameters and sexuality. male patients. Eur. J. Endocrinol. 174, 791–799 a potential link to increased mortality. Eur. J. Endocrinol.
219. Allolio, B., Arlt, W. & Hahner, S. DHEA: why, when, and (2016). 176, 471–480 (2017).
how much–DHEA replacement in adrenal insufficiency. 237. Werumeus Buning, J. et al. Effects of hydrocortisone Analysis of 42 patients with primary AI showing
Ann. Endocrinol. 68, 268–273 (2007). on the regulation of blood pressure: results from a normal neutrophil function but significantly
Comprehensive reflection of available evidence on randomized controlled trial. J. Clin. Endocrinol. Metab. decreased cytotoxicity of natural killer cells which
adrenal crisis and discussion of future directions. 101, 3691–3699 (2016). potentially compromises antiviral immune defence.


NATURE REvIEWS | DiSeASe PriMerS | Article citation ID: (2021) 7:19 23

0123456789();:
Primer

257. Smans, L. C., Souverein, P. C., Leufkens, H. G., 271. Mah, P. M. et al. Weight-​related dosing, timing and 286. Stirrat, L. I. et al. Transfer and metabolism of cortisol
Hoepelman, A. I. & Zelissen, P. M. Increased use monitoring hydrocortisone replacement therapy in by the isolated perfused human placenta. J. Clin.
of antimicrobial agents and hospital admission for patients with adrenal insufficiency. Clin. Endocrinol. Endocrinol. Metab. 103, 640–648 (2018).
infections in patients with primary adrenal insufficiency: 61, 367–375 (2004). 287. Coursin, D. B. & Wood, K. E. Corticosteroid
a cohort study. Eur. J. Endocrinol. 168, 609–614 272. Bratland, E. & Husebye, E. S. Cellular immunity and supplementation for adrenal insufficiency. JAMA 287,
(2013). immunopathology in autoimmune Addison’s disease. 236–240 (2002).
258. Tresoldi, A. S. et al. Increased infection risk in Mol. Cell Endocrinol. 336, 180–190 (2011). 288. Li, D. et al. Determinants of self-​reported health
Addison’s disease and congenital adrenal hyperplasia. 273. Rege, J., Turcu, A. F., Else, T., Auchus, R. J. & outcomes in adrenal insufficiency: a multi-​site survey
J. Clin. Endocrinol. Metab. 105, 418–429 (2020). Rainey, W. E. Steroid biomarkers in human adrenal study. J. Clin. Endocrinol. Metab. https://doi.org/
259. Gan, E. H. & Pearce, S. H. Management of endocrine disease. J. Steroid Biochem. Mol. Biol. 190, 273–280 10.1210/clinem/dgaa668 (2020).
disease: regenerative therapies in autoimmune (2019). 289. Fleming, L., Knafl, K., Knafl, G. & Van Riper, M.
Addison’s disease. Eur. J. Endocrinol. 176, 274. Mannstadt, M. et al. Hypoparathyroidism. Nat. Rev. Parental management of adrenal crisis in children with
R123–R135 (2017). Dis. Prim. 3, 17055 (2017). congenital adrenal hyperplasia. J. Spec. Pediatr. Nurs.
260. Balyura, M. et al. Transplantation of bovine 275. Lebbe, M. & Arlt, W. What is the best diagnostic and 22, e12190 (2017).
adrenocortical cells encapsulated in alginate. therapeutic management strategy for an Addison 290. Kienitz, T., Hahner, S., Burger-​Stritt, S. & Quinkler, M.
Proc. Natl Acad. Sci. USA 112, 2527–2532 (2015). patient during pregnancy? Clin. Endocrinol. 78, Therapeutic patient education for adrenal insufficiency
261. Gan, E. H. et al. Residual adrenal function in 497–502 (2013). under COVID-19 pandemic conditions. Exp. Clin.
autoimmune Addison’s disease: improvement after 276. Erichsen, M. M., Husebye, E. S., Michelsen, T. M., Endocrinol. Diabetes https://doi.org/10.1055/
tetracosactide (ACTH1-24) treatment. J. Clin. Dahl, A. A. & Lovas, K. Sexuality and fertility in women a-1217-7208 (2020).
Endocrinol. Metab. 99, 111–118 (2014). with Addison’s disease. J. Clin. Endocrinol. Metab. 95,
262. Pearce, S. H. et al. Adrenal steroidogenesis after 4354–4360 (2010). Acknowledgements
B lymphocyte depletion therapy in new-​onset 277. Remde, H., Zopf, K., Schwander, J. & Quinkler, M. S.H. is supported by the Deutsche Forschungsgemeinschaft
Addison’s disease. J. Clin. Endocrinol. Metab. 97, Fertility and pregnancy in primary adrenal insufficiency (DFG) (within the CRC/Transregio 205/1 “The Adrenal: Central
E1927–E1932 (2012). in Germany. Horm. Metab. Res. 48, 306–311 (2016). Relay in Health and Disease” and HA 6931/3-1). W.A. is sup-
263. Napier, C. et al. Residual adrenal function in 278. Bjornsdottir, S. et al. Addison’s disease in women is a ported by the Medical Research Council UK (programme
autoimmune Addison’s disease – effect of dual therapy risk factor for an adverse pregnancy outcome. J. Clin. grant G0900567). I.B. is supported by the National Institute
with rituximab and depot tetracosactide. J. Clin. Endocrinol. Metab. 95, 5249–5257 (2010). of Diabetes and Digestive and Kidney Diseases (NIDDK) of
Endocrinol. Metab. 105, e1250–e1259 (2020). 279. Schneiderman, M., Czuzoj-​Shulman, N., Spence, A. R. the National Institutes of Health (NIH) USA under award
264. Hahner, S. et al. Timelines in the management & Abenhaim, H. A. Maternal and neonatal outcomes K23DK121888. The views expressed are those of the
of adrenal crisis – targets, limits and reality. of pregnancies in women with Addison’s disease: a author(s) and not necessarily those of the National Institutes
Clin. Endocrinol. 82, 497–502 (2015). population-​based cohort study on 7.7 million births. of Health, USA. E.S.H. is supported by K.G. Jebsen Center of
265. Burger-​Stritt, S. et al. Management of adrenal BJOG 124, 1772–1779 (2017). Autoimmune Disorders, The Novonordisk Foundation, The
emergencies in educated patients with adrenal 280. Bothou, C. et al. Current management and outcome Norwegian Research Council. The authors thank S. Aslaksen
insufficiency–a prospective study. Clin. Endocrinol. of pregnancies in women with adrenal insufficiency: for her input in creating the figure on mechanisms underlying
89, 22–29 (2018). experience from a multicenter survey. J. Clin. Addison disease.
266. Repping-​Wuts, H. J., Stikkelbroeck, N. M., Noordzij, A., Endocrinol. Metab. 105, e2853–e2863 (2020).
Kerstens, M. & Hermus, A. R. A glucocorticoid Large retrospective multicentre analysis of the Author contributions
education group meeting: an effective strategy for course of pregnancy in women with adrenal Introduction (S.H. and M.Q.); Epidemiology (I.B. and E.S.H.);
improving self-​management to prevent adrenal crisis. insufficiency. Mechanisms/pathophysiology (S.H., E.S.H., R.J.R. and M.Q.);
Eur. J. Endocrinol. 169, 17–22 (2013). 281. Hirschberg, A. L. et al. Reproductive and perinatal Diagnosis, screening and prevention (W.A. and M.Q.);
267. Burger-​Stritt, S. et al. Standardised patient education outcomes in women with congenital adrenal Management (W.A., S.H., M.Q., R.J.R., S.B.-​S. and D.J.T.);
in adrenal insufficiency – a prospective multi-​centre hyperplasia: a population-​based cohort study. Quality of life (S.H.); Outlook (S.H., M.Q. and R.J.R.);
evaluation. Eur. J. Endocrinol. 183, 119–127 (2020). J. Clin. Endocrinol. Metab. 106, e957–e965 (2021). Overview of Primer (S.H.).
This longitudinal, prospective, questionnaire-​based, 282. Quinkler, M. et al. Agonistic and antagonistic
multicentre study included 526 patients with AI properties of progesterone metabolites at the human Competing interests
and proved that group education of patients with mineralocorticoid receptor. Eur. J. Endocrinol. 146, R.J.R. is a Director of Diurnal group PLC. W.A. served as con-
chronic AI represents a helpful tool for the 789–799 (2002). sultant and clinical investigator for Diurnal Ltd. All other
guidance of patients, their self-​assurance and their 283. Quinkler, M., Meyer, B., Oelkers, W. & Diederich, S. authors declare no competing interests.
knowledge on prevention of adrenal crises. Renal inactivation, mineralocorticoid generation,
268. Quinkler, M., Hahner, S., Johannsson, G. & and 11β-​hydroxysteroid dehydrogenase inhibition Peer review information
Stewart, P. M. Saving lives of patients with adrenal ameliorate the antimineralocorticoid effect of Nature Reviews Disease Primers thanks L. Chan,
insufficiency: a pan-​European initiative? Clin. Endocrinol. progesterone in vivo. J. Clin. Endocrinol. Metab. 88, G. Johannsson, L. Nieman and the other, anonymous,
80, 319–321 (2014). 3767–3772 (2003). reviewer(s) for their contribution to the peer review of this work.
269. Bornstein, S. R., Bornstein, T. D. & Andoniadou, C. L. 284. Cosimo, C. & Franco, C. Addison’s disease and
Novel medications inducing adrenal insufficiency. pregnancy: case report. J. Prenat. Med. 3, 53–54 Publisher’s note
Nat. Rev. Endocrinol. 15, 561–562 (2019). (2009). Springer Nature remains neutral with regard to jurisdictional
270. Nicolaides, N. C., Chrousos, G. P. & Charmandari, E. 285. Oelkers, W. K. Effects of estrogens and progestogens claims in published maps and institutional affiliations.
in Endotext (eds Feingold, K. R. et al) (MDText.com, Inc, on the renin-​aldosterone system and blood pressure.
2000). Steroids 61, 166–171 (1996). © Springer Nature Limited 2021

24 | Article citation ID: (2021) 7:19 www.nature.com/nrdp

0123456789();:

You might also like