Exp Clin Endocrinol Diabetes 2019; 127(02/03): 165-175
DOI: 10.1055/a-0804-2715
Review
© Georg Thieme Verlag KG Stuttgart · New York

An Update on Addison’s Disease

Andreas Barthel
1   Medicover, Bochum, Germany
2   Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
,
Georg Benker
1   Medicover, Bochum, Germany
,
Kai Berens
1   Medicover, Bochum, Germany
,
Sven Diederich
3   Medicover, Berlin-Mitte, Germany
,
Burkhard Manfras
4   Medicover, Ulm and Neu-Ulm, Germany
,
Matthias Gruber
2   Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
,
Waldemar Kanczkowski
2   Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
,
Greg Kline
5   University of Calgary, Calgary, AB,Canada
,
Virginia Kamvissi-Lorenz
2   Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
8   Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
,
Stefanie Hahner
6   Department of Medicine I, Würzburg University Hospital, Würzburg, Germany
,
Felix Beuschlein
7   Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, University Hospital, Zürich, Switzerland
,
Ana Brennand
8   Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
,
Bernhard O. Boehm
9   Lee Kong Chian School of Medicine, NTU Nanyang Technological University, Singapore, Singapore
,
David J. Torpy
10   Endocrine and Metabolic Unit, Royal Adelaide Hospital, University of Adelaide, Adelaide SA, Australia
,
Stefan R. Bornstein
2   Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
7   Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, University Hospital, Zürich, Switzerland
8   Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
9   Lee Kong Chian School of Medicine, NTU Nanyang Technological University, Singapore, Singapore
› Author Affiliations
Further Information

Correspondence

Andreas Barthel
Medicover Bochum
Universitätsstrasse 48
44789 Bochum
Germany   

Publication History

received   05 July 2018
revised   21 November 2018

accepted   21 November 2018

Publication Date:
18 December 2018 (online)

 

Abstract

Addison’s disease – the traditional term for primary adrenal insufficiency (PAI) – is defined as the clinical manifestation of chronic glucocorticoid- and/or mineralocorticoid deficiency due to failure of the adrenal cortex which may result in an adrenal crisis with potentially life-threatening consequences. Even though efficient and safe pharmaceutical preparations for the substitution of endogenous gluco- and mineralocorticoids are established in therapy, the mortality in patients with PAI is still increased and the health-related quality of life (HRQoL) is often reduced.

PAI is a rare disease but recent data report an increasing prevalence. In addition to the common “classical” causes of PAI like autoimmune, infectious, neoplastic and genetic disorders, other iatrogenic conditions – mostly pharmacological side effects (e. g., adrenal haemorrhage associated with anticoagulants, drugs affecting glucocorticoid synthesis, action or metabolism and some of the novel anti-cancer checkpoint inhibitors) are contributing factors to this phenomenon.

Due to the rarity of the disease and often non-specific symptoms at least in the early stages, PAI is frequently not considered resulting in a delayed diagnosis. Successful therapy is mainly based on adequate patient education as a cornerstone in the prevention and management of adrenal crisis. A focus of current research is in the development of pharmacokinetically optimized glucocorticoid preparations as well as regenerative therapies.


#

Introduction

The clinical picture of primary adrenal insufficiency (PAI) is based on the chronic deficiency of glucocorticoids and/or mineralocorticoids due to failure of the adrenal cortex to produce these hormones in sufficient amounts. Since they are essential regulators of water- and electrolyte homeostasis as well as energy balance, PAI is a very severe disease and may result in acute and potentially life-threatening adrenal crisis [1]. The dramatic consequences of adrenal failure with weakness, weight loss, anorexia, orthostatic hypotension due to dehydration, salt craving, hyperpigmentation, musculoskeletal and abdominal pain, nausea, vomiting, and ultimately the lethal outcome have first been recognized by Thomas Addison, a british surgeon – therefore, PAI is also commonly named as “Addison’s disease” [2].

The prognosis of the disease dramatically improved with the availability of steroid hormones for treatment and the development of feasible diagnostic tests. After successful isolation and characterization of cortisol and cortisone [3] [4] [5] [6] and the establishment of techniques to synthetize glucocorticoid hormones [7], the treatment of adrenal insufficiency as well as the diagnosis of the disease was substantially promoted mainly by the clinical work of Wilder in the 1930’s and then Thorn and Forsham in the 1940s and 1950s [8] [9] [10].

Here, we will provide an overview on current aspects in the diagnosis and treatment of Addison’s disease. Subsequently, as a synonym of Addison’s disease, we will exclusively use the term PAI for reasons of stringency and convenience.


#

Epidemiology and Etiology of Primary Adrenal Insufficiency

PAI is a rare disease with a current prevalence in our Western societies of about 100–140 cases per million [11]. However, the reported number has substantially increased over time from 40–70 cases per million in europe in the 1960s [12]. Interestingly, recent data suggest a continuation of this trend with a further increasing prevalence of PAI particularly in women [13]. In addition to a real increase of the prevalence the possibility should be considered that this phenomenon may be related to a general underestimation of the prevalence of PAI in the past and improvements of diagnostic and health care conditions over time [14]. It is also possible that etiologic changes may contribute to this observed effect. As described by Addison, based on 11 patients, the etiology of adrenal destruction in PAI was originally more than 50% tuberculosis, 30% neoplastic/metastatic and about 10% haemorrhagic at that time [2]. Today, in western societies, 80% of PAI is caused by autoimmune adrenalitis followed by tuberculosis or other infectious diseases (e. g. HIV/AIDS, CMV, candidiasis, histoplasmosis, syphilis and others) and malignant diseases (e. g. lung, breast, colon cancer and others) in about 10% of cases. The remaining causes include (bilateral) adrenalectomy (e. g. for Cushing’s syndrome or adrenal tumours), genetic diseases (e. g. congenital adrenal hyperplasia (CAH), adrenal hypoplasia congenita, Adrenoleukodystrophy in males) and adrenal haemorrhage (e. g. Waterhouse-Friderichsen-Syndrome in sepsis) [15] [16] [17].

PAI due to autoimmune adrenalitis may exist in isolation, but more than 50% of autoimmune adrenalitis occurs in combination with other autoimmune disorders as part of autoimmune polyglandular syndromes (APS). These syndromes sometimes have a known specific genetic background (e. g. AIRE gene mutations in APS type 1) but all forms of autoimmune adrenalitis show some association with specific gene variants of the major histocompatibility complex (e. g. HLA-DR3) or genes encoding for proteins involved in mechanisms of immunological regulation (e. g. CTLA-4). This emphasizes the central role of T-cell and cellular immunity in the pathophysiology of Addison’s disease [18] [19] [20] [21].

Due to the increasing age in our population with chronic illness often requiring complex medical treatment and polypharmacy, pharmacological adverse effects (e. g., adrenal haemorrhage associated with anticoagulants, drugs affecting glucocorticoid synthesis, action or metabolism like specific antimycotics and other compounds) are becoming more and more relevant as potential predisposing factors contributing to the manifestation of PAI [16]. This is distinct from the even more common scenario of secondary adrenal insufficiency (i. e., hypothalamic-pituitary suppression) of chronic glucocorticoid use for inflammatory conditions. Further, it is of increasing importance that novel drugs and therapeutic regimens may induce the manifestation of immune-mediated endocrine disorders including PAI. For example, recent pharmacovigilance data indicate an association of PAI with the use of different immune checkpoint inhibitors (ICIs), a novel class of drugs applied for the treatment of a variety of malignant tumours [22] [23]. Interestingly, it has been reported that PAI may develop as a late adverse event even more than a year after discontinuing treatment with pembrolizumab, a prototypic ICI [24]. Therefore, it is essential to carefully monitor patients being treated with drugs associated with an increased risk for the development of PAI and other immune-mediated endocrinopathies.

Unfortunately, symptoms and signs of PAI are non-specific in the early stages, therefore making the clinical diagnosis of PAI very difficult. Further, due to the fact that PAI is a rare disease with a reported incidence of 4–6 cases per million per year, the diagnosis is frequently not considered and therefore delayed [25] [26]. As a consequence, the first presentation of many patients with PAI is an acute and potentially life-threatening adrenal crisis in the emergency department.


#

Diagnosis of Primary Adrenal Insufficiency

PAI should be suspected in all acutely or chronically ill patients with general fatigue or severe weakness and unexplained dehydration, hypotension, weight loss, fever, abdominal pain and hyperpigmentation. Hyponatraemia, hyperkalaemia and - especially in children - hypoglycaemia are key lab characteristics of the disease reflecting the deficiency of gluco- and mineralocorticoids [27]. The diagnostic threshold should be even lower when patients have features of other autoimmune diseases like vitiligo, type-1-diabetes or autoimmune gastritis/Vitamin B12-deficiency. Additionally, in acutely ill patients with chronic infectious diseases – in particular HIV, CMV or tuberculosis – or in patients receiving drugs interfering with cortisol synthesis, action or metabolism - in particular anticonvulsants (carbamazepine, phenytoin), anti-fungals (ketoconazole), anti-cancer drugs (abiraterone, mitotane, ICIs) or specific over-the-counter medicines (e. g. St John’s Wort) - the index of suspicion should be high [16] [27].

In patients with symptoms and signs indicative of PAI, further diagnostic testing is required. Initially, a random blood sample for the simultaneous determination of ACTH and cortisol before glucocorticoid treatment is essential for the detection of glucocorticoid deficiency which is usually the leading clinical characteristic. The typical constellation in PAI is a low cortisol serum concentration in combination with an elevated ACTH. As a generally accepted clinical rule, PAI is regarded as very likely if cortisol is<5 µg/dl (138 nmol/L) with a concomitant ACTH increased more than 2-fold over the upper limit of the normal range [27] [28]. Mineralocorticoid deficiency is typically reflected by decreased sodium and elevated potassium serum concentrations. To verify mineralocorticoid deficiency, determination of plasma renin and aldosterone may be useful if there is no hyperkalemia present. The combination of an elevated plasma renin together with a normal or low serum aldosterone concentration is suggestive of mineralocorticoid deficiency [27] [29] [30].

Dynamic testing of the adrenocortical function with the corticotropin stimulation test – also termed as cosyntropin test, ACTH test or Synacthen test - is currently the best established and validated method for the diagnosis or exclusion of PAI [31]. If the diagnosis is not already obvious, this test may be performed as a confirmatory test if available and the patient is stable enough [27]. In this test, adults usually receive an injection of 250 µg intravenous (or alternatively intramuscular; in children<2 years 125 µg) of tetracosactide, a peptide representing the sequence of the first 24 (out of a total of 39) amino acids of ACTH and cortisol is determined immediately before and then 30 and 60 min after injection. As a common cut-off for the diagnosis of (primary) adrenal insufficiency, a peak cortisol concentration of less than 500 nmol/l (18 µg/dl) after stimulation is historically widely accepted [27]. However, cut-off levels for cortisol and other adrenal steroids after stimulation with cosyntropin may differ significantly based on the method used for detection (Liquid chromatography tandem mass spectrometry (LC-MS/MS) vs. immunoassay) [32]. Clinicians are strongly urged to consult with the local clinical chemist to verify the assay-specific definitions of normal cortisol responses to stimulation.

In contrast to the “high-dose” (250 µg) corticotropin test, the so-called “low-dose” corticotropin stimulation test utilizes 1 µg tetracosactide as a variation [33] [34]. The advantages and disadvantages of both tests in the diagnosis of adrenal insufficiency have been extensively discussed [35] [36]. Since both tests give comparable results for the serum cortisol concentration at least 30 min after stimulation and based on the fact that the “high-dose” (250 µg) corticotropin test is more comprehensively validated compared to the “low-dose” (1 µg) corticotropin test, it is suggested to perform the “low-dose” (1 µg) corticotropin test in the diagnosis of PAI only when the substance is in short supply [27] [34] [35] [37] [38].

Although a clear cut-off (500 nmol(l (18 µg/dl), as determined with common immunoassays) for the stimulated serum cortisol concentration has been defined to diagnose or exclude (primary) adrenal insufficiency, interpretation of the cosyntropin-test may be complicated by several factors. For example, the level of the binding globulin for cortisol, CBG, is increased by estrogens (e. g. in pregnancy or in oral contraceptives) – thereby resulting in increased measured cortisol levels, while patients with several disorders (e. g. liver disease or nephrotic syndrome) may have lower levels of measured cortisol due to decreased levels of CBG [39] [40] [41]. Concomitant or surreptitious use of synthetic glucocorticoids (administered orally or topically or inhaled) may also confound the results depending on the specificity of the cortisol assay in use. Therefore, especially in medically complex patients and pregnant women, diagnosis and interpretation of test results requires extensive experience in clinical endocrinology.

Once PAI has been confirmed at the biochemical level, further diagnostic testing (e. g. 21-OH antibody, 17-OH-progesterone, CT adrenals, very long chain fatty acids (VLCFA) in preadolescent boys) to determine the etiology of the disease (autoimmuneadrenalitis, CAH, infections, infiltrative disease, adrenoleukodystrophy…) is suggested, since this may have prognostic and therapeutic implications [27].


#

Treatment and Prevention of Primary Adrenal Insufficiency

Hydrocortisone is the drug of choice for the replacement of glucocorticoids. In adults, the usual dose is 15–25 mg/d given orally in 2 or 3 doses with the highest dose in the morning to mimic circadian rhythm [27]. The first dose should be taken with awakening early in the morning whereas the last dose should be taken at the latest 4–6 h before sleep to avoid insomnia and nocturnal insulin resistance with potential adverse metabolic consequences [42]. As an alternative to hydrocortisone, 3–5 mg/d prednisolone as a single dose or in 2 divided oral doses is suggested. This may be favourable in patients requiring a simplified therapeutic regimen due to specific individual circumstances – e. g. to improve comfort and compliance [43]. Dexamethasone should not be used due to its non-physiological pharmacokinetic properties (long half-life) and the resulting risks for adverse metabolic effects such as dyslipidaemia, hyperglycaemia and diabetes [44] [45]. In children, only hydrocortisone should be used for replacement therapy because its short half-life allows for best control; a typical starting dose is about 8 mg/m2 divided in 3 doses. Also, in pregnancy, hydrocortisone is the preferred drug for glucocorticoid replacement. In the last trimester of pregnancy, cortisol levels and free cortisol rise significantly, therefore it is suggested to increase the hydrocortisone dose by about 30% in the last trimester in pregnant patients with PAI [27] [46] [47]. Further, since high levels of progesterone counteract mineralocorticoid action, an increase of the fludrocortisone dose based on blood pressure and electrolyte status is frequently required especially in the third trimester.

Monitoring and management of glucocorticoid replacement therapy is primarily based on clinical parameters such as body weight, oedema, blood pressure, cushingoid changes as well as mental and physical performance. It is of prime importance to prevent and detect inadequate dosing and metabolic complications of long-term glucocorticoid treatment [48] [49]. In children, this includes monitoring of general development and growth velocity every 3–4 months. Laboratory measures of plasma ACTH and random serum cortisol are of little value. For example, patients with adequate hydrocortisone replacement frequently show increased ACTH levels because the short half life of orally administered hydrocortisone rarely fully mimics physiologic secretion to the point of normalization of all ACTH measures. In addition, the glucocorticoid dose needs to be adjusted dynamically according to specific stressful circumstances. For example, in acute febrile illness with temperatures exceeding 38°C the oral hydrocortisone dose should be increased 2–3 fold. For major surgery or in adrenal crisis, hydrocortisone should be given intravenously at an initial dose of 100 mg in adults (50 mg/m2 in children) followed by a continuous infusion of 200 mg hydrocortisone (50–100 mg/m2 in children) over 24 h together with intravenous fluid replacement to counteract dehydration [27] [50]. Due to volume depletion (salt wasting), hypotension and circulatory failure, acute adrenal crisis is a life-threatening condition. If adrenal crisis is suspected, a random cortisol and ACTH may be measured but therapy should then be instituted without the delay of awaiting results.

In mineralocorticoid deficiency, fludrocortisone is the drug of choice to stabilize water- and electrolyte balance. A common starting dose is 50–100 µg/d in children and adults with PAI, typically given orally once in the morning. Patients should be encouraged to include salt in their diet. Monitoring and management of mineralocorticoid replacement therapy is mainly based on clinical parameters like salt craving, oedema, blood pressure in combination with lab parameters like normal sodium and potassium concentrations in the serum; a plasma renin in the upper reference range is regarded as a helpful indicator for fine-tuning [29] [51] [52] [53] but should not supersede clinical assessment. Stress adjustment of mineralocorticoid replacement is not required.

In addition to the replacement with gluco- and mineralocorticoids which are vital, androgen substitution with dehydroepiandrosterone (DHEA) is frequently discussed in women with PAI. Based on current data, DHEA replacement may be considered in women receiving optimal gluco- and mineralocorticoid therapy that are still suffering from symptoms like low libido and depressive symptoms [27] [54] [55], particularly if post-menopausal or having concomitant primary ovarian insufficiency with loss of ovarian androgens.

Important and frequently discussed questions concern health related quality of life (HRQoL), co-morbidities and prognostic factors in patients with PAI. In this group of patients, self-reported quality of life has been documented to be lower as compared to the general population [56]. In addition, patients with PAI reportedly have a higher incidence of affective disorders such as depression and anxiety as well as a higher odds ratio for metabolic disorders like diabetes mellitus as compared to controls. This is reflected by more frequent hospital stays and substantially higher health care expenditures in these patients [49] [57] [58] [59]. Further, increased mortality has been reported in patients with PAI; in particular acute infections carry a risk of fatal adrenal crisis [11] [56] [60] [61]. However, other factors such as the impact of potentially adverse cardio-vascular risk profiles related to excessive glucocorticoid replacement regimes have been discussed in this context [45] [59].

In patients with known PAI, preventing adrenal crisis is of central importance. Currently, the reported incidence is about 6-8/100 patients per year with a mortality rate of 0.5/100 patient years [62] [63]. Therefore, patient education with regard to the management of specific situations requiring an increased glucocorticoid dose (e. g. flu-like febrile infections) including the use of techniques for parenteral administration of emergency glucocorticoids, as well as recognition of symptoms and signs signaling an emerging adrenal crisis is essential [27] [50]. Further, it is recommended that all patients with PAI should carry a medical alert notification or even better, a steroid emergency card to inform physicians and other medical personnel [27]. A standardised European emergency card for patients with PAI has been endorsed by the European Society of Endocrinology (ESE) and is sponsored by a variety of institutions [64].


#

Novel Aspects And Current Developments For the Treatment of Primary Adrenal Insufficiency

Steroid substitution in patients with PAI is effective but glucocorticoid replacement still has clear pharmacological limitations and normal physiology can not be completely mimicked by using oral hydrocortisone. In order to improve the pharmacokinetic properties and to better provide circadian cortisol concentrations with a morning peak, a dual-release preparation of hydrocortisone that needs to be given as a single oral dose in the morning has been developed [65]. Early clinical data are promising with regard to drug efficacy and safety [66]. In addition to more patient satisfaction due to the single-dose application, favourable effects with regard to body weight, blood pressure and glycaemic control have been described as compared to standard therapy [67] [68]. Other modified-release hydrocortisone formulations are currently in clinical development [69]. A challenging problem is glucocorticoid replacement in children; in order to help improve dosage and oral application, hydrocortisone capsules have been developed and successfully tested [70]. Restoration of normal circadian cortisol concentrations in patients with PAI compared to oral regimens may be accomplished by continuous subcutaneous cortisol infusion using insulin pumps. Although this approach is complex and therefore only applicable in selected individual patients, initial reports appear very promising [71] [72] [73].

Other experimental approaches in very early stages have been described including immunosuppression in patients with early forms of autoimmune adrenalitis, gene therapy for monogenic forms of PAI, transplantation and cell replacement as well as reprogramming techniques to induce a steroidogenic phenotype from cells of different origin [74] [75] [76] [77] [78].

In summary, PAI is a critically important and potentially life threatening disease but with modern therapy and education, affected patients can now return to a highly functional status. Further improvements are still needed to improve both quality of life and unravel potential disease mechanisms that may be exploited for possible prevention or cure of this serious disease.


#
#

Conflict of Interest

No conflict of interest has been declared by the authors.

  • References

  • 1 Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet. 2014; 383: 2152-2167
  • 2 Addison T. On the Constitutional and Local Effects of Disease of the Supra-renal Capsules. London, UK: Samuel Highley; 1855
  • 3 Wintersteiner OP, Pfiffner J. Chemical studies on the adrenal cortex: II. J Biol Chem 1935; 111: 599-612
  • 4 Kendall EC. A chemical and physiological investigation of the suprarenal cortex. Symp Quant Biol 1937; 5: 299
  • 5 de Fremery P, Laquer E, Reichstein T. et al. Corticosterone: A crystalloid compound with biological activity of the adrenal-cortical hormone. Nature 1937; 139: 26-27
  • 6 Grollman A. Physiological and chemical studies on the adrenal cortical hormone. Symp Quant Biol 1937; 5: 313
  • 7 Sarett L. The synthesis of hydrocortisone from desoxy- cholic acid. J Biol Chem 1946; 162: 601
  • 8 Cutler HH, Power MH, Wilder RM. Concentrations of chloride, sodium and potassium in urine and blood: Their diagnostic significance in adrenal insufficiency. JAMA 1938; 111: 117-122
  • 9 Forsham PH, Thorn GW, Recant L. et al. Clinical experience with the use of the ACTH test for adrenal cortical function. J Clin Invest 1948; 27: 534
  • 10 Thorn GW, Forsham PH, Frawley TF. et al. Clinical experience with the use of cortisone acetate pellets in the treatment of adrenal cortical insufficiency. J Clin Endocrinol Metab 1950; 10: 833-834
  • 11 Erichsen MM, Løvås K, Fougner KJ. et al. Normal overall mortality rate in Addison’s disease, but young patients are at risk of premature death. Eur J Endocrinol 2009; 160: 233-237
  • 12 Mason AS, Meade TW, Lee JA. et al. Epidemiological and clinical picture of Addison’s disease. Lancet 1968; 2: 744-747
  • 13 Meyer G, Neumann K, Badenhoop K. et al. Increasing prevalence of Addison’s disease in German females: Health insurance data 2008–2012. Eur J Endocrinol 2014; 170: 367-373
  • 14 Laureti S, Vecchi L, Santeusanio F. et al. Is the prevalence of Addison’s disease underestimated?. J Clin Endocrinol Metab 1999; 84: 1762
  • 15 Arlt W, Allolio B. Adrenal insufficiency. Lancet 2003; 361: 1881-1893
  • 16 Bornstein SR. Predisposing factors for adrenal insufficiency. N Engl J Med 2009; 360: 2328-2339
  • 17 Betterle C, Morlin L. Autoimmune Addison’s disease. Endocr Dev 2011; 20: 161-172
  • 18 Mitchell AL, Pearce SH. Autoimmune Addison disease: Pathophysiology and genetic complexity. Nat Rev Endocrinol 2012; 8: 306-316
  • 19 Husebye ES, Lovas K. Immunology of Addison’s disease and premature ovarian failure. Endocrinol Metab Clin North Am 2009; 38: 389-405
  • 20 Husebye E, Lovas K. Pathogenesis of primary adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 2009; 23: 147-157
  • 21 Husebye ES, Anderson MS, Kämpe O. Autoimmune polyendocrine syndromes. N Engl J Med 2018; 378: 1132-1141
  • 22 Konda B, Nabhan F, Shah MH. Endocrine dysfunction following immune checkpoint inhibitor therapy. Curr Opin Endocrinol Diabetes Obes 2017; 24: 337-347
  • 23 Barroso-Sousa R, Barry WT, Garrido-Castro AC. et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: A systematic review and meta-analysis. JAMA Oncol 2018; 4: 173-182
  • 24 Boudjemaa A, Rousseau-Bussac G, Monnet I. Late-Onset adrenal insufficiency more than 1 year after stopping pembrolizumab. J Thorac Oncol 2018; 13: e39-e40
  • 25 Lovas K, Husebye ES. High prevalence and increasing incidence of Addison’s disease in western Norway. Clin Endocrinol (Oxf) 2002; 56: 787-791
  • 26 Bleicken B, Hahner S, Ventz M. et al. Delayed diagnosis of adrenal insufficiency is common: A cross-sectional study in 216 patients. Am J Med Sci 2010; 339: 525-531
  • 27 Bornstein SR, Allolio B, Arlt W. et al. Diagnosis and treatment of primary adrenal insufficiency: An endocrine society clinical practice guideline. J Clin Endocrinol Metab 2016; 101: 364-389
  • 28 Kazlauskaite R, Evans AT, Villabona CV. et al. Corticotropin tests for hypothalamic-pituitary-adrenal insufficiency: A metaanalysis. J Clin Endocrinol Metab 2008; 93: 4245-4253
  • 29 Oelkers W, Diederich S, Bähr V. Diagnosis and therapy surveillance in Addison’s disease: Rapid adrenocorticotropin(ACTH) test and measurement of plasma ACTH, renin activity, and aldosterone. J Clin Endocrinol Metab 1992; 75: 259-264
  • 30 Rehan M, Raizman JE, Cavalier E. et al. Laboratory challenges in primary aldosteronism screening and diagnosis. Clin Biochem. 2015; 48: 377-387
  • 31 Pofi R, Feliciano C, Sbardella E. et al. The short Synacthen (corticotropin) test can be used to predict recovery of hypothalamo-pituitary-adrenal axis function. J Clin Endocrinol Metab 2018; 103: 3050-3059
  • 32 Ueland GÅ, Methlie P, Øksnes M. Et al. The short cosyntropin test revisited: New normal reference range using LC-MS/MS. J Clin Endocrinol Metab 2018; 103: 1696-1703
  • 33 Mayenknecht J, Diederich S, Bähr V. et al. Comparison of low and high dose corticotropin stimulation tests in patients with pituitary disease. J Clin Endocrinol Metab 1998; 83: 1558-1562
  • 34 Pura M, Kreze Jr A, Kentos P. et al. The low-dose (1 microg) cosyntropin test (LDT) for primary adrenocortical insufficiency: Defining the normal cortisol response and report on first patients with Addison disease confirmed with LDT. Exp Clin Endocrinol Diabetes 2010; 118: 151-157
  • 35 Magnotti M, Shimshi M. Diagnosing adrenal insufficiency: Which test is best–the 1-microg or the 250-microg cosyntropin stimulation test?. Endocr Pract. 2008; 14: 233-238
  • 36 Oelkers W. The role of high- and low-dose corticotropin tests in the diagnosis of secondary adrenal insufficiency. Eur J Endocrinol 1998; 139: 567-570
  • 37 Dekkers OM, Timmermans JM, Smit JW. et al. Comparison of the cortisol responses to testing with two doses of ACTH in patients with suspected adrenal insufficiency. Eur J Endocrinol 2011; 164: 83-87
  • 38 Lindholm J, Kehlet H. Re-evaluation of the clinical value of the 30 min ACTH test in assessing the hypothalamic-pituitary-adrenocortical function. Clin Endocrinol (Oxf) 1987; 26: 53-59
  • 39 Klose M, Lange M, Rasmussen AK. et al. Factors influencing the adrenocorticotropin test: role of contemporary cortisol assays, body composition, and oral contraceptive agents. J Clin Endocrinol Metab 2007; 92: 1326-1333
  • 40 Vincent RP, Etogo-Asse FE, Dew T. et al. Serum total cortisol and free cortisol index give different information regarding the hypothalamus-pituitary-adrenal axis reserve in patients with liver impairment. Ann Clin Biochem 2009; 46: 505-507
  • 41 Dhillo WS, Kong WM, Le Roux CW. et al. Cortisol-binding globulin is important in the interpretation of dynamic tests of the hypothalamic pituitary–adrenal axis. Eur J Endocrinol 2002; 146: 231-235
  • 42 Plat L, Leproult R, L’Hermite-Baleriaux M. et al. Metabolic effects of short-term elevations of plasma cortisol are more pronounced in the evening than in the morning. J Clin Endocrinol Metab 1999; 84: 3082-3092
  • 43 Forss M, Batcheller G, Skrtic S. et al. Current practice of glucocorticoid replacement therapy and patient-perceived health outcomes in adrenal insufficiency - A worldwide patient survey. BMC Endocr Disord 2012; 12: 8
  • 44 Fadeev VV, Gitel EP, Mel’nichenko GA. The diurnal rhythm of adrenocorticotropic hormone secretion in the assessment of the adequacy of replacement therapy in primary chronic adrenal failure. Neurosci Behav Physiol 2001; 31: 237-242
  • 45 Filipsson H, Monson JP, Koltowska-Häggström M. et al. The impact of glucocorticoid replacement regimens on metabolic outcome and comorbidity in hypopituitary patients. J Clin Endocrinol Metab 2006; 91: 3954-3961
  • 46 Lebbe M, Arlt W. What is the best diagnostic and therapeutic management strategy for an Addison patient during pregnancy?. Clin Endocrinol (Oxf) 2013; 78: 497-502
  • 47 Jung C, Ho JT, Torpy DJ. et al. A longitudinal study of plasma and urinary cortisol in pregnancy and postpartum. J Clin Endocrinol Metab 2011; 96: 1533-1540
  • 48 Hardy RS, Zhou H, Seibel MJ. et al. Glucocorticoids and bone: Consequences of endogenous and exogenous excess and replacement therapy. Endocr Rev 2018; 39: 519-548
  • 49 Ueland GA, Husebye ES. Metabolic Complications in Adrenal Insufficiency. Front Horm Res 2018; 49: 104-113
  • 50 Allolio B. Extensive expertise in endocrinology. Adrenal crisis. Eur J Endocrinol 2015; 172: R115-R124
  • 51 Oelkers W, L’Age M. Control of mineralocorticoid substitution in Addison’s disease by plasma renin measurement. Klin Wochenschr. 1976; 54: 607-612
  • 52 Flad TM, Conway JD, Cunningham SK. et al. The role of plasma renin activity in evaluating the adequacy of mineralocorticoid replacement in primary adrenalin sufficiency. ClinEndocrinol (Oxf) 1996; 45: 529-534
  • 53 Quinkler M, Oelkers W, Remde H. et al. Mineralocorticoid substitution and monitoring in primary adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 2015; 29: 17-24
  • 54 Lang K, Burger-Stritt S, Hahner S. Is DHEA replacement beneficial in chronic adrenal failure?. Best Pract Res Clin Endocrinol Metab 2015; 29: 25-32
  • 55 Wierman ME, Arlt W, Basson R. et al. Androgen therapy in women: A reappraisal: An Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2014; 99: 3489-3510
  • 56 Burger-Stritt S, Pulzer A, Hahner S. Quality of life and life expectancy in patients with adrenal insufficiency: what is true and what is urban myth?. Front Horm Res 2016; 46: 171-183
  • 57 Stewart PM, Biller BM, Marelli C. et al. Exploring inpatient hospitalizations and morbidity in patients with adrenal insufficiency. J Clin Endocrinol Metab 2016; 101: 4843-4850
  • 58 Gunnarsson C, Ryan MP, Marelli C. et al. Health care burden in patients with adrenal insufficiency. J Endocr Soc 2017; 1: 512-523
  • 59 Quinkler M, Ekman B, Marelli C. et al. Prednisolone is associated with a worse lipid profile than hydrocortisone in patients with adrenal insufficiency. 2017; 1: 512-523
  • 60 Bergthorsdottir R, Leonsson-Zachrisson M, Odén A. et al. Premature mortality in patients with Addison’s disease: A population-based study. J Clin Endocrinol Metab 2006; 91: 4849-4853
  • 61 Quinkler M, Ekman B, Zhang P. et al. Mortality data from the European Adrenal Insufficiency Registry-Patient characterization and associations. Clin Endocrinol (Oxf) 2018; 89: 30-35
  • 62 White K, Arlt W. Adrenal crisis in treated Addison’s disease: A predictable but under-managed event. Eur J Endocrinol 2010; 162: 115-120
  • 63 Hahner S, Spinnler C, Fassnacht M. et al. High incidence of adrenal crisis in educated patients with chronic adrenal insufficiency: A prospective study. J Clin Endocrinol Metab 2015; 100: 407-416
  • 64 Quinkler M, Dahlqvist P, Husebye ES. et al. A European Emergency Card for adrenal insufficiency can save lives. Eur J Intern Med 2015; 26: 75-76
  • 65 Johannsson G, Nilsson AG, Bergthorsdottir R. et al. Improved cortisol exposure-time profile and outcome in patients with adrenal insufficiency: A prospective randomized trial of a novel hydrocortisone dual-release formulation. J Clin Endocrinol Metab 2012; 97: 473-481
  • 66 Nilsson AG, Bergthorsdottir R, Burman P. et al. Long-term safety of once-daily, dual-release hydrocortisone in patients with adrenal insufficiency: A phase 3b, open-label, extension study. Eur J Endocrinol 2017; 176: 715-725
  • 67 Isidori AM, Venneri MA, Graziadio C. et al. Effect of once-daily, modified-release hydrocortisone versus standard glucocorticoid therapy on metabolism and innate immunity in patients with adrenal insufficiency (DREAM): A single-blind, randomised controlled trial. Lancet Diabetes Endocrinol 2018; 6: 173-185
  • 68 Quinkler M, Miodini Nilsen R. et al. Modified-release hydrocortisone decreases BMI and HbA1c in patients with primary and secondary adrenal insufficiency. Eur J Endocrinol 2015; 172: 619-626
  • 69 Mallappa A, Sinaii N, Kumar P. et al. A phase 2 study of Chronocort, a modified-release formulation of hydrocortisone, in the treatment of adults with classic congenital adrenal hyperplasia. J Clin Endocrinol Metab 2015; 100: 1137-1145
  • 70 Whitaker MJ, Spielmann S, Digweed D. et al. Development and testing in healthy adults of oral hydrocortisone granules with taste masking for the treatment of neonates and infants with adrenal insufficiency. J Clin Endocrinol Metab 2015; 100: 1681-1688
  • 71 Gagliardi L, Nenke MA, Thynne TR. et al. Continuous subcutaneous hydrocortisone infusiontherapy in Addison’s disease: a randomized, placebo-controlled clinical trial. J Clin Endocrinol Metab 2014; 99: 4149-4157
  • 72 Oksnes M, Björnsdottir S, Isaksson M. et al. Continuous subcutaneous hydrocortisone infusion versus oral hydrocortisone replacement for treatment of Addison’s disease: A randomized clinical trial. J Clin Endocrinol Metab 2014; 99: 1665-1674
  • 73 Russell GM, Durant C, Ataya A. et al. Subcutaneous pulsatile glucocorticoid replacement therapy. Clin Endocrinol (Oxf) 2014; 81: 289-293
  • 74 Pearce SH, Mitchell AL, Bennett S. et al. Adrenal steroidogenesis after B lymphocyte depletion therapy in new-onset Addison’s disease. J Clin Endocrinol Metab 2012; 97: E1927-E1932
  • 75 Grodstein E, Hardy MA, Goldstein MJ. A case of human intramuscular adrenal gland transplantation as a cure for chronic adrenal insufficiency. Am J Transplant 2010; 10: 431-433
  • 76 Tajima T, Okada T, Ma XM. et al. Restoration of adrenal steroidogenesis by adenovirus-mediated transfer of human cytochrome P450 21-hydroxylase into the adrenal gland of 21-hydroxylase-deficientmice. Gene Ther. 1999; 6: 1898-1903
  • 77 Ludwig B, Reichel A, Steffen A. et al. Transplantation of human islets without immunosuppression. Proc Natl Acad Sci USA 2013; 110: 19054-19058
  • 78 Ruiz-Babot G, Balyura M, Hadjidemetriou I. et al. Modeling congenital adrenal hyperplasia and testing interventions for adrenal insufficiency using donor-specific reprogrammed cells. Cell Rep. 2018; 22: 1236-1249

Correspondence

Andreas Barthel
Medicover Bochum
Universitätsstrasse 48
44789 Bochum
Germany   

  • References

  • 1 Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet. 2014; 383: 2152-2167
  • 2 Addison T. On the Constitutional and Local Effects of Disease of the Supra-renal Capsules. London, UK: Samuel Highley; 1855
  • 3 Wintersteiner OP, Pfiffner J. Chemical studies on the adrenal cortex: II. J Biol Chem 1935; 111: 599-612
  • 4 Kendall EC. A chemical and physiological investigation of the suprarenal cortex. Symp Quant Biol 1937; 5: 299
  • 5 de Fremery P, Laquer E, Reichstein T. et al. Corticosterone: A crystalloid compound with biological activity of the adrenal-cortical hormone. Nature 1937; 139: 26-27
  • 6 Grollman A. Physiological and chemical studies on the adrenal cortical hormone. Symp Quant Biol 1937; 5: 313
  • 7 Sarett L. The synthesis of hydrocortisone from desoxy- cholic acid. J Biol Chem 1946; 162: 601
  • 8 Cutler HH, Power MH, Wilder RM. Concentrations of chloride, sodium and potassium in urine and blood: Their diagnostic significance in adrenal insufficiency. JAMA 1938; 111: 117-122
  • 9 Forsham PH, Thorn GW, Recant L. et al. Clinical experience with the use of the ACTH test for adrenal cortical function. J Clin Invest 1948; 27: 534
  • 10 Thorn GW, Forsham PH, Frawley TF. et al. Clinical experience with the use of cortisone acetate pellets in the treatment of adrenal cortical insufficiency. J Clin Endocrinol Metab 1950; 10: 833-834
  • 11 Erichsen MM, Løvås K, Fougner KJ. et al. Normal overall mortality rate in Addison’s disease, but young patients are at risk of premature death. Eur J Endocrinol 2009; 160: 233-237
  • 12 Mason AS, Meade TW, Lee JA. et al. Epidemiological and clinical picture of Addison’s disease. Lancet 1968; 2: 744-747
  • 13 Meyer G, Neumann K, Badenhoop K. et al. Increasing prevalence of Addison’s disease in German females: Health insurance data 2008–2012. Eur J Endocrinol 2014; 170: 367-373
  • 14 Laureti S, Vecchi L, Santeusanio F. et al. Is the prevalence of Addison’s disease underestimated?. J Clin Endocrinol Metab 1999; 84: 1762
  • 15 Arlt W, Allolio B. Adrenal insufficiency. Lancet 2003; 361: 1881-1893
  • 16 Bornstein SR. Predisposing factors for adrenal insufficiency. N Engl J Med 2009; 360: 2328-2339
  • 17 Betterle C, Morlin L. Autoimmune Addison’s disease. Endocr Dev 2011; 20: 161-172
  • 18 Mitchell AL, Pearce SH. Autoimmune Addison disease: Pathophysiology and genetic complexity. Nat Rev Endocrinol 2012; 8: 306-316
  • 19 Husebye ES, Lovas K. Immunology of Addison’s disease and premature ovarian failure. Endocrinol Metab Clin North Am 2009; 38: 389-405
  • 20 Husebye E, Lovas K. Pathogenesis of primary adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 2009; 23: 147-157
  • 21 Husebye ES, Anderson MS, Kämpe O. Autoimmune polyendocrine syndromes. N Engl J Med 2018; 378: 1132-1141
  • 22 Konda B, Nabhan F, Shah MH. Endocrine dysfunction following immune checkpoint inhibitor therapy. Curr Opin Endocrinol Diabetes Obes 2017; 24: 337-347
  • 23 Barroso-Sousa R, Barry WT, Garrido-Castro AC. et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: A systematic review and meta-analysis. JAMA Oncol 2018; 4: 173-182
  • 24 Boudjemaa A, Rousseau-Bussac G, Monnet I. Late-Onset adrenal insufficiency more than 1 year after stopping pembrolizumab. J Thorac Oncol 2018; 13: e39-e40
  • 25 Lovas K, Husebye ES. High prevalence and increasing incidence of Addison’s disease in western Norway. Clin Endocrinol (Oxf) 2002; 56: 787-791
  • 26 Bleicken B, Hahner S, Ventz M. et al. Delayed diagnosis of adrenal insufficiency is common: A cross-sectional study in 216 patients. Am J Med Sci 2010; 339: 525-531
  • 27 Bornstein SR, Allolio B, Arlt W. et al. Diagnosis and treatment of primary adrenal insufficiency: An endocrine society clinical practice guideline. J Clin Endocrinol Metab 2016; 101: 364-389
  • 28 Kazlauskaite R, Evans AT, Villabona CV. et al. Corticotropin tests for hypothalamic-pituitary-adrenal insufficiency: A metaanalysis. J Clin Endocrinol Metab 2008; 93: 4245-4253
  • 29 Oelkers W, Diederich S, Bähr V. Diagnosis and therapy surveillance in Addison’s disease: Rapid adrenocorticotropin(ACTH) test and measurement of plasma ACTH, renin activity, and aldosterone. J Clin Endocrinol Metab 1992; 75: 259-264
  • 30 Rehan M, Raizman JE, Cavalier E. et al. Laboratory challenges in primary aldosteronism screening and diagnosis. Clin Biochem. 2015; 48: 377-387
  • 31 Pofi R, Feliciano C, Sbardella E. et al. The short Synacthen (corticotropin) test can be used to predict recovery of hypothalamo-pituitary-adrenal axis function. J Clin Endocrinol Metab 2018; 103: 3050-3059
  • 32 Ueland GÅ, Methlie P, Øksnes M. Et al. The short cosyntropin test revisited: New normal reference range using LC-MS/MS. J Clin Endocrinol Metab 2018; 103: 1696-1703
  • 33 Mayenknecht J, Diederich S, Bähr V. et al. Comparison of low and high dose corticotropin stimulation tests in patients with pituitary disease. J Clin Endocrinol Metab 1998; 83: 1558-1562
  • 34 Pura M, Kreze Jr A, Kentos P. et al. The low-dose (1 microg) cosyntropin test (LDT) for primary adrenocortical insufficiency: Defining the normal cortisol response and report on first patients with Addison disease confirmed with LDT. Exp Clin Endocrinol Diabetes 2010; 118: 151-157
  • 35 Magnotti M, Shimshi M. Diagnosing adrenal insufficiency: Which test is best–the 1-microg or the 250-microg cosyntropin stimulation test?. Endocr Pract. 2008; 14: 233-238
  • 36 Oelkers W. The role of high- and low-dose corticotropin tests in the diagnosis of secondary adrenal insufficiency. Eur J Endocrinol 1998; 139: 567-570
  • 37 Dekkers OM, Timmermans JM, Smit JW. et al. Comparison of the cortisol responses to testing with two doses of ACTH in patients with suspected adrenal insufficiency. Eur J Endocrinol 2011; 164: 83-87
  • 38 Lindholm J, Kehlet H. Re-evaluation of the clinical value of the 30 min ACTH test in assessing the hypothalamic-pituitary-adrenocortical function. Clin Endocrinol (Oxf) 1987; 26: 53-59
  • 39 Klose M, Lange M, Rasmussen AK. et al. Factors influencing the adrenocorticotropin test: role of contemporary cortisol assays, body composition, and oral contraceptive agents. J Clin Endocrinol Metab 2007; 92: 1326-1333
  • 40 Vincent RP, Etogo-Asse FE, Dew T. et al. Serum total cortisol and free cortisol index give different information regarding the hypothalamus-pituitary-adrenal axis reserve in patients with liver impairment. Ann Clin Biochem 2009; 46: 505-507
  • 41 Dhillo WS, Kong WM, Le Roux CW. et al. Cortisol-binding globulin is important in the interpretation of dynamic tests of the hypothalamic pituitary–adrenal axis. Eur J Endocrinol 2002; 146: 231-235
  • 42 Plat L, Leproult R, L’Hermite-Baleriaux M. et al. Metabolic effects of short-term elevations of plasma cortisol are more pronounced in the evening than in the morning. J Clin Endocrinol Metab 1999; 84: 3082-3092
  • 43 Forss M, Batcheller G, Skrtic S. et al. Current practice of glucocorticoid replacement therapy and patient-perceived health outcomes in adrenal insufficiency - A worldwide patient survey. BMC Endocr Disord 2012; 12: 8
  • 44 Fadeev VV, Gitel EP, Mel’nichenko GA. The diurnal rhythm of adrenocorticotropic hormone secretion in the assessment of the adequacy of replacement therapy in primary chronic adrenal failure. Neurosci Behav Physiol 2001; 31: 237-242
  • 45 Filipsson H, Monson JP, Koltowska-Häggström M. et al. The impact of glucocorticoid replacement regimens on metabolic outcome and comorbidity in hypopituitary patients. J Clin Endocrinol Metab 2006; 91: 3954-3961
  • 46 Lebbe M, Arlt W. What is the best diagnostic and therapeutic management strategy for an Addison patient during pregnancy?. Clin Endocrinol (Oxf) 2013; 78: 497-502
  • 47 Jung C, Ho JT, Torpy DJ. et al. A longitudinal study of plasma and urinary cortisol in pregnancy and postpartum. J Clin Endocrinol Metab 2011; 96: 1533-1540
  • 48 Hardy RS, Zhou H, Seibel MJ. et al. Glucocorticoids and bone: Consequences of endogenous and exogenous excess and replacement therapy. Endocr Rev 2018; 39: 519-548
  • 49 Ueland GA, Husebye ES. Metabolic Complications in Adrenal Insufficiency. Front Horm Res 2018; 49: 104-113
  • 50 Allolio B. Extensive expertise in endocrinology. Adrenal crisis. Eur J Endocrinol 2015; 172: R115-R124
  • 51 Oelkers W, L’Age M. Control of mineralocorticoid substitution in Addison’s disease by plasma renin measurement. Klin Wochenschr. 1976; 54: 607-612
  • 52 Flad TM, Conway JD, Cunningham SK. et al. The role of plasma renin activity in evaluating the adequacy of mineralocorticoid replacement in primary adrenalin sufficiency. ClinEndocrinol (Oxf) 1996; 45: 529-534
  • 53 Quinkler M, Oelkers W, Remde H. et al. Mineralocorticoid substitution and monitoring in primary adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 2015; 29: 17-24
  • 54 Lang K, Burger-Stritt S, Hahner S. Is DHEA replacement beneficial in chronic adrenal failure?. Best Pract Res Clin Endocrinol Metab 2015; 29: 25-32
  • 55 Wierman ME, Arlt W, Basson R. et al. Androgen therapy in women: A reappraisal: An Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2014; 99: 3489-3510
  • 56 Burger-Stritt S, Pulzer A, Hahner S. Quality of life and life expectancy in patients with adrenal insufficiency: what is true and what is urban myth?. Front Horm Res 2016; 46: 171-183
  • 57 Stewart PM, Biller BM, Marelli C. et al. Exploring inpatient hospitalizations and morbidity in patients with adrenal insufficiency. J Clin Endocrinol Metab 2016; 101: 4843-4850
  • 58 Gunnarsson C, Ryan MP, Marelli C. et al. Health care burden in patients with adrenal insufficiency. J Endocr Soc 2017; 1: 512-523
  • 59 Quinkler M, Ekman B, Marelli C. et al. Prednisolone is associated with a worse lipid profile than hydrocortisone in patients with adrenal insufficiency. 2017; 1: 512-523
  • 60 Bergthorsdottir R, Leonsson-Zachrisson M, Odén A. et al. Premature mortality in patients with Addison’s disease: A population-based study. J Clin Endocrinol Metab 2006; 91: 4849-4853
  • 61 Quinkler M, Ekman B, Zhang P. et al. Mortality data from the European Adrenal Insufficiency Registry-Patient characterization and associations. Clin Endocrinol (Oxf) 2018; 89: 30-35
  • 62 White K, Arlt W. Adrenal crisis in treated Addison’s disease: A predictable but under-managed event. Eur J Endocrinol 2010; 162: 115-120
  • 63 Hahner S, Spinnler C, Fassnacht M. et al. High incidence of adrenal crisis in educated patients with chronic adrenal insufficiency: A prospective study. J Clin Endocrinol Metab 2015; 100: 407-416
  • 64 Quinkler M, Dahlqvist P, Husebye ES. et al. A European Emergency Card for adrenal insufficiency can save lives. Eur J Intern Med 2015; 26: 75-76
  • 65 Johannsson G, Nilsson AG, Bergthorsdottir R. et al. Improved cortisol exposure-time profile and outcome in patients with adrenal insufficiency: A prospective randomized trial of a novel hydrocortisone dual-release formulation. J Clin Endocrinol Metab 2012; 97: 473-481
  • 66 Nilsson AG, Bergthorsdottir R, Burman P. et al. Long-term safety of once-daily, dual-release hydrocortisone in patients with adrenal insufficiency: A phase 3b, open-label, extension study. Eur J Endocrinol 2017; 176: 715-725
  • 67 Isidori AM, Venneri MA, Graziadio C. et al. Effect of once-daily, modified-release hydrocortisone versus standard glucocorticoid therapy on metabolism and innate immunity in patients with adrenal insufficiency (DREAM): A single-blind, randomised controlled trial. Lancet Diabetes Endocrinol 2018; 6: 173-185
  • 68 Quinkler M, Miodini Nilsen R. et al. Modified-release hydrocortisone decreases BMI and HbA1c in patients with primary and secondary adrenal insufficiency. Eur J Endocrinol 2015; 172: 619-626
  • 69 Mallappa A, Sinaii N, Kumar P. et al. A phase 2 study of Chronocort, a modified-release formulation of hydrocortisone, in the treatment of adults with classic congenital adrenal hyperplasia. J Clin Endocrinol Metab 2015; 100: 1137-1145
  • 70 Whitaker MJ, Spielmann S, Digweed D. et al. Development and testing in healthy adults of oral hydrocortisone granules with taste masking for the treatment of neonates and infants with adrenal insufficiency. J Clin Endocrinol Metab 2015; 100: 1681-1688
  • 71 Gagliardi L, Nenke MA, Thynne TR. et al. Continuous subcutaneous hydrocortisone infusiontherapy in Addison’s disease: a randomized, placebo-controlled clinical trial. J Clin Endocrinol Metab 2014; 99: 4149-4157
  • 72 Oksnes M, Björnsdottir S, Isaksson M. et al. Continuous subcutaneous hydrocortisone infusion versus oral hydrocortisone replacement for treatment of Addison’s disease: A randomized clinical trial. J Clin Endocrinol Metab 2014; 99: 1665-1674
  • 73 Russell GM, Durant C, Ataya A. et al. Subcutaneous pulsatile glucocorticoid replacement therapy. Clin Endocrinol (Oxf) 2014; 81: 289-293
  • 74 Pearce SH, Mitchell AL, Bennett S. et al. Adrenal steroidogenesis after B lymphocyte depletion therapy in new-onset Addison’s disease. J Clin Endocrinol Metab 2012; 97: E1927-E1932
  • 75 Grodstein E, Hardy MA, Goldstein MJ. A case of human intramuscular adrenal gland transplantation as a cure for chronic adrenal insufficiency. Am J Transplant 2010; 10: 431-433
  • 76 Tajima T, Okada T, Ma XM. et al. Restoration of adrenal steroidogenesis by adenovirus-mediated transfer of human cytochrome P450 21-hydroxylase into the adrenal gland of 21-hydroxylase-deficientmice. Gene Ther. 1999; 6: 1898-1903
  • 77 Ludwig B, Reichel A, Steffen A. et al. Transplantation of human islets without immunosuppression. Proc Natl Acad Sci USA 2013; 110: 19054-19058
  • 78 Ruiz-Babot G, Balyura M, Hadjidemetriou I. et al. Modeling congenital adrenal hyperplasia and testing interventions for adrenal insufficiency using donor-specific reprogrammed cells. Cell Rep. 2018; 22: 1236-1249