Mifepristone is the first and only available glucocorticoid receptor antagonist. It was initially mainly considered as a so-called ‘contragestive’ pill due to its antiprogestin activity. In this review, we summarize the results of mifepristone reported in the literature as a treatment of Cushing’s syndrome. Most of the patients were treated due to unsuccessful surgery and/or partially effective anticortisolic drugs. The majority of them presented a rapid decrease of clinical signs of hypercortisolism during the first month of treatment; about half experienced a reduction in their elevated blood pressure, and half of the diabetic patients presented improved blood glucose levels. Mifepristone treatment has 2 main drawbacks: (1) the blockade of glucocorticoid receptors leads to increased ACTH and cortisol levels, making it difficult to adapt the treatment and diagnose adrenal deficiency, and (2) increased cortisol levels can also lead to severe hypokalemia. Follow-up of efficacy should only be clinical (weight, blood pressure, skin lesions) and biological (regular blood potassium sampling). Dose adjustment will be performed based on these parameters. The lack of a large available prospective cohort of patients on mifepristone, and the scarcity of data on its long-term effects, does not allow recommending it as a first-line drug in the treatment of hypercortisolism. However, as mifepristone is a rapidly effective drug, it can play a role in the management of hypercortisolism. The main indication is the partial efficacy or bad tolerance of other well-known anticortisolic drugs, either by replacement (bad tolerance, lack of effectiveness) or addition (multimodal approach) of mifepristone.

Cushing’s syndrome can be caused by adrenal, pituitary or ectopic tumors. Surgery remains the first-line treatment, and can be followed by pituitary irradiation or bilateral adrenalectomy if necessary. Medical management remains, however, of major interest when surgery is impossible, in preparation for surgery, or when surgery is partially effective. The efficacy of usual anticortisolic drugs (ketoconazole, metyrapone, mitotane, etc.) is not constant and may be limited by their poor tolerance [1,2]. Mifepristone, a glucocorticoid receptor antagonist initially considered as a so-called ‘contragestive’ pill, may represent an alternative [3]. Our recent European collaborative retrospective study [3] allowed to discuss the potential future role of this drug in the management of Cushing’s syndrome. In the present review, we will mainly talk about mifepristone, as data about other potential glucocorticoid receptor antagonists are only experimental.

Mifepristone is a substituted 19-nor steroid compound chemically designated as 11β-[p-(dimethylamino)phenyl]-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one. It is the first and currently only available glucocorticoid receptor antagonist treatment [4]. Other glucocorticoid receptors are currently evaluated, but no clinical trial has been published to date [5]. Mifepristone has also a strong anti-progestin activity, and a weak anti-androgen activity: its relative binding affinity at the glucocorticoid receptor is more than three times that of dexamethasone and more than ten times that of cortisol; affinity at the progesterone receptor is more than twice that of progesterone, and affinity at the androgen receptor is less than one third that of testosterone. It does not bind to the estrogen receptor or the mineralocorticoid receptor. Following oral administration of a single dose of 400–600 mg, mifepristone is rapidly absorbed, with a peak plasma concentration occurring approximately 90 min after ingestion. Of note, in the absence of progesterone or cortisol, mifepristone can also act as a weak agonist [5,6].

Mifepristone appears as a rapidly effective drug in controlling signs of hypercortisolism. To date, 37 patients, mainly adults, have been reported in the literature as having received mifepristone for various etiologies of Cushing’s syndrome (table 1) [7,8,9,10,11,12,13,14,15,16]. Most of them were treated due to unsuccessful surgery and partially effective or badly tolerated medical treatment. Up to 85% presented a decrease of their clinical signs of hypercortisolism (weight loss, improvement of skin signs) during the first month of mifepristone (fig. 1). About half experienced a reduction in their elevated blood pressure and half of the diabetic patients also presented improved blood glucose levels, sometimes leading to withdrawal of antidiabetic drugs. The majority of the patients have been treated for an advanced adrenal carcinoma, ectopic ACTH secretion or Cushing’s disease.

Table 1

Individual data of patients treated with mifepristone for adrenal carcinoma (patients 1–17), ectopic ACTH secretion (18–29), Cushing’s disease (30–34) or other causes (35–37)

Individual data of patients treated with mifepristone for adrenal carcinoma (patients 1–17), ectopic ACTH secretion (18–29), Cushing’s disease (30–34) or other causes (35–37)
Individual data of patients treated with mifepristone for adrenal carcinoma (patients 1–17), ectopic ACTH secretion (18–29), Cushing’s disease (30–34) or other causes (35–37)
Fig. 1

Clinical picture of a patient before (left) and 6 months after (right) treatment with mifepristone for an ectopic ACTH syndrome. Note the decrease in upper body obesity and facial erythrosis after treatment.

Fig. 1

Clinical picture of a patient before (left) and 6 months after (right) treatment with mifepristone for an ectopic ACTH syndrome. Note the decrease in upper body obesity and facial erythrosis after treatment.

Close modal

Adrenal Carcinoma

Seventeen patients have been reported to date. In the majority of cases, mifepristone was given in advanced adrenal carcinomas after failed surgery, and lack of efficacy of classical anticortisolic drugs (including mitotane). Median duration of treatment was 3 months (range 0.25–9). Clinical signs of hypercortisolism improved in 76% of the cases during the first month of treatment. In our series, 60% of the patients presented improved blood pressure levels. Interestingly, one patient with severe psychosis had a drastic improvement of psychiatric signs in the first few days of mifepristone treatment.

Ectopic ACTH Secretion

Twelve patients have been treated with mifepristone after unsuccessful surgery or lack of identified tumor. Median duration of treatment was 5.5 months (range 0.3–18). All patients presented an improvement of their clinical signs. About half of them experienced decreased blood pressure levels. Severe psychosis signs improved in the first few days of mifepristone treatment in 1 patient.

Cushing’s Disease

Five patients have been treated to date because of the lack of pituitary adenoma visualized on MRI, failed surgery, bad tolerance to other anticortisolic drugs or partial efficacy of a radiosurgical procedure. Mean duration of treatment was 11 months (range 0.5–24). All but one patient presented improved clinical signs. Again, 1 patient with severe psychosis had a drastic improvement of psychiatric signs in the first few days of mifepristone treatment.

Of note, 1 patient has been treated for bilateral adrenal hyperplasia, and 2 for a likely benign adrenal adenoma (but the precise etiology is unclear). All of them experienced a rapid improvement of clinical signs of hypercortisolism during the first month of mifepristone treatment [10].

In Cushing’s syndrome, hypokalemia is directly related to glucocorticoid excess as massive hypercortisolemia leads to incomplete renal inactivation of cortisol by 11β-dehydrogenase, and hence mineralocorticoid excess. Mifepristone requires careful medical attention and a close follow-up of this specific point. As mifepristone blocks only glucocorticoid action, the mineralocorticoid activity of cortisol excess is not affected by mifepristone treatment, thus leading to hypokalemia. On the contrary, mifepristone may lead to an increase of plasma ACTH and consecutively of cortisol levels in some patients with Cushing’s syndrome, particularly with Cushing’s disease due to alterations in negative feedback (a mechanism of hypokalemia similar to the one observed in Cushing’s syndrome without treatment) [5]. Mifepristone-induced hypokalemia was observed in one third of the 37 Cushing’s patients reported to date. In a few cases, hypokalemia was very severe, requiring high doses of oral and intravenous potassium, and intensive anti-aldosterone treatment. Severe hypokalemia should thus represent a contraindication for this treatment and potassium levels need to be carefully monitored. The same mechanism can induce increased blood pressure levels, which was observed in 5 patients, despite an improvement of clinical signs of hypercortisolism. The ideal treatment remains high dose anti-aldosterone treatment: there is no need for a systematic prescription at the onset of mifepristone, except in case of low-normal hypokalemia.

Moreover, the lack of available biological parameters of follow-up on treatment obviously represents a limitation for the use of mifepristone. Signs suggestive of adrenal insufficiency have been reported in 16% of the 37 patients reported to date. The blockade of glucocorticoid receptors does not allow determining the efficacy of the drug by measuring ACTH and cortisol levels that are increased by mifepristone treatment. However, this low rate of adrenal deficiency is puzzling taking into account the mechanism of action of mifepristone: it is probably due to a partial blockade of glucocorticoid receptor antagonists, or a weak agonist effect of the drug [5]. Management of adrenal deficiency is also difficult: hydrocortisone is not effective; dexamethasone (1 mg for 400 mg of mifepristone) should be given during 48 h in parallel with mifepristone withdrawal. Mifepristone can be re-initiated at a lower dose after the treatment of adrenal deficiency [17].

Antiprogestin effects are also important to notice as they can induce an endometrial hyperplasia (relative hyperestrogenia). Three patients have been reported in the literature with this adverse effect [14]. Pelvic ultrasonography should be performed yearly in every woman before menopause.

Mifepristone should be initiated at a low dose (200–400 mg/day), and increased every 2–4 weeks based on clinical efficacy and tolerance. The maximal dose given should be around 400–800 mg/day. Increasing the dose up to 1,000 mg/day is usually not necessary, and increases the risk of adverse effects. However, increase in the dose of mifepristone could be performed more rapidly in case of severe signs of psychosis [10].

Follow-up of efficacy, and dose adjustments should mainly rely on clinical and necessarily imperfect parameters like weight, blood pressure or skin lesions. ACTH and cortisol levels will be increased, and do not represent a good marker of the efficacy of the drug. Adrenal deficiency should be suspected in case of weakness, fatigue, nausea, vomiting, and hypoglycemic episodes [10].

As mentioned previously, only a limited number of patients have been treated long-term with mifepristone, probably because more than a third of patients reported to date presented an advanced adrenal carcinoma. If long-term treatment is considered, pelvic ultrasonography should be performed at least yearly in nonmenopausal women. No specific adverse effects other than the ones seen at short-term have been reported.

The potential risks and benefits of mifepristone have to be weighed against alternative treatment options. The majority of other anticortisolic drugs have the major advantage to induce decreased cortisol levels: they are thus easier to follow (regular measures of plasma cortisol levels), whereas during mifepristone, cortisol concentration provides no guidance for treatment. However, the other anticortisolic drugs classically expose patients to frequent and/or severe side effects without superior efficacy. Ketoconazole has been reported to induce serious and life-threatening hepatotoxicity, although this side effect is rare (1/15,000 cases). Moreover, ketoconazole is no longer available in some European countries (e.g. Germany) [2]. Metyrapone, an inhibitor of 11β-hydroxylase, is not easily available in several European countries and may also be associated with significant side effects [18]. Etomidate can only be used intravenously and, therefore, should be reserved for severe cases [19]. The use of mitotane can be difficult due to a narrow therapeutic window, and overall bad tolerance (gastrointestinal and neurotoxic effects). Moreover, most of these treatments are not as rapidly effective as mifepristone. This is particularly true for mitotane, which requires several weeks before being fully effective [20,21].

Thus, treatment with mifepristone may be of significant value in the medical treatment of Cushing’s syndrome in a high percentage of cases. Its place must be defined in regards of the severity of the disease. Due to the low number of patients reported, mifepristone cannot be recommended as a first-line treatment, even in comparison with more classical anticortisolic drugs. Mifepristone should be reserved to patients with lack of/partial efficacy and/or bad tolerance of well-known therapeutic procedures, i.e. surgery, radiotherapy and other anticortisolic drugs. Of note, mifepristone can be added to other anticortisolic drugs, as they have different mechanisms of actions. In this case, follow-up (mainly in terms of signs of adrenal deficiency and potassium levels) should be very strict.

Mifepristone represents a rapidly effective treatment to control signs of hypercortisolism with the main drawback of the impossibility to follow blood cortisol levels. The risk of severe hypokalemia needs to be closely monitored and further limits its widespread use. Despite these promising results, the lack of a large available prospective cohort of patients on mifepristone, and the scarcity of data on its long-term effects, does not allow recommending it as a first-line drug in the treatment of hypercortisolism. However, as mifepristone is a rapidly effective drug, its main indication is the partial efficacy or bad tolerance of other well-known anticortisolic drugs especially in patients with psychiatric signs of hypercortisolism. Only a limited number of patients have been treated long-term with mifepristone and if long-term treatment is considered, one should keep in mind the anti-progestin activity of mifepristone, as this can lead to endometrial hyperplasia.

The authors have nothing to disclose.

1.
Biller BM, Grossman AB, Stewart PM, Melmed S, Bertagna X, Bertherat J, Buchfelder M, Colao A, Hermus AR, Hofland LJ, Klibanski A, Lacroix A, Lindsay JR, Newell-Price J, Nieman LK, Petersenn S, Sonino N, Stalla GK, Swearingen B, Vance ML, Wass JA, Boscaro M: Treatment of adrenocorticotropin-dependent Cushing’s syndrome: a consensus statement. J Clin Endocrinol Metab 2008;93:2454–2462.
2.
Castinetti F, Morange I, Jaquet P, Conte-Devolx B, Brue T: Ketoconazole revisited: a preoperative or postoperative treatment in Cushing’s disease. Eur J Endocrinol 2008;158:91–99.
3.
Johanssen S, Allolio B: Mifepristone (ru 486) in Cushing’s syndrome. Eur J Endocrinol 2007;157:561–569.
4.
Bertagna X, Bertagna C, Luton JP, Husson JM, Girard F: The new steroid analog ru 486 inhibits glucocorticoid action in man. J Clin Endocrinol Metab 1984;59:25–28.
5.
McMaster A, Ray DW: Drug insight: selective agonists and antagonists of the glucocorticoid receptor. Nat Clin Pract 2008;4:91–101.
6.
Miller JW, Crapo L: The medical treatment of Cushing’s syndrome. Endocr Rev 1993;14:443–458.
7.
Beaufrere B, de Parscau L, Chatelain P, Morel Y, Aguercif M, Francois R: Ru 486 administration in a child with Cushing’s syndrome. Lancet 1987;ii:217.
8.
Bilgin YM, van der Wiel HE, Fischer HR, De Herder WW: Treatment of severe psychosis due to ectopic Cushing’s syndrome. J Endocrinol Invest 2007;30:776–779.
9.
Cassier PA, Abou-Amara-Olivieri S, Artru P, Lapalus MG, Riou JP, Lombard-Bohas C: Mifepristone for ectopic acth secretion in metastatic endocrine carcinomas: report of two cases. Eur J Endocrinol 2008;158:935–938.
10.
Castinetti F, Fassnacht M, Johanssen S, Terzolo M, Bouchard P, Chanson P, Do Cao C, Morange I, Pico A, Ouzounian S, Young J, Hahner S, Brue T, Allolio B, Conte-Devolx B: Merits and pitfalls of mifepristone in Cushing’s syndrome. Eur J Endocrinol 2009;160:1003–1010.
11.
Chu JW, Matthias DF, Belanoff J, Schatzberg A, Hoffman AR, Feldman D: Successful long-term treatment of refractory cushing’s disease with high-dose mifepristone (ru 486). J Clin Endocrinol Metab 2001;86:3568–3573.
12.
Contreras P: Adrenal cancer: tumor regression with ketoconazole or mifepristone (ru 486): in vivo and in vitro evidence supporting tumoral hormone dependency. Proc 69th Ann Endocrine Society Meeting, Indianapolis, 1987, p 23.
13.
Donckier JE, Michel LA, Berbinschi A, De Coster PM, De Plaen JF, Ketelslegers JM, Buysschaert M: Late recurrence of operated adrenocortical carcinoma: atrial natriuretic factor before and after treatment with mitotane. Surgery 1989;105:690–692.
14.
Newfield RS, Spitz IM, Isacson C, New MI: Long-term mifepristone (ru486) therapy resulting in massive benign endometrial hyperplasia. Clin Endocrinol 2001;54:399–404.
15.
Nieman LK, Chrousos GP, Kellner C, Spitz IM, Nisula BC, Cutler GB, Merriam GR, Bardin CW, Loriaux DL: Successful treatment of cushing’s syndrome with the glucocorticoid antagonist ru 486. J Clin Endocrinol Metab 1985;61:536–540.
16.
van der Lely AJ, Foeken K, van der Mast RC, Lamberts SW: Rapid reversal of acute psychosis in the Cushing syndrome with the cortisol-receptor antagonist mifepristone (ru 486). Ann Intern Med 1991;114:143–144.
17.
Raux-Demay MC, Pierret T, Bouvier d’Yvoire M, Bertagna X, Girard F: Transient inhibition of ru 486 antiglucocorticoid action by dexamethasone. J Clin Endocrinol Metab 1990;70:230–233.
18.
Verhelst JA, Trainer PJ, Howlett TA, Perry L, Rees LH, Grossman AB, Wass JA, Besser GM: Short and long-term responses to metyrapone in the medical management of 91 patients with Cushing’s syndrome. Clin Endocrinol 1991;35:169–178.
19.
Schulte HM, Benker G, Reinwein D, Sippell WG, Allolio B: Infusion of low dose etomidate: correction of hypercortisolemia in patients with Cushing’s syndrome and dose-response relationship in normal subjects. J Clin Endocrinol Metab 1990;70:1426–1430.
20.
Hahner S, Fassnacht M: Mitotane for adrenocortical carcinoma treatment. Curr Opin Investig Drugs 2005;6:386–394.
21.
Terzolo M, Angeli A, Fassnacht M, et al: Adjuvant mitotane treatment for adrenocortical carcinoma. N Engl J Med 2007;356:2372–2380.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.