Which drug would be administered to control glucose levels in a patient with endogenous Cushing syndrome?

Endogenous Cushing’s syndrome is a rare condition caused by too much cortisol in your body. “Endogenous” means that something inside your body causes too much cortisol production.

Pharmacotherapy

Medications used in the management of Cushing syndrome include the following:

  • 11-beta-hydroxylase inhibitor: Osilodrostat

  • Somatostatin analogs: Pasireotide

  • Adrenal steroid inhibitors: Metyrapone, ketoconazole, etomidate

  • Glucocorticoid receptor antagonist: Mifepristone

  • Adrenolytic agents: Mitotane

Pasireotide (Signifor) is a somatostatin analog that binds and activates human somatostatin receptors resulting in inhibition of ACTH secretion, which leads to decreased cortisol secretion. It is indicated for treatment of adults with Cushing disease in whom pituitary surgery is not an option or has not been curative.

In a phase 3 trial [32]  in which 162 patients were randomly assigned to receive either 0.6 or 0.9 mg of pasireotide subcutaneously twice a day, twelve (15%) participants in the lower-dose group and 21 (26%) of the higher-dosage group met the trial's primary endpoint (free urinary cortisol levels at or below the upper limit of normal with no dose increase in 6 months). Patients with baseline levels not exceeding 5 times the upper limit of the normal range were found to achieve normal urinary free cortisol level more often than those with higher baseline levels. Other observations included improvements in systolic and diastolic blood pressure, low-density lipoprotein cholesterol, weight, and health-related quality of life. However, 48% of patients who did not have diabetes at baseline became diabetic. Seventy-three percent of the study participants experienced hyperglycemia-related adverse events, leading to 6% leaving the study and 46% requiring a new glucose-lowering medication. Cholelithiasis was also associated with its use.

In an extension treatment study of pasireotide from an original 15-day, phase 2 study, pasireotide was shown to maintain the effect on lowering free urinary cortisol levels in some patients up to 24 months. This supports the extended treatment of pasireotide in some patients with Cushing disease. [30]

Agents that inhibit steroidogenesis, such as mitotane, ketoconazole, metyrapone, and etomidate, have been used to cause medical adrenalectomy. These medications are often are toxic at the doses required to reduce cortisol secretion. For instance, ketoconazole's prescribing information was revised to include a black box warning regarding hepatotoxicity, including fatalities and liver transplantation. Thus, medical treatment should be initiated cautiously and, ideally, in consultation with a specialist. Efficacy of these medical interventions can be assessed with serial measurements of 24-hour urinary free cortisol. Patients receiving these medications may require glucocorticoid replacement to avoid adrenal insufficiency. Patients should be counseled on the signs and symptoms of adrenal insufficiency when starting these drugs.

An orally administered steroidogenesis inhibitor, osilodrostat (Isturisa) acts on 11-beta-hydroxylase, an enzyme that catalyzes the last step of cortisol synthesis in the adrenal cortex. It is indicated for adults with Cushing disease who cannot undergo pituitary surgery or in whom the operation has not been curative.

Approval of osilodrostat by the US Food and Drug Administration (FDA) in March 2020 was based on a phase-3, multicenter, double-blind, randomized withdrawal study by Biller et al in which the drug was significantly more successful than placebo in maintaining mean urinary free cortisol (mUFC) within normal limits at the end of the study’s withdrawal period (86% vs 29% of patients, respectively). The median baseline value for mUFC had been 3.5 times the upper limit of normal. [33]

Metyrapone is an agent that competitively inhibits a single steroidogenic enzyme. Ketoconazole acts at several sites. In ACTH-dependent Cushing syndrome, ACTH secretion continues to stimulate steroidogenesis, which counters the actions of these medications.

Because ACTH production may persist or increase in patients with Cushing disease, radiation therapy of the pituitary is often required after unsuccessful initial therapy, either surgical or medical. These agents have higher efficacy when used in combination because they may act synergistically.

Ketoconazole has been the most popular and effective of these agents for long-term use and usually has been the agent of choice. However, the FDA has issued a warning that states clinicians should no longer prescribe ketoconazole, except to treat some life-threatening fungal infections; this is due to increased risk for severe liver injury, adrenal insufficiency, and adverse drug interactions. Ketoconazole acts on several of the P450 enzymes, including the first step in cortisol synthesis, cholesterol side-chain cleavage, and conversion of 11-deoxycortisol to cortisol. A daily dose of 600-800 mg often decreases cortisol production. If this agent is ineffective at controlling hypercortisolism, the dose may be maintained while another steroid enzyme inhibitor, typically metyrapone, is initiated.

Adverse effects of ketoconazole include headache, sedation, nausea, irregular menses, decreased libido, impotence, gynecomastia, and elevated liver function tests. The drug is contraindicated during pregnancy.

Ketoconazole is less effective in patients on H2 blockers or proton-pump inhibitors because gastric acidity is required for metabolism.

Patients should be informed of the hepatotoxicity risks and monitored closely while taking this drug. Drug-to-drug interaction should also be reviewed. Certain drugs when used concurrently with Ketoconazole can increase risk of prolong QT interval or changes in drug metabolism. Concomitant use with cisapride, disopyramide, dofetilide, dronedarone, methadone, pimozide, quinidine, and ranolazine is contraindicated due to the possible occurrence of life-threatening ventricular arrhythmias such as torsade de pointes.

Metyrapone blocks 11-beta-hydroxylase activity, the final step in cortisol synthesis. Therapy is begun at 1 g/d divided into 4 doses and increased to a maximum dose of 4.5 g/d. Adverse effects present from increases in androgen and mineralocorticoid precursors and include hypertension, acne, and hirsutism.

Etomidate, an imidazole-derivative anesthetic agent, blocks 11-beta-hydroxylase. It is used intravenously at 0.3 mg/kg/h. Its use is limited by the requirement for administration by the intravenous route. However, it rapidly decreases cortisol concentration and may be used as an adjunct to impending surgical procedure.

Mitotane is an adrenolytic agent that acts by inhibiting 11-beta hydroxylase and cholesterol side-chain cleavage enzymes. This drug also leads to mitochondrial destruction and necrosis of adrenocortical cells in the zona fasciculata and reticularis. For this reason, it is used in treatment of adrenal cancer at doses of 2-4 g daily. Its survival benefit is unclear. It can be used in addition to radiation therapy for treatment of Cushing disease and in combination with metyrapone for treatment of ectopic ACTH secretion.

Unfortunately, mitotane is expensive, and its utility is limited by the adverse gastrointestinal and neurologic effects, including nausea, diarrhea, dizziness, and ataxia. Other adverse effects include rash, arthralgias, and leukopenia. Mitotane is taken up by adipose tissues and persists in the circulation long after discontinuation. It is a potential teratogen and can cause abortion; therefore, it is relatively contraindicated in women interested in remaining fertile.

Mifepristone (Korlym) is an antiprogestational agent, which, at high doses, competitively binds to the glucocorticoid and progesterone receptors. [34] In February 2012, the FDA approved mifepristone to control hyperglycemia secondary to hypercortisolism in adult patients with endogenous Cushing syndrome who have type 2 diabetes mellitus or glucose intolerance and have failed surgery, or are not candidates for surgery.

The effect of mifepristone was reported in a 24-week, multicenter, open-label trial after failed multimodality therapy at 14 United States academic medical centers and three private research centers. [35] Fifty Cushing syndrome patients were enrolled in the study. Forty-three had Cushing disease, of which 42 had prior surgery, four patients had ectopic ACTH-producing tumors and three had adrenal cancer. Response was defined as at least a 25% decrease in area under the curve for glucose on a standard oral glucose tolerance test from baseline to 24 weeks, an amount considered clinically meaningful improvement in glucose control. Twenty-nine patients enrolled in the study were glucose intolerant. Sixty percent of patients met the study’s primary endpoint. There was also a statistically significant reduction in mean HbA1c over the course of the study, from 7.43% at baseline to 6.29% at study conclusion (p< 0.001). Diastolic blood pressure response, weight reduction, and waist circumference decrease were also observed in study subjects. Overall, 87% had significant improvement in clinical status. Insulin resistance, depression, cognition, and quality of life also improved.

Patients should be monitored for adrenal insufficiency, hypokalemia, prolonged QT interval. Use of mifepristone will result in termination of pregnancy. When Mifepristone is used in women with Cushing’s syndrome, pregnancy must be excluded prior to initiation of therapy. Nonhormonal form of contraception must be used during treatment and for 1 month after stopping therapy.

Agents that decrease CRH or ACTH release have been studied for the treatment of Cushing disease. Such agents include Carbegoline, bromocriptine, Cyproheptadine, valproic acid, and octreotide.

Treatment options for patients with persistent or recurrent Cushing disease have included pituitary irradiation and repeat surgery. A small study found that a percentage of such patients respond to cabergoline therapy. [36] Longer-term follow-up and a larger number of patients are needed to confirm these encouraging preliminary results.

An ongoing phase 3 study is being conducted to confirm long-term efficacy and safety of agent LCI699 for the treatment of patients with Cushing disease. [37] LCI699 is a potent inhibitor of 11β-hydroxylase, which catalyzes the final step of cortisol synthesis. In a prior multicenter proof-of-concept 10-week study in which 12 patients were enrolled and completed the study, all 12 patients achieved urinary free cortisol level less than or equal to upper limit of normal or a 50% or greater decrease from baseline at day 70; 11 (92%) had normal UFC levels at that time. No serious side effects were reported. Common side effects were headache, fatigue, and nausea. [38]

Surgical Therapy

The treatment of choice for endogenous Cushing syndrome is surgical resection of the causative tumor. The primary therapy for Cushing disease is transsphenoidal surgery, and the primary therapy for adrenal tumors is adrenalectomy.

Other surgical interventions include the following:

  • Bilateral adrenalectomy

  • Unilateral adrenalectomy

  • Resection of carcinomas

Patients with endogenous Cushing syndrome who undergo resection of pituitary, adrenal, or ectopic tumors should receive stress doses of glucocorticoid in the intraoperative and immediate postoperative period. Typically, hydrocortisone is infused intravenously, either continuously (10 mg/h) or in boluses (80-100 mg q8h) starting prior to surgery and for the first 24 hours afterward. If the patient does well, intravenous glucocorticoid replacement may be tapered over 1-2 days and replaced with an oral formulation. The rate of steroid taper may be slowed if severe preoperative hypercortisolism was present. In the event of pituitary destruction or bilateral adrenalectomy, lifelong glucocorticoid replacement is necessary. Lifelong mineralocorticoid replacement is also necessary in those patients who undergo bilateral adrenalectomy.

Cushing disease

Treatment of choice for classic Cushing disease is transsphenoidal surgery by an experienced neurosurgeon. The goal of surgery is to remove the adenoma, preserving as much pituitary function as possible.

  • The more extensive the mass and the resulting resection, the greater the risk for loss of pituitary function. Successful amelioration of hypercortisolism occurs in 60-80% of cases. Lateralization of ACTH secretion via IPS catheterization and sampling is sometimes helpful in difficult cases.

  • Pituitary irradiation is employed when transsphenoidal surgery is not successful or not possible. The procedure is less successful than surgery in adults, with a 40-50% cure rate in adults and 85% cure rate in children. Late-onset adverse effects include hypopituitarism.

  • Bilateral adrenalectomy is an option if transsphenoidal surgery, pituitary irradiation, and medical therapy fail or if rapid normalization of cortisol levels is required. The patient then requires lifelong glucocorticoid and mineralocorticoid therapy.

  • Individuals who undergo bilateral adrenalectomy might develop Nelson syndrome, which is symptomatic enlargement of the pituitary gland and adenoma. This may occur in one quarter to one half of adults not treated with pituitary irradiation and in as many as one quarter of patients pretreated with radiation therapy.

Ectopic adrenocorticotropic production

Surgical resection of the source of ACTH production may not always be possible. Ectopic ACTH-producing tumors are often difficult to locate.

Medical therapy or bilateral adrenalectomy may be required.

Adrenal source

Adenomas may be removed with unilateral adrenalectomy, often with a laparoscopic approach.

Carcinomas should be resected for possible cure and palliation.

Micronodular or macronodular hyperplasia causing Cushing syndrome may be treated effectively by bilateral adrenalectomy. Unilateral or subtotal adrenalectomy may lead to recurrence.

Consultations

Effective diagnosis and management of Cushing disease is often facilitated by involvement of an endocrinologist and the appropriate experienced neurosurgeon.

Complications

The successful treatment of hypercortisolemia, with the accompanying recovery of the immune system, permits the subsequent manifestation of autoimmune disease. A study by Tatsi et al found that following the resolution of hypercortisolemia in 129 children with endogenous Cushing syndrome, 10 patients (7.8%) were found to have a new autoimmune disorder or related condition. Specifically, these individuals were diagnosed—6 to 19 months after treatment—with celiac disease, psoriasis, Hashimoto thyroiditis, Graves disease, optic neuritis, skin hypopigmented lesions/vitiligo, allergic rhinitis/asthma, or neuropathy (with this last responding to glucocorticoid therapy). [39]

  1. Nieman LK. Recent Updates on the Diagnosis and Management of Cushing's Syndrome. Endocrinol Metab (Seoul). 2018 Jun. 33 (2):139-46. [QxMD MEDLINE Link]. [Full Text].

  2. Chaudhry HS, Bhimji SS. Cushing Syndrome. 2018 Jan. [QxMD MEDLINE Link]. [Full Text].

  3. Marieb NJ, Spangler S, Kashgarian M, Heimann A, Schwartz ML, Schwartz PE. Cushing's syndrome secondary to ectopic cortisol production by an ovarian carcinoma. J Clin Endocrinol Metab. 1983 Oct. 57 (4):737-40. [QxMD MEDLINE Link].

  4. Carey RM, Varma SK, Drake CR Jr, Thorner MO, Kovacs K, Rivier J, et al. Ectopic secretion of corticotropin-releasing factor as a cause of Cushing's syndrome. A clinical, morphologic, and biochemical study. N Engl J Med. 1984 Jul 5. 311 (1):13-20. [QxMD MEDLINE Link].

  5. Tatsi C, Boden R, Sinaii N, et al. Decreased lymphocytes and increased risk for infection are common in endogenous pediatric Cushing syndrome. Pediatr Res. 2018 Feb. 83 (2):431-7. [QxMD MEDLINE Link]. [Full Text].

  6. Brown RJ, Kelly MH, Collins MT. Cushing syndrome in the McCune-Albright syndrome. J Clin Endocrinol Metab. 2010 Apr. 95 (4):1508-15. [QxMD MEDLINE Link].

  7. Assie G, Bahurel H, Coste J, Silvera S, Kujas M, Dugue MA, et al. Corticotroph tumor progression after adrenalectomy in Cushing's Disease: A reappraisal of Nelson's Syndrome. J Clin Endocrinol Metab. 2007 Jan. 92 (1):172-9. [QxMD MEDLINE Link].

  8. Lindholm J, Juul S, Jorgensen JO, Astrup J, Bjerre P, Feldt-Rasmussen U, et al. Incidence and late prognosis of cushing's syndrome: a population-based study. J Clin Endocrinol Metab. 2001 Jan. 86 (1):117-23. [QxMD MEDLINE Link].

  9. Graversen D, Vestergaard P, Stochholm K, Gravholt CH, Jorgensen JO. Mortality in Cushing's syndrome: a systematic review and meta-analysis. Eur J Intern Med. 2012 Apr. 23 (3):278-82. [QxMD MEDLINE Link].

  10. Hara T, Akutsu H, Yamamoto T, Ishikawa E, Matsuda M, Matsumura A. Cushing's disease presenting with gastrointestinal perforation: a case report. Endocrinol Diabetes Metab Case Rep. 2013. 2013:130064. [QxMD MEDLINE Link].

  11. Graham BS, Tucker WS Jr. Opportunistic infections in endogenous Cushing's syndrome. Ann Intern Med. 1984 Sep. 101 (3):334-8. [QxMD MEDLINE Link].

  12. Davi' MV, Cosaro E, Piacentini S, et al. Prognostic factors in ectopic Cushing's syndrome due to neuroendocrine tumors: a multicenter study. Eur J Endocrinol. 2017 Apr. 176 (4):451-9. [QxMD MEDLINE Link].

  13. Pereira AM, Delgado V, Romijn JA, Smit JW, Bax JJ, Feelders RA. Cardiac dysfunction is reversed upon successful treatment of Cushing's syndrome. Eur J Endocrinol. 2010 Feb. 162 (2):331-40. [QxMD MEDLINE Link].

  14. Tyrrell JB, Findling JW, Aron DC, Fitzgerald PA, Forsham PH. An overnight high-dose dexamethasone suppression test for rapid differential diagnosis of Cushing's syndrome. Ann Intern Med. 1986 Feb. 104 (2):180-6. [QxMD MEDLINE Link].

  15. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, et al. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2008 May. 93 (5):1526-40. [QxMD MEDLINE Link].

  16. Meikle AW. Dexamethasone suppression tests: usefulness of simultaneous measurement of plasma cortisol and dexamethasone. Clin Endocrinol (Oxf). 1982 Apr. 16 (4):401-8. [QxMD MEDLINE Link].

  17. Carroll T, Raff H, Findling JW. Late-night salivary cortisol for the diagnosis of Cushing syndrome: a meta-analysis. Endocr Pract. 2009 May-Jun. 15 (4):335-42. [QxMD MEDLINE Link].

  18. Elamin MB, Murad MH, Mullan R, Erickson D, Harris K, Nadeem S, et al. Accuracy of diagnostic tests for Cushing's syndrome: a systematic review and metaanalyses. J Clin Endocrinol Metab. 2008 May. 93 (5):1553-62. [QxMD MEDLINE Link].

  19. Elias PC, Martinez EZ, Barone BF, Mermejo LM, Castro M, Moreira AC. Late-night salivary cortisol has a better performance than urinary free cortisol in the diagnosis of Cushing's syndrome. J Clin Endocrinol Metab. 2014 Jun. 99 (6):2045-51. [QxMD MEDLINE Link].

  20. Yanovski JA, Cutler GB Jr, Chrousos GP, Nieman LK. Corticotropin-releasing hormone stimulation following low-dose dexamethasone administration. A new test to distinguish Cushing's syndrome from pseudo-Cushing's states. JAMA. 1993 May 5. 269 (17):2232-8. [QxMD MEDLINE Link].

  21. Yanovski JA, Cutler GB Jr, Chrousos GP, Nieman LK. Corticotropin-releasing hormone stimulation following low-dose dexamethasone administration. A new test to distinguish Cushing's syndrome from pseudo-Cushing's states. JAMA. 1993 May 5. 269 (17):2232-8. [QxMD MEDLINE Link].

  22. Martin NM, Dhillo WS, Banerjee A, Abdulali A, Jayasena CN, Donaldson M, et al. Comparison of the dexamethasone-suppressed corticotropin-releasing hormone test and low-dose dexamethasone suppression test in the diagnosis of Cushing's syndrome. J Clin Endocrinol Metab. 2006 Jul. 91 (7):2582-6. [QxMD MEDLINE Link].

  23. Wester VL, Reincke M, Koper JW, et al. Scalp hair cortisol for diagnosis of Cushing's syndrome. Eur J Endocrinol. 2017 Jun. 176 (6):695-703. [QxMD MEDLINE Link].

  24. Elhomsy G, Staros E. Dexamethasone Suppression Test. Medscape Drugs & Diseases. Available at http://emedicine.medscape.com/article/2114191-overview. Accessed: 5/30/15.

  25. Aron DC, Raff H, Findling JW. Effectiveness versus efficacy: the limited value in clinical practice of high dose dexamethasone suppression testing in the differential diagnosis of adrenocorticotropin-dependent Cushing's syndrome. J Clin Endocrinol Metab. 1997 Jun. 82 (6):1780-5. [QxMD MEDLINE Link].

  26. Nieman LK, Oldfield EH, Wesley R, Chrousos GP, Loriaux DL, Cutler GB Jr. A simplified morning ovine corticotropin-releasing hormone stimulation test for the differential diagnosis of adrenocorticotropin-dependent Cushing's syndrome. J Clin Endocrinol Metab. 1993 Nov. 77 (5):1308-12. [QxMD MEDLINE Link].

  27. Reimondo G, Paccotti P, Minetto M, Termine A, Stura G, Bergui M, et al. The corticotrophin-releasing hormone test is the most reliable noninvasive method to differentiate pituitary from ectopic ACTH secretion in Cushing's syndrome. Clin Endocrinol (Oxf). 2003 Jun. 58 (6):718-24. [QxMD MEDLINE Link].

  28. Oldfield EH, Doppman JL, Nieman LK, Chrousos GP, Miller DL, Katz DA, et al. Petrosal sinus sampling with and without corticotropin-releasing hormone for the differential diagnosis of Cushing's syndrome. N Engl J Med. 1991 Sep 26. 325 (13):897-905. [QxMD MEDLINE Link].

  29. Mulligan GB, Eray E, Faiman C, Gupta M, Pineyro MM, Makdissi A, et al. Reduction of false-negative results in inferior petrosal sinus sampling with simultaneous prolactin and corticotropin measurement. Endocr Pract. 2011 Jan-Feb. 17 (1):33-40. [QxMD MEDLINE Link].

  30. Tucker ME. New guidelines address Cushing’s syndrome treatment. Medscape Medical News. Available at http://www.medscape.com/viewarticle/849084. Aug 5,2015; Accessed: Sept 3, 2015.

  31. Aymes S. Endocrine Society releases guidelines on treatment of Cushing’s Syndrome. Endocrinology Advisor. Aug 26, 2015.

  32. Colao A, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. A 12-month phase 3 study of pasireotide in Cushing's disease. N Engl J Med. 2012 Mar 8. 366 (10):914-24. [QxMD MEDLINE Link].

  33. Biller BMK, Newell-Price J, Fleseriu M, et al. OR16-2 Osilodrostat Treatment in Cushing's Disease (CD): Results from a Phase III, Multicenter, Double-Blind, Randomized Withdrawal Study (LINC 3). J Endocr Soc. 2019 Apr-May. 3(Suppl_1):[Full Text].

  34. Johanssen S, Allolio B. Mifepristone (RU 486) in Cushing's syndrome. Eur J Endocrinol. 2007 Nov. 157 (5):561-9. [QxMD MEDLINE Link].

  35. Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart DE, Gross C, et al. Mifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing's syndrome. J Clin Endocrinol Metab. 2012 Jun. 97 (6):2039-49. [QxMD MEDLINE Link].

  36. Lila AR, Gopal RA, Acharya SV, George J, Sarathi V, Bandgar T, et al. Efficacy of cabergoline in uncured (persistent or recurrent) Cushing disease after pituitary surgical treatment with or without radiotherapy. Endocr Pract. 2010 Nov-Dec. 16 (6):968-76. [QxMD MEDLINE Link].

  37. Safety and Efficacy of LC1699 for the Treatment of Patients With Cushing's Disease. ClinicalTrials.gov. Available at https://clinicaltrials.gov/ct2/show/NCT02180217. Accessed: 7/13/15.

  38. Bertagna X, Pivonello R, Fleseriu M, Zhang Y, Robinson P, Taylor A, et al. LCI699, a potent 11β-hydroxylase inhibitor, normalizes urinary cortisol in patients with Cushing's disease: results from a multicenter, proof-of-concept study. J Clin Endocrinol Metab. 2014 Apr. 99 (4):1375-83. [QxMD MEDLINE Link].

  39. Tatsi C, Keil M, Lyssikatos C, Belyavskaya E, Stratakis CA, Lodish MB. Incidence of Autoimmune and Related Disorders After Resolution of Endogenous Cushing Syndrome in Children. Horm Metab Res. 2018 Apr. 50 (4):290-5. [QxMD MEDLINE Link].

  40. Dekkers OM, Horvath-Puho E, Jorgensen JO, Cannegieter SC, Ehrenstein V, Vandenbroucke JP, et al. Multisystem morbidity and mortality in Cushing's syndrome: a cohort study. J Clin Endocrinol Metab. 2013 Jun. 98 (6):2277-84. [QxMD MEDLINE Link].

  41. Boscaro M, Bertherat J, Findling J, Fleseriu M, Atkinson AB, Petersenn S, et al. Extended treatment of Cushing's disease with pasireotide: results from a 2-year, Phase II study. Pituitary. 2014 Aug. 17 (4):320-6. [QxMD MEDLINE Link].

  • Physical findings in Cushing syndrome.

  • Diagnosis of Cushing syndrome.

  • Etiology of Cushing syndrome.

  • High power H and E of a pituitary adenoma showing a monotonous proliferation of bland cells. Not all pituitary adenomas result in Cushing syndrome, only those that produce ACTH.

  • Intermediate power H and E of an adrenal adenoma showing a monotonous proliferation of bland cells. Not all adrenal adenomas or carcinomas result in Cushing's syndrome; only those that produce ACTH.

Author

Coauthor(s)

Catherine Anastasopoulou, MD, PhD, FACE Associate Professor of Medicine, The Steven, Daniel and Douglas Altman Chair of Endocrinology, Sidney Kimmel Medical College of Thomas Jefferson University; Einstein Endocrine Associates, Einstein Medical Center

Catherine Anastasopoulou, MD, PhD, FACE is a member of the following medical societies: American Association of Clinical Endocrinologists, American Society for Bone and Mineral Research, Endocrine Society, Philadelphia Endocrine Society

Disclosure: Nothing to disclose.

Specialty Editor Board

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Romesh Khardori, MD, PhD, FACP (Retired) Professor, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Eastern Virginia Medical School

Romesh Khardori, MD, PhD, FACP is a member of the following medical societies: American Association of Clinical Endocrinologists, American College of Physicians, American Diabetes Association, Endocrine Society

Disclosure: Nothing to disclose.

Acknowledgements

Gail K Adler, MD, PhD, Associate Professor of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School.

Disclosure: Nothing to disclose.

Susanna L Dipp, MD Fellow, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School

Disclosure: Nothing to disclose

Don S Schalch, MD Professor Emeritus, Department of Internal Medicine, Division of Endocrinology, University of Wisconsin Hospitals and Clinics

Disclosure: Nothing to disclose

Which drug would be administered to control glucose levels in a patient with endogenous Cushing syndrome and who has a history of type 2 diabetes?

In 2012, the FDA approved mifepristone (Korlym, Corcept Therapeutics) as a once-daily oral medication to treat adults with Cushing's syndrome who had elevated blood glucose due to type 2 diabetes.
The adrenal-directed drug ketoconazole is the most commonly used drug, mainly because of its rapid action, whereas the glucocorticoid receptor antagonist, mifepristone, is highly effective in controlling clinical comorbidities, mainly glucose intolerance, thus being a useful treatment for CD when it is associated with ...

What is the treatment of choice to patients with Cushing's disease?

Treatment of choice for classic Cushing disease is transsphenoidal surgery by an experienced neurosurgeon. The goal of surgery is to remove the adenoma, preserving as much pituitary function as possible. The more extensive the mass and the resulting resection, the greater the risk for loss of pituitary function.

How blood glucose level is changed in Cushing's syndrome patient?

Impaired glucose metabolism is prevalent in Cushing's syndrome. Cushing's syndrome causes a significant reduction in insulin sensitivity and glucose disappearance (peripheral uptake of glucose). Excess cortisol induces both lipolysis and lipogenesis.