575
Views
52
CrossRef citations to date
0
Altmetric
Review

Cushing’s syndrome: epidemiology and developments in disease management

, &
Pages 281-293 | Published online: 17 Apr 2015

Abstract

Cushing’s syndrome is a rare disorder resulting from prolonged exposure to excess glucocorticoids. Early diagnosis and treatment of Cushing’s syndrome is associated with a decrease in morbidity and mortality. Clinical presentation can be highly variable, and establishing the diagnosis can often be difficult. Surgery (resection of the pituitary or ectopic source of adrenocorticotropic hormone, or unilateral or bilateral adrenalectomy) remains the optimal treatment in all forms of Cushing’s syndrome, but may not always lead to remission. Medical therapy (steroidogenesis inhibitors, agents that decrease adrenocorticotropic hormone levels or glucocorticoid receptor antagonists) and pituitary radiotherapy may be needed as an adjunct. A multidisciplinary approach, long-term follow-up, and treatment modalities customized to each individual are essential for optimal control of hypercortisolemia and management of comorbidities.

Introduction

Cushing’s syndrome is a rare disorder caused by prolonged exposure to excess glucocorticoids. The clinical presentation is highly variable. While the diagnosis can be straightforward in florid cases, establishing the diagnosis can be challenging in cases with mild hypercortisolism and subtle clinical features, especially given the overlap in symptoms in individuals with and without the syndrome.Citation1 Here, we discuss the epidemiology, diagnosis, and advances in management of endogenous Cushing’s syndrome.

Epidemiology and prognosis

Administration of supraphysiologic doses of glucocorticoids is the most common cause of Cushing’s syndrome (exogenous or iatrogenic Cushing’s syndrome).Citation2 As glucocorticoids are used to treat inflammatory, autoimmune, and neoplastic disorders, a detailed medication history is essential. Although the oral route is most commonly associated with iatrogenic Cushing’s syndrome, any mode of delivery, including inhaled, topical, or injectable glucocorticoids, should be sought.

Endogenous Cushing’s syndrome is rare, with an incidence of 0.7–2.4 per million population per year.Citation1,Citation3,Citation4 A population-based study from Denmark reported a diagnosis of Cushing’s syndrome in 166 patients over an 11-year period (1985–1995), yielding an incidence of two cases per million inhabitants per year.Citation3 Of the 139 patients with nonmalignant disease, 23 (16.5%) died during follow-up (median 8 years), yielding a standard mortality ratio of 3.7 (95% confidence interval 2.3–5.3), with the highest mortality during the first year after initial presentation. Eight deaths occurred before initiation of treatment. Causes of death included suicide (n=1), cardiac rupture (n=1), stroke (n=1), and severe infections (n=3; peritonitis, septicemia, and pneumonia). The cause of death could not be ascertained in two patients. A similar study from Spain reported 49 cases of Cushing’s syndrome over 18 years, yielding an incidence of 2.4 cases per million inhabitants per year, with a standard mortality ratio of 3.8.Citation4 Although the mortality risk decreases with remission of hypercortisolism,Citation5 it is not equivalent to that of the general population. Several studies have shown that increased morbidity, especially related to cardiovascular disease, persists for several years after biochemical remission.Citation6Citation9 Furthermore, varying criteria for remission of hypercortisolemia across studies make it difficult to interpret these outcome measures accurately.

Although population-based studies demonstrate a low incidence of endogenous Cushing’s syndrome, more evaluations of patients with uncontrolled diabetes mellitus or hypertension suggest that this may be an underestimation.Citation10Citation13 Leibowitz et al screened 90 obese subjects with uncontrolled diabetes mellitus (hemoglobin A1c >9%), and found three (3.3%) to have Cushing’s syndrome.Citation10 Similarly, Catargi et al diagnosed Cushing’s syndrome in four (2%) of 200 obese subjects with uncontrolled diabetes mellitus.Citation11 However, this reported prevalence of 2%–5% was not confirmed in other studies.Citation14Citation17 Also, an evaluation of 369 overweight and obese subjects with at least two other features of Cushing’s syndrome did not identify any with Cushing’s syndrome, but 84 subjects had at least one abnormal screening test result.Citation18 Widespread screening for Cushing’s syndrome in overweight individuals or patients with type 2 diabetes mellitus is therefore not recommended.Citation18,Citation19 Instead, a case-finding approach in patients with other features of Cushing’s syndrome or uncontrolled diabetes or hypertension despite appropriate treatment may be indicated.Citation19

Currently, extensive use of computed tomography (CT) and magnetic resonance imaging (MRI) scans has led to an increasing number of incidentally found adrenal masses. The prevalence of these adrenal “incidentalomas” increases from 0.2% to 7% with increasing age.Citation20Citation22 “Subclinical” or subtle Cushing’s syndrome has been reported in 5%–10% of these patients, who represent a population in which Cushing’s syndrome is more common.Citation22,Citation23

Classification of Cushing’s syndrome

Traditionally, endogenous Cushing’s syndrome is classified as adrenocorticotropic hormone (ACTH)-dependent or ACTH-independent. ACTH-dependent Cushing’s syndrome accounts for 80%–85% of cases. Of these, 75%–80% are due to ACTH production from a pituitary adenoma (Cushing’s disease [CD]), 15%–20% are due to ACTH production from nonpituitary tumors (ectopic ACTH syndrome [EAS]) and <1% are caused by corticotropin-releasing hormone (CRH)-producing tumors.Citation1,Citation23 Most pituitary tumors are sporadic, resulting from monoclonal expansion of a single mutated cell. Rarely, they may occur as part of a genetic syndrome, the most common being multiple endocrine neoplasia type 1 and familial isolated pituitary adenomas. Ectopic ACTH secretion most often derives from small-cell carcinoma of the lung or pulmonary carcinoid tumor. Other causes include pancreatic neuroendocrine tumors (NETs), thymic NETs, gastrinomas, medullary thyroid cancer, and pheochromocytoma.Citation24

ACTH-independent Cushing’s syndrome accounts for 15%–20% of endogenous Cushing’s syndrome in adults; 90% are unilateral adrenal tumors. Of these, adenomas are the cause in ~80% of the cases, while the others are adrenocortical carcinoma.Citation25 Rare adrenal causes of Cushing’s syndrome include macronodular adrenal hyperplasia, primary pigmented nodular adrenal disease (sporadic or as part of Carney’s complex) and McCune–Albright syndrome. In a recent study, Louiset et al described intra-adrenal production of ACTH and paracrine regulation of cortisol secretion in 30 cases of bilateral macronodular adrenal hyperplasia.Citation26 Proopiomelanocortin messenger ribonucleic acid expression was detected in all hyperplastic tissue samples and ACTH was detected in steroidogenic cell clusters throughout adrenal tissue specimens. ACTH levels were also found to be higher in adrenal venous samples of two patients compared to the periphery.Citation26 These findings bring into question the traditional classification of “ACTH-independent Cushing’s syndrome”.Citation27

In childhood and adolescence, as in adults, exogenous glucocorticoids are the most common cause of Cushing’s syndrome. The causes of endogenous Cushing’s syndrome in children are similar to those in adults, with some differences. Cushing’s syndrome in infancy is commonly associated with McCune–Albright syndrome, adrenocortical tumors are usually the cause in children less than 5–7 years of age, CD is the commonest cause after 7 years of age, and EAS is extremely rare.Citation28

Clinical features of Cushing’s syndrome

The clinical presentation of Cushing’s syndrome is variable (). It is influenced by age and sex and the severity and duration of the disease.Citation1,Citation24,Citation28Citation31 No single sign or symptom is pathognomonic.

Table 1 Clinical features of Cushing’s syndrome

While weight gain is the most common sign of Cushing’s syndrome, it is also extremely common in patients without the syndrome.Citation18 Moreover, an underlying malignancy or paraneoplastic wasting syndrome can mask the weight gain associated with hypercortisolism. As may be concluded from , establishing a diagnosis based on clinical features alone can be difficult. Some of the features that are thought to distinguish Cushing’s syndrome more reliably from simple obesity include proximal muscle weakness, easy bruising, and violaceous striae greater than 1 cm wide. Decreased linear growth along with progressive weight gain is one of the hallmarks of pediatric Cushing’s syndrome.Citation32 Detailed description of differences in presentation between pediatric and adult Cushing’s syndrome is beyond the scope of this review.Citation28,Citation33,Citation34

The clinical presentation of Cushing’s syndrome can vary by sex. Men with CD are more likely to present at a younger age with more florid clinical features. Violaceous striae, muscle weakness and atrophy, osteoporosis, and kidney stones are much more common.Citation29 Gonadal dysfunction is common in both men and women, presenting as decreased libido, infertility, menstrual irregularity or amenorrhea in women, and erectile dysfunction in men. While CD has a female preponderance (female:male ratio 3–4:1), EAS is equally common in both sexes among the adult population.Citation24 Unlike adults, pediatric CD is characterized by significant male preponderance in the prepubertal years. With increasing age, the sex distribution of CD equalizes toward puberty, with a trend toward female preponderance in adulthood.Citation28,Citation33,Citation34

Classically, patients with ectopic Cushing’s have severe hypercortisolemia associated with a rapid onset of symptoms and a florid presentation, hypokalemia, and severe or opportunistic infections.Citation24,Citation35,Citation36 However, these features reflect the severity of hypercortisolism, and can be seen in patients with CD. By contrast, the clinical presentation of many ACTH-secreting carcinoid tumors may be indistinguishable from that of CD, reflecting less extreme hypercortisolism.Citation24,Citation37

Decreased bone mineral density, osteoporosis, and fractures are present in 50%–80% of patients with Cushing’s syndrome. After cure, bone mineral density improves, but additional specific treatment for fractures and related pain may be needed. Bone loss can be more severe in primary adrenal disease compared to CD. This may be related to a protective effect of the higher adrenal androgen levels in CD.Citation38,Citation39 However, these findings have not been reproduced in other studies, and the significance of disease etiology in bone loss and fractures remains controversial.Citation40,Citation41

Psychiatric and cognitive dysfunction is present in 70%–85% of patients with Cushing’s syndrome. Depression, emotional lability, and irritability are the most common manifestations; acute psychosis, mania, anxiety, panic attacks, suicidal ideation, and paranoia are rarer.Citation42Citation45 Hypercortisolemia is also associated with a decrease in brain volume, particularly the hippocampus, and related impairment in learning and short-term memory.Citation46,Citation47 Although psychiatric and cognitive symptoms improve after remission, many symptoms may persist.Citation45,Citation46

Excess glucocorticoids have a catabolic effect on skeletal muscles, skin, and connective tissue. Increased protein wasting and type II muscle-fiber atrophy is associated with significant muscle weakness, with predominant involvement of the pelvic girdle musculature.Citation48,Citation49 Vertebral fractures, back pain, and depression further lead to decreased mobility and disuse muscle atrophy. A persistently impaired quality of life, primarily of the physical domain, and persistent muscle weakness have been documented even several years after remission.Citation50Citation53

Dyslipidemia (increased low-density lipoprotein, decreased high-density lipoprotein) and glucose intolerance occur in 45%–70% of patients.Citation54Citation56 This reported prevalence of glucose intolerance is likely an underestimate, as many patients with normal fasting glucose have underlying glucose intolerance, and not all patients with Cushing’s syndrome undergo glucose-tolerance testing. Hypertension (cortisol-mediated enhancement of vascular reactivity to vasoconstrictors and the mineralocorticoid effects of cortisol), though not invariably present, is frequently seen in patients with Cushing’s syndrome, with a prevalence of approximately 80%.Citation35,Citation55 Hepatic steatosis and increased visceral adipose tissue are common in patients with Cushing’s syndrome.Citation57,Citation58 These features of metabolic syndrome, along with a hypercoagulable state,Citation59 lead to an increased cardiovascular risk that may not return to baseline after successful treatment.Citation60Citation62

Diagnosis of Cushing’s syndrome

Clinical suspicion in patients with multiple and progressive signs and symptoms suggestive of Cushing’s syndrome should provoke diagnostic testing. Screening may be considered in patients with other features of Cushing’s syndrome, particularly with poorly controlled diabetes or hypertension, or unexplained osteoporosis. Patients with an incidentally discovered adrenal mass should be evaluated.

It is important to differentiate between the pathological hypercortisolemia of endogenous Cushing’s syndrome and that associated with pregnancy, glucocorticoid resistance, and pseudo-Cushing’s states like alcoholism, depression, severe obesity, anorexia nervosa, and bulimia. The hypercortisolism of pseudo-Cushing’s states is thought to be mediated via increased hypothalamic secretion of CRH. In contrast, hypothalamic CRH is suppressed in true Cushing’s syndrome.

Biochemical tests in Cushing’s syndrome are based on the cardinal features of increased endogenous secretion of cortisol, loss of normal feedback of the hypothalamic–pituitary–adrenal axis, and loss of the normal cortisol circadian rhythm. According to the 2008 Endocrine Society guidelines, the following tests should be used for the diagnosis of Cushing’s syndrome: 24-hour urinary free cortisol (UFC), late-night salivary cortisol, and/or a low-dose dexamethasone-suppression test (DST; 1 mg overnight or 2 mg/day over 48 hours).Citation19 None of these tests has 100% diagnostic accuracy; each test has its own caveats, and multiple tests are usually needed to establish the diagnosis ().Citation1,Citation19,Citation31,Citation63

Table 2 Sensitivity, specificity, and caveats of screening tests for Cushing’s syndrome

UFC is an index of circulating free (biologically active) cortisol. Unlike plasma cortisol, which measures total cortisol (bound and unbound), UFC results are not affected by factors that modulate the levels of corticosteroid-binding globulin (CBG). Inappropriate urine collection, increased fluid intake, and abnormal renal function can be associated with false-positive and -negative results.Citation19,Citation31,Citation63 Review of urine volume and creatinine excretion can help assess the adequacy of urine collection. UFC values in Cushing’s syndrome are variable.Citation64,Citation65 Cushing’s syndrome is highly unlikely in the setting of three or more normal UFC levels, except in rare cases of cyclic Cushing’s syndrome. Values more than three- to fourfold the upper limit of normal are generally diagnostic of Cushing’s syndrome.Citation1,Citation19,Citation31

DSTs rely on the loss of glucocorticoid negative-feedback inhibition of CRH and ACTH secretion. In the overnight test, 1 mg of dexamethasone is given at 11 pm, followed by measurement of fasting plasma cortisol between 8 and 9 am the next morning. In the 2-day test, 0.5 mg of dexamethasone is given every 6 hours (starting at 9 am) for 2 days, with cortisol measurement at the beginning and end of the test. A cortisol level of ≤1.8 μg/dL (50 nmol/L) at the end of either test excludes Cushing’s syndrome ().Citation19 Variable absorption and metabolism of dexamethasone can affect the results of the DSTs. Estrogen treatment and pregnancy can increase CBG levels, leading to false-positive DST results. For accurate results, estrogen treatment should be stopped for 4–6 weeks to allow CBG levels to return to baseline.Citation1,Citation19 Some (3%–8%) CD patients show suppression of cortisol levels below 1.8 μg/dL (50 nmol/L) on DST.Citation1,Citation66 Multiple repeated tests may be needed to establish the diagnosis in these cases.

Salivary cortisol, like UFC, measures free cortisol, and is therefore not affected by CBG levels. Diagnostic ranges vary between studies, due to the different assays and comparison groups used to set cutoff points.Citation63 Salivary cortisol can be measured by immunoassay or liquid chromatography–tandem mass spectrometry (LC/MS-MS). Given the lower sensitivity of LC/MS-MS (as it may not measure cortisol metabolites/precursors), immunoassay remains the preferred methodology for screening of Cushing’s syndrome.Citation67Citation69 Educating the patient on optimal timing of specimen collection while taking into account their normal sleep–wake cycle, as well as avoiding exciting or stressful experiences on the evening of the test, are important for an accurate salivary cortisol result. False-positive results can also be seen, due to contamination of the saliva sample with widely available nonprescription topical hydrocortisone creams and ointments. However, overall, ease of collection, stability at room temperature, and a greater than 90% sensitivity and specificity make it a highly useful test, especially in outpatients, children, and in assessment of cyclic Cushing’s syndrome.Citation67Citation69 Midnight plasma cortisol levels also can be used for screening. A single sleeping midnight plasma cortisol level of less than 1.8 μg/dL (50 nmol/L) excludes active Cushing’s syndrome.Citation70 A midnight plasma cortisol level, drawn while the patient is resting but not fully asleep, of more than 7.5 μg/dL (207 nmol/L) is a more specific cutoff, but can miss up to 7% cases of mild Cushing’s syndrome.Citation71,Citation72

If the diagnosis remains unclear, dexamethasone-suppressed CRH stimulation (Dex-CRH) testCitation73,Citation74 or the desmopressin testCitation75,Citation76 can be performed to distinguish further between CD and pseudo-Cushing’s states. However, the exact diagnostic accuracy of these tests and the optimal cutoffs for diagnosis need further evaluation.Citation77 Moreover, similarly to other dexamethasone tests, variable absorption and metabolism of dexamethasone, especially in the setting of concomitant use of other commonly prescribed medications, can affect the accuracy of the test.Citation78 Recently, a prospective study of 73 patients with clinical features of hypercortisolism and an abnormal DST or UFC result reported a positive predictive value of 100% and a negative predictive value of 90% for the Dex-CRH test (sensitivity 94%, specificity 100%) using a 15-minute post-CRH cortisol cutoff of 3.2 μg/dL (87 nmol/L).Citation79

Establishing the cause of Cushing’s syndrome

Once the diagnosis of Cushing’s syndrome has been established, the next step is to differentiate between the three causes. Measurement of plasma ACTH levels is the initial step in the differential diagnosis. A two-site immunoradiometric assay is preferred over radioimmunoassay, because it better discriminates low or suppressed ACTH levels.Citation80 To avoid falsely low results, ACTH levels should be measured on multiple occasions, and samples should be collected in prechilled ethylenediaminetetraacetic acid tubes, transported in an ice bath, and processed immediately.Citation1 Values <5 pg/mL (1.1 pmol/L) suggest ACTH-independent Cushing’s syndrome. Imaging studies of the adrenal gland can then identify unilateral or bilateral lesions. The glands may appear normal in primary pigmented nodular adrenal disease. Patients with proven ACTH-independent hypercortisolism but normal adrenal imaging studies should undergo screening for exogenous glucocorticoids, assessment for other features of Carney’s complex, and possibly genetic testing for PRKAR1A mutations.Citation81 A paradoxical increase in glucocorticoid excretion in response to dexamethasone during the Liddle test (0.5 mg dexamethasone every 6 hours for 48 hours, followed by 2 mg every 6 hours for 48 hours) can further help identify patients with primary pigmented nodular adrenocortical disease.Citation82 Recently, an inactivating germ-line mutation of ARMC5, a putative tumor-suppressor gene, has been implicated in the development of primary bilateral macronodular hyperplasia.Citation83Citation85 Patients with this mutation show familial clustering and bilateral disease, and often present with a more severe clinical phenotype. Genetic testing for this mutation may help in earlier diagnosis and better management of these cases.Citation85

An inappropriately normal or elevated ACTH level (>20 pg/ml, 4.4 pmol/L) is consistent with an ACTH-dependent form of Cushing’s syndrome. Patients with mild adrenal Cushing’s may not have suppressed ACTH levels. Moreover, recently, intra-adrenal production of ACTH has been reported in macronodular adrenal hyperplasia, although peripheral ACTH levels were suppressed.Citation26 CRH stimulation and high-dose DSTs may help differentiate between adrenal and ACTH-dependent forms of Cushing’s syndrome when ACTH results are indeterminate.Citation1

Although there is significant overlap in ACTH levels between CD and EAS, extremely high levels (>500 pg/mL, 110 pmol/L) usually reflect EAS.Citation1,Citation31 Sex may help: 90% of cases of ACTH-dependent Cushing’s syndrome in women are caused by CD.

A pituitary MRI should be obtained in ACTH-dependent cases. A large mass (>6 mm) strongly suggests CD. However, ACTH-secreting pituitary tumors are usually small and may not be detected, even with newer, more advanced MRI techniques (spoiled gradient-recalled acquisition or dynamic MRI sequences) in 20%–58% of patients with CD.Citation86,Citation87 Moreover, ~10% of “healthy” individuals can have incidental pituitary lesions up to 6 mm in size.Citation88 Differential diagnosis of ACTH-dependent Cushing’s syndrome can therefore be very challenging.

The high-dose DST (2 mg given every 6 hours for 48 hours, or the overnight test with a single 8 mg dose) is based on the concept that corticotrope adenomas arise clonally from normal cells and retain some sensitivity to glucocorticoid negative feedback, while ectopic ACTH-secreting tumors do not. In the overnight test, a greater than 69% suppression of cortisol levels following 8 mg of dexamethasone suggests a pituitary source of ACTH.Citation89 Logistic regression modeling has shown that the diagnostic accuracy of this test is less than the pretest likelihood based on clinical features alone.Citation90 Due to the poor diagnostic accuracy (~80%) of a high-dose DST, many endocrinologists do not recommend performing the test unless inferior petrosal sinus sampling (IPSS) is not available.Citation1,Citation91

In the CRH-stimulation test, recombinant ovine or human sequence CRH (1 μg/kg, maximum 100 μg dose) is used to stimulate corticotrope tumors to secrete ACTH. Most patients with CD respond with an increase in ACTH (>34%) and/or cortisol (>20%) levels within 45 minutes of intravenous administration of ovine CRH (sensitivity 93%).Citation92 Following administration of human CRH, most CD patients have at least a 14% increase in cortisol levels (sensitivity 85%, specificity 100%).Citation93 While some reports have shown ovine CRH to be superior to recombinant human CRH,Citation94 others have found similar responses.Citation95 A systematic review of all published series on the CRH-stimulation test revealed that 7%–14% of patients with CD fail to respond to CRH.Citation31 Some patients with ACTH-secreting pulmonary carcinoids (~10%) can respond to dexamethasone and/or CRH.Citation24

If the CRH-stimulation test and high-dose DST are both consistent with a pituitary source and imaging studies identify a pituitary lesion consistent with an adenoma (>6 mm), no further testing is necessary. On the other hand, if biochemical testing is discordant and/or the pituitary MRI is normal or equivocal (lesion <6 mm), IPSS with ACTH measurements before and after CRH administration should be performed.Citation1,Citation96 A central-to-peripheral ACTH gradient of ≥2.0 before and/or ≥3.0 after CRH administration is consistent with CD. A systematic review of all published studies on IPSS showed an overall sensitivity of 96% and specificity of 100% using these criteria.Citation31

Although IPSS is the gold-standard test to distinguish between a pituitary and ectopic source of ACTH, it is an invasive procedure requiring a high degree of skill, and thus is best performed in experienced centers. False-negative IPSS results of 1%–10% have been attributed to anomalous venous drainage, abnormal venous anatomy, lack of expertise, and technical problems.Citation97Citation99 Review of the IPSS venogram and/or prolactin measurement during IPSS can improve diagnostic accuracy and decrease false-negative results.Citation100Citation102 A baseline prolactin inferior petrosal sinus to peripheral (IPS/P) ratio (ipsilateral to the dominant post-CRH ACTH IPS/P ratio) of 1.8 or more suggests successful catheterization during IPSS. Prolactin-normalized ACTH IPS/P ratios can then be used to differentiate between a pituitary and ectopic source of ACTH. Values ≤0.7 suggest EAS, and those ≥1.3 suggest CD. Indeterminate values (0.7–1.3) need further study.Citation102 False-positive IPSS results can occur in rare cases of ectopic CRH productionCitation103,Citation104 and in patients with cyclic Cushing’s syndrome if the normal corticotropes are not completely suppressed.Citation102 Documentation of sustained hypercortisolism prior to dynamic testing therefore remains crucial.

Localization of the source of ACTH

Cushing’s disease

As discussed earlier, advanced MRI techniques often do not identify pituitary adenomas. While IPSS localizes the tumor to the pituitary gland, its role in determining the precise tumor location remains controversial. Using an intersinus ratio of 1.4 or more before or after CRH to predict the location of pituitary adenomas,Citation105 a review of 19 studies (313 cases) reported poor localizing accuracy (78% overall, 50%–100% in individual studies).Citation31 More recently, Wind et al found that IPSS correctly predicted tumor location in 273 of 396 patients (positive predictive value 69%) with surgically proven CD and a lateral adenoma.Citation87 Mulligan et al demonstrated that the use of prolactin-normalized ACTH intersinus ratios led to improvement in the lateralization accuracy of IPSS (75% versus 54%) in 28 patients with surgically proven CD.Citation106 Although these data are promising, larger prospective studies are needed. Therefore, at present IPSS lateralization results can only be used as a guide for where to begin initial transsphenoidal exploration in MRI negative cases. A thorough exploration of the pituitary gland is essential before using lateralization results for hemihypophysectomy.

Ectopic ACTH syndrome

Structural (CT and MRI) and functional (somatostatin scintigraphy and positron emission tomography [PET] scans) imaging studies are used to identify the source of ACTH in EAS, but the optimal imaging strategy has not been well defined. Since no single imaging modality identifies all tumors, correlation of multiple studies is needed.Citation24,Citation107

Because the majority of these tumors are intrathoracic, initial imaging should focus on the chest, with additional studies obtained as needed. Thin-cut multislice CT scans and 3 T MRI improve tumor detection, but also can give false-positive results.Citation107 As many NETs express somatostatin-subtype (sst) receptors, they may be detected via somatostatin scintigraphy (eg, octreotide scan). The ability of these scans to identify a tumor depends on the type (sst1–5) and degree of sst-receptor expression, size of the tumor, and the dose (6–18 mCi pentetreotide) of the radiopharmaceutical and the use of single photon emission CT.Citation108 In vitro studies have shown that glucocorticoids downregulate sst-receptor expression in human neuroendocrine cells.Citation109 This has been described in vivo, as well in two patients with ACTH-secreting pulmonary NETs, where mifepristone (a glucocorticoid receptor antagonist) treatment led to a change in the octreotide-scan status and correct localization of the tumor.Citation110 This phenomenon of improved diagnostic ability of the octreotide scan after medical control of hypercortisolism needs to be further evaluated in larger studies. GaCitation68-DOTATATE and DOTATOC, PET radiopharmaceuticals with high affinity for sst2, have shown promise in identifying gastrointestinal–pancreatic and pulmonary NETs. However, patients with ACTH-secreting tumors were not included.Citation111Citation113 The use of 68Ga-DOTATATE PET scans in identifying the source of ACTH in ectopic Cushing’s syndrome needs further investigation.

As most EAS tumors are slow growing and have low metabolic activity, fluorodeoxyglucose (18F) PET scans have limited utility in tumor localization; they may help define the extent of metastatic disease.Citation114 Radiolabeled L-3,4-dihydroxyphenylalanine and (11C) 5-hydroxytryptamine PET scans can help with localization.Citation107,Citation115,Citation116 Non-IPSS venous sampling usually does not provide any additional information, and is not considered necessary in the evaluation of ectopic Cushing’s syndrome.Citation24,Citation117

Management

Surgery

Surgical resection of the source of glucocorticoid excess (pituitary adenoma, nonpituitary tumor-secreting ACTH or adrenal tumor[s]) remains the first-line treatment of all forms of Cushing’s syndrome. The initial remission rate after transsphenoidal surgery is 60%–80% (<15% in macroadenomas),Citation118 with a relapse rate of up to 20% within 10 years.Citation1,Citation119,Citation120 However, the success rate depends on the skill and experience of the neurosurgeon, and can be as high as 90% at experienced centers. Patients with hypocortisolism in the immediate postoperative period need glucocorticoid replacement until the recovery of the hypothalamic–pituitary–adrenal axis (usually 6–18 months after surgery).Citation1 Although long-term remission is more likely when postoperative cortisol levels are less than 2 μg/dL (<54 nmol/L), no cortisol value excludes the possibility of recurrence.Citation120 These data emphasize the need for ongoing surveillance and alternative treatment modalities for CD.

As in CD, surgical removal of an ectopic ACTH-secreting tumor is the optimal treatment. However, occult or metastatic tumors require medical therapy or bilateral adrenalectomy.Citation24

Laparoscopic unilateral or bilateral adrenalectomy is the treatment of choice in adrenal causes of Cushing’s syndrome, and has an excellent prognosis in benign cases.Citation121 Bilateral adrenalectomy in ACTH-dependent Cushing’s syndrome may be used when surgery and medical therapy are unsuccessful, or based on patient preference. It leads to rapid resolution of hypercortisolemia and related morbidity.Citation122 However, after bilateral adrenalectomy, patients need lifelong glucocorticoid and mineralocorticoid replacement. Another concern with bilateral adrenalectomy in patients with CD is the development of Nelson’s syndrome (local tumor growth with mass effects and increased ACTH levels causing hyperpigmentation). Modern imaging techniques allow early detection and management of corticotrope tumor progression after bilateral adrenalectomy in these patients.Citation123 Some physicians advocate prophylactic pituitary radiotherapy to decrease the risk of development of Nelson’s syndrome.Citation124

Pituitary radiotherapy

Persistent hypercortisolemia after transsphenoidal surgery due to residual tumor can be treated with radiotherapy. Adjunctive medical control of hypercortisolemia may be needed while awaiting the effects of radiotherapy. Conventional fractionated radiotherapy is very effective, but its effects may be delayed up to 10 years, and it can be associated with long-term hypopituitarism.Citation125 Stereotactic radiosurgery is more rapidly effective, but has been associated with a relapse rate of 20%.Citation126

Medical therapy

Medical control of hypercortisolemia may be needed in occult cases, while awaiting surgery, when surgery is contraindicated or unsuccessful, and while awaiting the effect of radiation treatment. Medical treatments for hypercortisolemia include agents that inhibit steroidogenesis (ketoconazole, metyrapone, mitotane, and etomidate), modulate ACTH release (somatostatin and dopamine agonists) or block glucocorticoid action at its receptor (mifepristone) ().Citation127 A major concern with all medical therapies is the risk of overtreatment and adrenal insufficiency. Medical control of hypercortisolemia can be achieved in two ways: either by blocking cortisol production to achieve normal levels, or by blocking cortisol secretion completely along with glucocorticoid replacement (block and replace). Regardless of the strategy, all patients on medical therapy should be educated about the symptoms of adrenal insufficiency and the emergency use of glucocorticoids.

Table 3 Drugs used to treat Cushing’s syndrome: mechanism of action, dosage, and important side effects and concerns

Ketoconazole, a steroidogenesis inhibitor, has a rapid onset of action. It inhibits the first step in cortisol biosynthesis (side-chain cleavage) and to a lesser degree 11β-hydroxylase and 17,20-desmolase.Citation128 It requires an acidic environment for maximal absorption, and thus has reduced efficacy if used in combination with proton-pump inhibitors. Although used off-label, ketoconazole is usually the first-line agent for medical control of hypercortisolism in the US. However, gastrointestinal side effects, hepatocellular dysfunction, gynecomastia, and decreased libido in men may limit its use.Citation129,Citation130 The European Medicines Agency (EMA) has recently withdrawn ketoconazole from the market because of hepatic dyscrasia in patients treated for fungal infections.Citation131 A similar safety announcement from the US Food and Drug Administration (FDA) advised against its use for any fungal infection.Citation132 When used for the control of hypercortisolemia, liver-function tests need to be monitored closely. Despite the risks associated, ketoconazole remains an important drug in the physician’s armamentarium for medical control of hypercortisolism. The EMA has therefore invited national authorities with expertise in this area to make ketoconazole available for patients with Cushing’s syndrome under controlled conditions.Citation131

Metyrapone inhibits 11β-hydroxylase and has a rapid onset of action. In the US, it is primarily used as adjunctive therapy with other steroidogenesis inhibitors and with radiation therapy in CD.Citation133,Citation134 Increased deoxycorticosterone levels, due to inhibition of 11β-hydroxylase by metyrapone, can lead to salt retention and (rarely) hypertension, while increased adrenal androgens can lead to new or worsening hirsutism and acne in women. Both ketoconazole and metyrapone have been associated with an increase in ACTH secretion and loss of control of hypercortisolism over time in CD (escape phenomena).

Mitotane, another steroidogenesis inhibitor, is also adrenolytic. It has a slow-onset, long-lasting action. Mitotane increases CBG levels, leading to falsely high total cortisol levels, complicating titration of therapy. It is teratogenic, not well tolerated, and is primarily used in the treatment of adrenocortical carcinoma.Citation135 During treatment, plasma mitotane concentrations must be monitored.Citation135 Etomidate is a substituted imidazole anesthetic agent that inhibits 11β-hydroxylase. It has been used for rapid control of hypercortisolemia in hospitalized patients in an intensive care unit setting, and is the only available intravenous parenteral agent.Citation136

Recently, somatostatin (sst5 and sst2) and dopamine (D2) receptors have been identified as therapeutic targets in CD.Citation137 Pituitary-directed therapy with pasireotide (a somatostatin analog with high affinity for sst1, -2, -3, and -5) or cabergoline (a dopamine agonist) normalizes UFC in 20%–40% of patients with CD.Citation138,Citation139 This represents an off-label use for cabergoline. Pasireotide (Signifor), the first agent approved by the EMA and FDA for the treatment of patients who have failed surgery or are not surgical candidates, is most effective when UFC is less than twice the upper limit of normal. Seventy-three percent of patients treated with pasireotide develop glucose intoleranceCitation138 due to inhibition of incretin secretion, with a concomitant decrease in insulin secretion. Glucagon-like peptide-1 agonists or dipeptidyl peptidase-4 inhibitors can effectively control glucose levels in this setting.Citation140

Mifepristone (RU-486, Korlym), a glucocorticoid and progesterone-receptor antagonist, has been approved in the US for control of hyperglycemia secondary to hypercortisolism in patients with Cushing’s syndrome who have failed surgery or are not surgical candidates. It has a rapid onset of action, and can be used to treat acute complications of Cushing’s syndrome, especially cortisol-induced psychosis.Citation141,Citation142 It is important to note that cortisol and ACTH levels remain unchanged or increase with the use of mifepristone, and cannot be used to gauge treatment success or to diagnose adrenal insufficiency. Moreover, increases in ACTH and cortisol levels can lead to (worsening of) mineralocorticoid effects (hypertension, edema, hypokalemia), and women can develop endometrial thickening during prolonged treatment.Citation141,Citation142

Other agents currently under (preclinical) study for the medical control of hypercortisolemia include LCI699 (an 11β-hydroxylase inhibitor), retinoic acid, and gefitinib (tyrosine-kinase inhibitor with EGFR [epidermal growth factor receptor] as target) ().Citation143Citation145

Combination therapy with different agents may be needed to achieve normal plasma cortisol levels in patients with moderate-to-severe hypercortisolism. This can reduce drug-related adverse events if lower combined doses are effective. A small study of 17 patients showed an overall response rate of 88% after sequential addition of ketoconazole and/or cabergoline when initial pasireotide therapy was not successful.Citation146 Overall, medical treatment in Cushing’s syndrome needs to be individualized according to patient characteristics, potential side effects, and other pharmacological properties of the drugs.Citation127

Conclusion

Cushing’s syndrome, a rare disorder, is associated with significant morbidity and mortality. Clinical presentation can be broad, and establishing the diagnosis can be difficult. Early recognition and rapid control of hypercortisolemia is necessary to decrease morbidity and mortality in these patients. Surgery remains the optimal treatment in all forms of Cushing’s syndrome, but may not be curative. Individualized medical treatment and a multidisciplinary approach are needed for optimal control of hypercortisolemia and management of comorbidities. New alternative modalities of medical treatment are needed.

Disclosure

STS reports no conflicts of interest in this work. LKN has received funding from Laboratoire-HRA Pharma as part of a Cooperative Research and Development Agreement to conduct research on the antiglucocorticoid agent, mifepristone. RAF has received research grants from Novartis.

References

  • Newell-PriceJBertagnaXGrossmanABNiemanLKCushing’s syndromeLancet20063671605161716698415
  • HopkinsRLLeinungMCExogenous Cushing’s syndrome and glucocorticoid withdrawalEndocrinol Metab Clin N Am200534371384
  • LindholmJJuulSJørgensonJOIncidence and late prognosis of Cushing’s syndrome: a population-based studyJ Clin Endocrinol Metab20018611712311231987
  • EtxabeJVazquezJAMorbidity and mortality in Cushing’s disease: an epidemiological approachClin Endocrinol (Oxf)1994404794848187313
  • FeeldersRAPulgarSJKempelAPereiraAMThe burden of Cushing’s disease: clinical and health-related quality of life aspectsEur J Endocrinol201216731132622728347
  • NtaliGAsimakopoulouASiamatrasTMortality in Cushing’s syndrome: systematic analysis of a large series with prolonged follow-upEur J Endocrinol201316971572323996696
  • ColaoAPivonelloRSpieziaSPersistence of increased cardiovascular risk in patients with Cushing’s disease after five years of successful cureJ Clin Endocrinol Metab1999842664267210443657
  • FaggianoAPivonelloRSpieziaSCardiovascular risk factors and common carotid artery caliber and stiffness in patients with Cushing’s disease during active disease and 1 year after disease remissionJ Clin Endocrinol Metab2003882527253312788849
  • DekkersOMHorváth-PuhóEJørgensenJOMultisystem morbidity and mortality in Cushing’s syndrome: a cohort studyJ Clin Endocrinol Metab2013982277228423533241
  • LeibowitzGTsurAChayenSDPre-clinical Cushing’s syndrome: an unexpected frequent cause of poor glycaemic control in obese diabetic patientsClin Endocrinol (Oxf)1996447177228759185
  • CatargiBRigalleauVPoussinAOccult Cushing’s syndrome in type-2 diabetesJ Clin Endocrinol Metab2003885808581314671173
  • OmuraMSaitoJYamaguchiKKakutaYNishikawaTProspective study on the prevalence of secondary hypertension among hypertensive patients visiting a general outpatient clinic in JapanHypertens Res20042719320215080378
  • TerzoloMReimondoGChiodiniIScreening of Cushing’s syndrome in outpatients with type 2 diabetes: results of a prospective multicentric study in ItalyJ Clin Endocrinol Metab2012973467347522767639
  • MullanKBlackNThiraviarajAIs there value in routine screening for Cushing’s syndrome in patients with diabetes?J Clin Endocrinol Metab2010952262226520237165
  • ReimondoGPiaAAllasinoBScreening of Cushing’s syndrome in adult patients with newly diagnosed diabetes mellitusClin Endocrinol (Oxf)20076722522917547690
  • NewsomeSChenKHoangJWilsonJDPotterJMHickmanPECushing’s syndrome in a clinic population with diabetesIntern Med J20083817818217645502
  • GagliarduLChapmanIMLoughlinPOTorpyDJScreening for subclinical Cushing’s syndrome in type 2 diabetes mellitus: low false-positive rates with nocturnal salivary cortisolHorm Metab Res20104228028420119887
  • BaidSKRubinoDSinaiiNRamseySFrankANiemanLKSpecificity of screening tests for Cushing’s syndrome in an overweight and obese populationJ Clin Endocrinol Metab2009943857386419602562
  • NiemanLKBillerBMFindlingJWThe diagnosis of Cushing’s syndrome: an Endocrine Society Clinical Practice GuidelineJ Clin Endocrinol Metab2008931526154018334580
  • HerraraMFGrantCSvan HeerdenJASheedyPFIlstrupDMIncidentally discovered adrenal tumors: an institutional perspectiveSurgery1991110101410211745970
  • BovioSCataldiAReimondoGPrevalence of adrenal incidentaloma in a contemporary computerized tomography seriesJ Endocrinol Invest20062929830216699294
  • NiemanLKApproach to the patient with an adrenal incidentalomaJ Clin Endocrinol Metab2010954106411320823463
  • ManteroFTerzoloMArnaldiGA survey on adrenal incidentaloma in Italy. Study Group on Adrenal Tumors of the Italian Society of EndocrinologyJ Clin Endocrinol Metab20008563764410690869
  • IliasITorpyDJPacakKCushing’s syndrome due to ectopic corticotrophin secretion: twenty years experience at National Institutes of HealthJ Clin Endocrinol Metab2005904955496215914534
  • LacroixABourdeauIBilateral adrenal Cushing’s syndrome: macronodular adrenal hyperplasia and primary pigmented nodular adrenocortical diseaseEndocrinol Metab Clin N Am200534441458
  • LouisetEDuparcCYoungJIntraadrenal corticotrophin in bilateral macronodular adrenal hyperplasiaN Eng J Med201336921152125
  • LacroixAHeredity and cortisol regulation in bilateral macronodular adrenal hyperplasiaN Eng J Med201336921472149
  • StorrHLChanLFGrossmanABSavageMOPaediatric Cushing’s syndrome: epidemiology, investigation and therapeutic advancesTrends Endocrinol Metab20071816717417412607
  • Pecori GiraldiFMoroMCavagniniFGender-related differences in the presentation and course of Cushing’s diseaseJ Clin Endocrinol Metab2003881554155812679438
  • FaggianoAPivonelloRMelisDNephrolithiasis in Cushing’s disease: prevalence, etiopathogenesis, and modification after disease cureJ Clin Endocrinol Metab2003882076208012727957
  • Newell-PriceJTrainerPBesserMGrossmanAThe diagnosis and differential diagnosis of Cushing’s syndrome and pseudo-Cushing’s statesEndocr Rev1998196476729793762
  • GreeningJEStorrHLMcKenzieSALinear growth and body mass index in pediatric patients with Cushing’s disease or simple obesityJ Endocrinol Invest20062988588717185896
  • MagiakouMAMastorakosGOldfieldEHCushing’s syndrome in children and adolescents. Presentation, diagnosis, and therapyN Engl J Med19943316296368052272
  • StorrHLIsidoriAMMonsonJPBesserGMGrossmanABSavageMOPrepubertal Cushing’s disease is more common in males, but there is no increase in severity at diagnosisJ Clin Endocrinol Metab2004893818382015292311
  • TorpyDJMullenNIliasINiemanLKAssociation of hypertension and hypokalemia with Cushing’s syndrome caused by ectopic ACTH secretion: a series of 58 casesAnn N Y Acad Sci200297013414412381548
  • SarlisNJChanockSJNiemanLKCortisolemic indices predict severe infections in Cushing syndrome due to ectopic production of adrenocorticotropinJ Clin Endocrinol Metab200085424710634361
  • IsidoriAMKaltsasGAPozzaCThe ectopic adrenocorticotropin syndrome: clinical features, diagnosis, management, and long-term follow-upJ Clin Endocrinol Metab20069137137716303835
  • MinettoMReimondoGOsellaGVenturaMAngeliATerzoloMBone loss is more severe in primary adrenal than in pituitary-dependent Cushing’s syndromeOsteoporosis Int200415855861
  • OhmoriNNomuraKOhmoriKKatoYItohTTakanoKOsteoporosis is more prevalent in adrenal than in pituitary Cushing’s syndromeEndocr J2003501712733704
  • TrementinoLAppolloniGCeccoliLBone complications in patients with Cushing’s syndrome: looking for clinical, biochemical, and genetic determinantsOsteoporos Int20142591392124126765
  • ValassiESantosAYanevaMThe European Registry on Cushing’s syndrome: 2-year experience. Baseline demographic and clinical characteristicsEur J Endocrinol201116538339221715416
  • StarkmanMNSchteingartDENeuropsychiatric manifestations of patients with Cushing’s syndrome. Relationship to cortisol and adrenocorticotropic hormone levelsArch Intern Med1981412152196257194
  • HaskettRFDiagnostic categorization of psychiatric disturbance in Cushing’s syndromeAm J Psychiatry19851429119162992298
  • DornLDBurgessESDubbertBPsychopathology in patients with endogenous Cushing’s syndrome: ‘atypical’ or melancholic featuresClin Endocrinol (Oxf)1995434334427586617
  • PereiraAMTiemensmaJRomijnJANeuropsychiatric disorders in Cushing’s syndromeNeuroendocrinology201092Suppl 1657020829621
  • BourdeauIBardCNoelBLoss of brain volume in endogenous Cushing’s syndrome and its reversibility after correction of hypercortisolismJ Clin Endocrinol Metab2002871949195411994323
  • StarkmanMNGebarskiSSBerentSSchteingartDEHippocampal formation volume, memory dysfunction, and cortisol levels in patients with Cushing’s syndromeBiol Psychiatry1992327567651450290
  • MandelSSteroid myopathy. Insidious cause of muscle weaknessPostgrad Med1982722072102132157134072
  • Rebuffé-ScriveMKrotkiewskiMElfversonJBjörntorpPMuscle and adipose tissue morphology and metabolism in Cushing’s syndromeJ Clin Endocrinol Metab198867112211283142910
  • LindsayJRNanselTBaidSGumowskiJNiemanLKLong-term quality of life in Cushing’s syndrome despite initial improvement after surgical improvement after surgical remissionJ Clin Endocrinol Metab20069144745316278266
  • PikkarainenLSaneTReunsnenAThe survival and well-being of patients treated for Cushing’s syndromeJ Intern Med199924546346810363746
  • MishraAKAgarwalAGuptaSAgarwalGVermaAKMishraSKOutcome of adrenalectomy for Cushing’s syndrome: experience from a tertiary care centerWorld J Surg2007311425143217534556
  • FeeldersRAPulgarSJKempelAPereiraAMThe burden of Cushing’s disease: clinical and health-related quality of life aspectsEur J Endocrinol201216731132622728347
  • PivonelloRDe LeoMVitalePPathophysiology of diabetes mellitus in Cushing’s syndromeNeuroendocrinology201092Suppl 1778120829623
  • SharmaSTNiemanLKCushing’s syndrome: all variants, detection, and treatmentEndocrinol Metab Clin North Am201140379391viiiix21565673
  • ArnaldiGScandaliVMTrementinoLCardinalettiMAppolloniGBoscaroMPathophysiology of dyslipidemia in Cushing’s syndromeNeuroendocrinology201092Suppl 1869020829625
  • RockallAGSohaibSAEvansDHepatic steatosis in Cushing’s syndrome: a radiological assessment using computerized tomographyEur J Endocrinol200314954354814640995
  • RockallAGSohaibSAEvansDComputed tomography assessment of fat distribution in male and female patients with Cushing’s syndromeEur J Endocrinol200314956156714640998
  • van der PasRLeebeekFWHoflandLJde HerderWWFeeldersRAHypercoagulability in Cushing’s syndrome: prevalence, pathogenesis and treatmentClin Endocrinol (Oxf)20137848148823134530
  • GiordanoRPicuAMarinazzoEMetabolic and cardiovascular outcomes in patients with Cushing’s syndrome of different aetiologies during active disease and 1 year after remissionClin Endocrinol (Oxf)20117535436021521323
  • BarahonaMJResminiEViladésDCoronary artery disease detected by multislice computed tomography in patients after long-term cure of Cushing’s syndromeJ Clin Endocrinol Metab2013981093109923393183
  • GeerEBShenWStrohmayerEPostKDFredaPUBody composition and cardiovascular risk markers after remission of Cushing’s disease: a prospective study using whole-body MRIJ Clin Endocrinol Metab2012971702171122419708
  • de CastroMMoreiraACScreening and diagnosis of Cushing’s syndromeArq Bras Endocrinol Metab20075111911198
  • PetersennSNewell-PriceJFindlingJWHigh variability in baseline urinary free cortisol values in patients with Cushing’s diseaseClin Endocrinol (Oxf)20148026126923746264
  • AlexandrakiKIGrossmanABIs urinary free cortisol of value in the diagnosis of Cushing’s syndrome?Curr Opin Endocrinol Diabetes Obes20111825926321681089
  • FindlingJWRaffHAronDCThe low-dose dexamethasone suppression test: a reevaluation in patients with Cushing’s syndromeJ Clin Endocrinol Metab2004891222122615001614
  • RaffHUtility of salivary cortisol measurements in Cushing’s syndrome and adrenal insufficiencyJ Clin Endocrinol Metab2009943647365519602555
  • GafniRIPapanicolaouDANiemanLKNighttime salivary cortisol measurement as a simple, noninvasive, outpatient screening test for Cushing’s syndrome in children and adolescentsJ Pediatr2000137303510891818
  • RaffHUpdate on late-night salivary cortisol for the diagnosis of Cushing’s syndrome: methodological considerationsEndocrine20134434634923839587
  • Newell-PriceJTrainerPPerryLWassJGrossmanABesserMA single sleeping midnight cortisol has 100% sensitivity for the diagnosis of Cushing’s syndromeClin Endocrinol (Oxf)1995435455508548938
  • ReimondoGAllasinoBBovioSPaccottiPAngeliATerzoloMEvaluation of the effectiveness of midnight serum cortisol in the diagnostic procedures for Cushing’s syndromeEur J Endocrinol200515380380916322385
  • PapanicolaouDAYanovskiJACutlerGBJrChrousosGPNiemanLKA single midnight serum cortisol measurement distinguishes Cushing’s syndrome from pseudo-Cushing statesJ Clin Endocrinol Metab199883116311679543134
  • YanovskiJACutlerGBJrChrousosGPNiemanLKThe dexamethasone-suppressed corticotropin-releasing hormone stimulation test differentiates mild Cushing’s disease from normal physiologyJ Clin Endocrinol Metab1998833483529467539
  • YanovskiJACutlerGBJrChrousosGPNiemanLKCorticotropin- releasing hormone stimulation following low-dose dexamethasone administration. A new test to distinguish Cushing’s syndrome from pseudo-Cushing’s statesJAMA1993269223222388386285
  • TirabassiGFaloiaEPapaRFurlaniGBoscaroMArnaldiGUse of the desmopressin test in the differential diagnosis of pseudo-Cushing state from Cushing’s diseaseJ Clin Endocrinol Metab2010951115112220080839
  • MoroMPutignanoPLosaMInvittiCMaraschiniCCavagniniFThe desmopressin test in the differential diagnosis between Cushing’s disease and pseudo-Cushing statesJ Clin Endocrinol Metab2000853569357411061503
  • NiemanLEditorial: The dexamethasone-suppressed corticotropin-releasing hormone test for the diagnosis of Cushing’s syndrome: what we have learned in 14 yearsJ Clin Endocrinol Metab2007922876287817682087
  • ValassiESwearingenBLeeHConcomitant medication use can confound interpretation of the combined dexamethasone-corticotropin releasing hormone test in Cushing’s syndromeJ Clin Endocrinol Metab2009944851485919850679
  • AlwaniRASchmit JongbloedLWde JongFHVan der LelyAJde HerderWFeeldersRADifferentiating between Cushing’s disease and pseudo-Cushing’s syndrome: comparison of four testsEur J Endocrinol201417047748624394725
  • RaffHFindlingJWA new immunoradiometric assay for corticotropin evaluated in normal subjects and patients with Cushing’s syndromeClin Chem1989355966002539271
  • HorvathAStratakisCPrimary pigmented nodular adrenocortical disease and Cushing’s syndromeArq Bras Endocrinol Metabol2007511238124418209861
  • StratakisCASarlisNKirschnerLSParadoxical response to dexamethasone in the diagnosis of primary pigmented nodular adrenocortical diseaseAnn Intern Med199913158559110523219
  • AssiéGLibéREspiardSARMC5 mutations in macronodular adrenal hyperplasia with Cushing’s syndromeN Eng J Med201336921052114
  • AlencarGALerarioAMNishiMYARMC5 mutations are a frequent cause of primary macronodular adrenal hyperplasiaJ Clin Endocrinol Metab201499E1501E150924708098
  • FauczFRZilbermintMLodishMBMacronodular adrenal hyperplasia due to mutations in an armadillo repeat containing 5 (ARMC5) gene: a clinical and genetic investigationJ Clin Endocrinol Metab201499E1113E111924601692
  • PatronasNBulakbasiNStratakisCASpoiled gradient recalled acquisition in the steady state technique is superior to conventional postcontrast spin echo technique for magnetic resonance imaging detection of adrenocorticotropin-secreting pituitary tumorsJ Clin Endocrinol Metab2003881565156912679440
  • WindJJLonserRRNiemanLKDevroomHLChangROldfieldEHThe lateralization accuracy of inferior petrosal sinus sampling in 501 patients with Cushing’s diseaseJ Clin Endocrinol Metab2013982285229323553862
  • HallWALucianoMGDoppmanJLPatronasNJOldfieldEHPituitary magnetic resonance imaging in normal human volunteers: occult adenomas in the general populationAnn Intern Med19941208178208154641
  • DicheckALNiemanLKOldfieldEHPassHIMalleyJDCutlerGBJrA comparison of the standard high dose dexamethasone suppression test and the overnight 8-mg dexamethasone suppression test for the differential diagnosis of adrenocorticotropin-dependent Cushing’s syndromeJ Clin Endocrinol Metab1994784184228106630
  • AronDCRaffHFindlingJWEffectiveness versus efficacy: the limited value in clinical practice of high dose dexamethasone suppression testing in the differential diagnosis of adrenocorticotropin-dependent Cushing’s syndromeJ Clin Endocrinol Metab199782178017859177382
  • FindlingJWRaffHDiagnosis and differential diagnosis of Cushing’s syndromeEndocrinol Metab Clin North Am20013072974711571938
  • NiemanLKOldfieldEHWesleyRChrousosGPLoriauxDLCutlerGBJrA simplified morning ovine corticotropin-releasing hormone stimulation test for the differential diagnosis of adrenocorticotropin-dependent Cushing’s syndromeJ Clin Endocrinol Metab199377130813128077325
  • Newell-PriceJMorrisDGDrakeWMOptimal response criteria for the human CRH test in the differential diagnosis of ACTH-dependent Cushing’s syndromeJ Clin Endocrinol Metab2002871640164511932295
  • NiemanLKCutlerGBJrOldfieldEHLoriauxDLChrousosGPThe ovine corticotropin-releasing hormone (CRH) stimulation test is superior to the human CRH stimulation test for the diagnosis of Cushing’s diseaseJ Clin Endocrinol Metab1989691651692543689
  • TrainerPJFariaMNewell-PriceJA comparison of the effects of human and ovine corticotropin-releasing hormone on the pituitary-adrenal axisJ Clin Endocrinol Metab1995804124177852498
  • OldfieldEHDoppmanJLNiemanLKPetrosal sinus sampling with and without corticotropin-releasing hormone for the differential diagnosis of Cushing’s syndromeN Eng J Med1991325897905
  • SwearingenBKatznelsonLMillerKDiagnostic errors after inferior petrosal sinus samplingJ Clin Endocrinol Metab2004893752376315292301
  • DoppmanJLChangROldfieldEHChrousosGStratakisCANiemanLKThe hypoplastic inferior petrosal sinus: a potential source of false-negative results in petrosal sampling for Cushing’s diseaseJ Clin Endocrinol Metab19998453354010022412
  • LópezJBarcelóBLucasTPetrosal sinus sampling for diagnosis of Cushing’s disease: evidence of false negative resultsClin Endocrinol (Oxf)1996451471568881446
  • FindlingJWKehoeMERaffHIdentification of patients with Cushing’s disease with negative pituitary adrenocorticotropin gradients during inferior petrosal sinus sampling: prolactin as an index of pituitary venous effluentJ Clin Endocrinol Metab2004896005600915579751
  • MulliganGBErayEFaimanCReduction of false-negative results in inferior petrosal sinus sampling with simultaneous prolactin and corticotrophin measurementEndocr Pract201117334020713347
  • SharmaSTRaffHNiemanLKProlactin as a marker of successful catheterization during IPSS in patients with ACTH-dependent Cushing’s syndromeJ Clin Endocrinol Metab2011963687369422031511
  • ShahaniSNudelmanRJNaliniRKimHSSamsonSLEctopic corticotropin-releasing hormone (CRH) syndrome from metastatic small cell carcinoma: a case report and review of the literatureDiagn Pathol201055620807418
  • YoungJDeneuxCGrinoMOliverCChansonPSchaisonGPitfall of petrosal sinus sampling in a Cushing’s syndrome secondary to ectopic adrenocorticotropin-corticotropin releasing hormone (ACTH-CRH) secretionJ Clin Endocrinol Metab1998833053089467532
  • OldfieldEHChrousosGPSchulteHMPreoperative localization of ACTH-secreting pituitary microadenomas by bilateral and simultaneous inferior petrosal venous sinus samplingN Eng J Med1985312100103
  • MulliganGBFaimanCGuptaMProlactin measurement during inferior petrosal sinus sampling improves the localization of pituitary adenomas in Cushing’s diseaseClin Endocrinol (Oxf)20127726827422243215
  • ZemskovaMSGundaboluBSinaiiNUtility of various functional and anatomic imaging modalities for detection of ectopic adrenocorticotropin-secreting tumorsJ Clin Endocrinol Metab2010951207121920089611
  • KwekkeboomDJKrenningEPBakkerWHOeiHYKooijPPLambertsSWSomatostatin analogue scintigraphy in carcinoid tumoursEur J Nucl Med1993202832928491220
  • de BruinCFeelderRAWaaijersAMDifferential regulation of human dopamine D2 and somatostatin receptor subtype expression by glucocorticoids in vitroJ Mol Endocrinol200942475618852217
  • de BruinCHoflandLJNiemanLKMifepristone effects on tumor somatostatin receptor expression in two patients with Cushing’s syndrome due to ectopic adrenocorticotropin secretionJ Clin Endocrinol Metab20129745546222090282
  • GabrielMDecristoforoCKendlerD68Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and CTJ Nucl Med20074850851817401086
  • KayaniIConryBGGrovesAMA comparison of 68Ga-DOTATATE and 18F-FDG PET/CT in pulmonary neuroendocrine tumorsJ Nucl Med2009501927193219910422
  • JindalTKumarAVenkitaramanBDuttaRKumarRRole of 68Ga-DOTATOC PET/CT in the evaluation of primary pulmonary carcinoidsKorean J Intern Med20102538639121179276
  • PacakKIliasIChenCCCarrasquilloJAWhatleyMNiemanLKThe role of [(18)F]fluorodeoxyglucose positron emission tomography and [(111)In]-diethylenetriaminepentaacetate-D-Phe-pentetreotide scintigraphy in the localization of ectopic adrenocorticotropin-secreting tumors causing Cushing’s syndromeJ Clin Endocrinol Metab2004892214222115126544
  • KauhanenSSeppänenMOvaskaJThe clinical value of [18F] fluoro-dihydroxyphenylalanine positron emission tomography in primary diagnosis, staging, and restaging of neuroendocrine tumorsEndocr Relat Cancer20091625526519088184
  • OrleforsHSundinAGarskeUWhole-body (11) C-5-hydroxytryptophan positron emission tomography as a universal imaging technique for neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and computed tomographyJ Clin Endocrinol Metab2005903392340015755858
  • DoppmanJLPassHINiemanLKCorticotropin-secreting carcinoid tumors of the thymus: diagnostic unreliability of thymic vein samplingRadiology199218471741319078
  • WooYSIsidoriAMWatWZClinical and biochemical characteristics of adrenocorticotropin-secreting macroadenomasJ Clin Endocrinol Metab2005904963496915886242
  • ReesDAHannaFWDaviesJSMillsRGVafidisJScanlonMFLong-term follow-up results of transsphenoidal surgery for Cushing’s disease in a single center using strict criteria for remissionClin Endocrinol (Oxf)20025654155111966748
  • LindsayJROldfieldEHStratakisCANiemanLKThe postoperative basal cortisol and CRH tests for prediction of long-term remission from Cushing’s disease after transsphenoidal surgeryJ Clin Endocrinol Metab2011962057206421508126
  • BruntLMMoleyJFDohertyGMLairmoreTCDeBenedettiMKQuasebarthMAOutcomes analysis in patients undergoing laparoscopic adrenalectomy for hormonally active adrenal tumorsSurgery200113062963511602893
  • RitzelKBeuschleinFMickischAClinical review: outcome of bilateral adrenalectomy in Cushing’s syndrome: a systematic reviewJ Clin Endocrinol Metab201398103939394823956347
  • AssiéGBahurelHCosteJCorticotroph tumor progression after adrenalectomy in Cushing’s disease: a reappraisal of Nelson’s syndromeJ Clin Endocrinol Metab20079217217917062771
  • MehtaGUSheehanJPVanceMLEffect of stereotactic radio-surgery before bilateral adrenalectomy for Cushing’s disease on the incidence of Nelson’s syndromeJ Neurosurg20131191493149723952749
  • EstradaJBoronatMMielgoMThe long-term outcome of pituitary irradiation after unsuccessful transsphenoidal surgery in Cushing’s diseaseN Eng J Med1997336172177
  • SheehanJMVanceMLSheehanJPEllegalaDBLawsERJrRadiosurgery for Cushing’s disease after failed transsphenoidal surgeryJ Neurosurg20009373874211059652
  • FeeldersRAHoflandLJMedical treatment of Cushing’s diseaseJ Clin Endocrinol Metab20139842543823345100
  • EngelhardtDWeberMMTherapy of Cushing’s syndrome with steroid biosynthesis inhibitorsJ Steroid Biochem Mol Biol1994492612678043488
  • LewisJHZimmermanHJBensonGDIshakKGHepatic injury associated with ketoconazole therapy. Analysis of 33 casesGastroenterology1984865035136319220
  • CastinettiFGuignatLGiraudPKetoconazole in Cushing’s disease: is it worth a try?J Clin Endocrinol Metab2014991623163024471573
  • European Medicines AgencyEuropean Medicines Agency recommends suspension of marketing authorizations for oral ketoconazole throughout the European Union [press release]LondonEMA7262013 Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Press_release/2013/07/WC500146613.pdfAccessed November 13, 2014
  • Food and Drug AdministrationFDA Drug Safety Communication: FDA limits usage of Nizoral (ketoconazole) oral tablets due to potentially fatal liver injury and risk of drug interactions and adrenal gland problems [press release]Silver Spring (MD)FDA7262013 Available from: http://www.fda.gov/Drugs/DrugSafety/ucm362415.htmAccessed November 13, 2014
  • OrthDNMetyrapone is useful only as adjunctive therapy in Cushing’s diseaseAnn Intern Med197889128130208445
  • ValassiECrespoIGichIRodríguezJWebbSMA reappraisal of the medical therapy with steroidogenesis inhibitors in Cushing’s diseaseClin Endocrinol (Oxf)20127773574222533782
  • BaudryCCosteJBouKREfficiency and tolerance of mitotane in Cushing’s disease in 76 patients from a single centerEur J Endocrinol201216747348122815335
  • PredaVASenJKaravitakiNGrossmanABEtomidate in the management of hypercortisolaemia in Cushing’s syndrome: a reviewEur J Endocrinol201216713714322577107
  • de BruinCPereiraAMFeeldersRACoexpression of dopamine and somatostatin receptor subtypes in corticotroph adenomasJ Clin Endocrinol Metab2009941118112419141584
  • ColaoAPetersennSNewell-PriceJA 12-month phase 3 study of pasireotide in Cushing’s diseaseN Engl J Med201236691492422397653
  • GodboutAManavelaMDanilowiczKBeauregardHBrunoODLacroixACabergoline monotherapy in the long-term treatment of Cushing’s diseaseEur J Endocrinol201016370971620702648
  • HenryRRCiaraldiTPArmstrongDBurkePLigueros-SaylanMMudaliarSHyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteersJ Clin Endocrinol Metab2013983446345323733372
  • CastinettiFFassnachtMJohanssenSMerits and pitfalls of mifepristone in Cushing’s syndromeEur J Endocrinol20091601003101019289534
  • FleseriuMBillerNMFindlingJWMolitchMESchteingartDEMifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing’s syndromeJ Clin Endocrinol Metab2012972039204922466348
  • BertagnaXPivonelloRFleseriuMLCI699, a potent 11β-hydroxylase inhibitor, normalizes urinary cortisol in patients with Cushing’s disease: results from a multicenter, proof-of-concept studyJ Clin Endocrinol Metab2014991375138324423285
  • Pecori GiraldiFAmbrogioAGAndrioliMPotential role for retinoic acid in patients with Cushing’s diseaseJ Clin Endocrinol Metab2012973577358322851491
  • FukuokaHCooperOBen-ShlomoAEGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomasJ Clin Invest20111214712472122105169
  • FeeldersRAde BruinCPereiraAMPasireotide alone or with cabergoline and ketoconazole in Cushing’s diseaseN Eng J Med201036218461848
  • RossEJLinchDCCushing’s syndrome-killing disease: discriminatory value of signs and symptoms aiding early diagnosisLancet198226466496125785
  • YanovskiJACutlerGBJrGlucocorticoid action and the clinical features of Cushing’s syndromeEndocrinol Metab Clin North Am1994234875097805650
  • WeberATrainerPJGrossmanABInvestigation, management and therapeutic outcome in 12 cases of childhood and adolescent Cushing’s syndromeClin Endocrinol (Oxf)19954319287641408
  • SofferLIannacconeAGabriloveJCushing’s syndrome: a study of fifty patientsAmerican Journal of Medicine196130129146