You are on page 1of 16

Review

doi: 10.1111/joim.12975

Current management of Cushing’s disease


N. A. Tritos & B. M. K. Biller
From the Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA

Abstract. Tritos NA, Biller BMK (Neuroendocrine experienced surgeon. Patients who are not in
Unit, Massachusetts General Hospital and remission postoperatively or those who relapse
Harvard Medical School, Boston, MA, USA). may benefit from undergoing a second pituitary
Current management of Cushing’s disease. J operation. Alternatively, radiation therapy to the
Intern Med 2019; 286: 526–541. sella with interim medical therapy, or bilateral
adrenalectomy, can be effective as definitive treat-
Cushing’s disease (CD) is caused by a pituitary ments of CD. Medical therapy is currently adjunc-
tumour that secretes adrenocorticotropin (ACTH) tive in most patients with CD and is generally
autonomously, leading to excess cortisol secretion prescribed to patients who are about to receive
from the adrenal glands. The condition is associ- radiation therapy and will be awaiting its salutary
ated with increased morbidity and mortality that effects to occur. Available treatment options
can be mitigated by treatments that result in include steroidogenesis inhibitors, centrally acting
sustained endocrine remission. Transsphenoidal agents and glucocorticoid receptor antagonists.
pituitary surgery (TSS) remains the mainstay of Several novel agents are in clinical trials and may
treatment for CD but requires considerable neuro- eventually constitute additional treatment options
surgical expertise and experience in order to opti- for this serious condition.
mize patient outcomes. Up to 90% of patients with
microadenomas (tumour below 1 cm in largest Keywords: Cushing’s disease, medical therapy, pitu-
diameter) and 65% of patients with macroadeno- itary adenoma, radiotherapy, transsphenoidal
mas (tumour at or above 1 cm in greatest diameter) pituitary surgery.
achieve endocrine remission after TSS by an

importance of prompt diagnosis and timely man-


Introduction
agement of CD [3, 8, 9].
Cushing’s disease (CD) is caused by a pituitary
tumour, generally benign (adenoma) but rarely The goals of treatment in patients with CD are to
malignant (carcinoma), which secretes adrenocor- abolish cortisol excess and restore normal cortisol
ticotropin (ACTH) autonomously, leading to secretion, maintain normal pituitary function and
hypercortisolism (Figure 1) [1–3]. It accounts for achieve tumour control. At present, transsphe-
approximately 70% of patients with endogenous noidal pituitary surgery (TSS), performed by an
Cushing’s syndrome (CS); the ectopic ACTH syn- experienced pituitary neurosurgeon, is the treat-
drome and adrenal tumours or hyperplasia ment of choice for the vast majority of patients with
account for 10% and 20% of patients with CS, CD and comes closest to fulfilling the above-men-
respectively [3]. Of note, CD is estimated to occur tioned goals [8–12]. Radiation therapy (RT) to the
in 1.2-1.8 persons per million population per sella with interim medical therapy and bilateral
year and is associated with increased morbidity adrenalectomy constitute additional treatment
and mortality, primarily as a consequence of options and are generally second- and third-line
increased risk of cardiovascular disease and therapies as outlined in Figure 2 [8, 11].
secondarily infection [4–6]. The diagnosis and
differential diagnosis of CD are complex and have The aims of the present manuscript include dis-
been the subject of Endocrine Society guidelines cussing the role, outcomes, risks and benefits of
[3] and a recent review article [7]. Patients who diverse treatment options currently available for
achieve endocrine remission generally have a patients with CD and highlighting emerging ther-
more favourable prognosis, underscoring the apies. To identify pertinent articles from the

526 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

Fig. 1 Coronal T1-weighted, gadolinium-enhanced, magnetic resonance imaging (MRI) of a 35-year-old woman, obtained
before (left panel) and 12 months after (right panel) transsphenoidal pituitary surgery (TSS). This patient had presented
with weight gain in a central distribution, oligomenorrhoea, acne and proximal myopathy of recent onset. Laboratory tests
showed elevated 24-h urine free cortisol and late-night salivary cortisol on several occasions (3–4 times above the upper
limit of normal). A pituitary MRI examination showed a 5 mm by 4 mm right-sided sellar hypodensity, consistent with
microadenoma (left panel). Additional tests were consistent with Cushing’s disease. She underwent TSS, leading to
resection of a pituitary adenoma (positive for ACTH on immunohistochemistry), which was followed by clinical and
endocrine remission. A follow-up MRI examination (right panel) showed no evidence of a sellar mass.

primary literature, electronic searches were con- (such as fluoroscopy and MRI), operating micro-
ducted using the keywords ‘Cushing’s syndrome’, scopes, endoscopes and intraoperative neuronavi-
‘Cushing’s disease’, ‘treatment’, ‘transsphenoidal gation were not available. It is the advent of these
pituitary surgery’, ‘radiation therapy’, ‘medical technological advances that have made the
therapy’, ‘bilateral adrenalectomy’. Articles were transsphenoidal route increasingly safer for pitu-
included in the present bibliography at the itary surgery, thus leading to its widespread adop-
authors’ discretion. tion amongst neurosurgeons [13, 14].

The large majority (90%) of pituitary adenomas


Pituitary surgery
causing CD are microadenomas, measuring less
Transsphenoidal pituitary surgery is currently than 10 mm in largest diameter, and are detectable
first-line treatment for almost all patients with on MRI only in ~60% of patients [3, 7]. Here lies one
CD, the exception being the occasional patients of the challenges for the pituitary neurosurgeon,
with contraindications to surgery, such as the who may often have to meticulously examine the
requirement for uninterrupted anticoagulation pituitary gland for evidence of an adenoma by
therapy, or those rare patients who decline sur- obtaining intraoperative biopsies from suspicious
gery. Patients whose condition is not immediately areas for cytological and frozen section pathologi-
stable enough for TSS, such as those with active cal analysis [15], if the tumour has not been
sinusitis or psychosis, can temporize with medical visualized preoperatively. It is therefore not sur-
therapy until they are well enough to undergo prising that the outcome of TSS is significantly
surgery (as will be subsequently discussed). influenced by the experience and expertise of the
pituitary neurosurgeon [16]. In expert hands,
Although Harvey Cushing had initially used the postoperative remission of hypercortisolism, usu-
transsphenoidal route for pituitary surgery, he ally defined as very low morning serum cortisol
later abandoned it in favour of craniotomy, recog- levels in the early postoperative period [17, 18] [and
nizing the technical difficulties associated with the in some centres, supported by very low 24-hour
former approach [1, 2]. At that time, however, high- urine free cortisol (UFC) and late-night salivary
resolution cross-sectional pituitary imaging [in- cortisol (LNSC) during that time [19, 20]], is
cluding magnetic resonance imaging (MRI)], mod- achieved in up to ~90% of patients with microade-
ern surgical instruments, intraoperative imaging nomas and ~65% of patients with macroadenomas

ª 2019 The Association for the Publication of the Journal of Internal Medicine 527
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

Fig. 2 Management of patients with Cushing’s disease.

(Table 1) [9, 18, 21–64]. Other predictors of post- supraphysiologic doses of glucocorticoids in order
operative remission include preoperative or intra- to alleviate glucocorticoid withdrawal symptoms.
operative tumour visualization and pathological Once shown to have recovered normal function of
confirmation of the diagnosis [65–67]. Use of the the hypothalamic–pituitary–adrenal axis and no
endoscope (versus operating microscope) is not longer need glucocorticoid replacement, patients
associated with higher remission rates in patients with CD require lifelong periodic surveillance to
with CD who have microadenomas but may be detect recurrence of hypercortisolism. Recurrent
associated with higher likelihood of remission in CD occurs at a rate of approximately 2% of patients
those with macroadenomas [68]. per year and peaks in the first 5 years after tumour
resection, but may also develop a decade or longer
Experienced pituitary neurosurgeons perform TSS after endocrine remission was achieved, the cumu-
with very low mortality (approaching zero in some lative risk approaching 30% on long-term follow-up
case series) and low morbidity [9, 14, 16]. However, [69]. Patients with macroadenomas and those
complications may still occur, including epistaxis requiring glucocorticoid replacement for shorter
(0.4-6.0%), tumour bed haemorrhage (1-6%), cere- intervals (less than 12 months) after TSS are at
brospinal fluid leak (up to 8%), meningitis (up to higher risk for recurrence [21, 66, 70]. Patients
3%) and venous thromboembolism (up to 4%) [16]. with ‘normal’ rather than low morning serum
Endocrine complications include transient or per- cortisol levels in the early postoperative period are
sistent central diabetes insipidus (3-9%), hypona- also at higher risk of recurrence [29].
traemia (10-25%, often caused by the syndrome of
the inappropriate antidiuretic hormone secretion) Patients with persistent or recurrent CD after
and anterior hypopituitarism (2-40%) [16]. initial TSS may undergo a second surgical proce-
dure by an experienced pituitary neurosurgeon
Patients who are in endocrine remission after TSS (Figure 2). Of note, endocrine remission is achieved
require glucocorticoid replacement until their pitu- in 55–70% of patients with CD after repeat TSS and
itary–adrenal axis recovers, which generally takes the risk of surgical complications appears to be
many months. Some patients initially require higher [71, 72]. Other treatment options for

528 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

Table 1. Outcomes of pituitary surgery in patients with Cushing’s disease

Number of Follow-up (mean,


Study (first author, year) patients median or range, months) Remission rate (%) Recurrence rate (%)
Pikkarainen et al. (1999) [22] 43 89 83.7 41.7
Swearingen et al. (1999) [9] 154 104 87 7.5
Kurosaki et al. (2000) [23] 51 6-36 94.1 4.2
Vallette-Kasic et al. (2000) [24] 53 139 81.1 11.6
Barbetta et al. (2001) [25] 68 58 89.7 21.3
Chee et al. (2001) [26] 61 88 78.7 14.6
Imaki et al. (2001) [27] 49 88 95.9 14.9
Shimon et al. (2002) [28] 74 50 78.4 5.2
Yap et al. (2002) [29] 97 92 62.9 11.5
Chen et al. (2003) [30] 174 NA 81.6 15.5
Flitsch et al. (2003) [31] 147 61 93.2 5.8
Hammer et al. (2004) [32] 227 133 83.2 NA
Mortini et al. (2005) [33] 262 NA 77.5 5.9
Esposito et al. (2006) [18] 40 34 77.5 3.2
Frank et al. (2006) [34] 56 54 67.8 NA
Shah et al. (2006) [35] 65 NA 61.5 12.5
Acebes et al. (2007) [36] 44 49 88.6 7.7
Rollin et al. (2007) [37] 103 72 85.4 4.5
Hofmann et al. (2008) [38] 426 72 65.7 15
Jehle et al. (2008) [39] 193 62 80.8 13.5
Patil et al. (2008) [40] 215 46 85.6 17.4
Jagannathan et al. (2009) [41] 483 84 93.8 1.3
Alwani et al. (2010) [42] 79 84 64.6 19.6
Ammini et al. (2011) [43] 81 18-132 66.7 18.5
Ciric et al. (2012) [44] 121 0-396 83.5 8.9
Honegger et al. (2012) [45] 83 38 84.3 7.1
Alexandraki et al. (2013) [46] 124 185 67.7 23.8
Barbot et al. (2013) [47] 57 77 68.4 38.4
Berker et al. (2013) [48] 69 32 95.6 6
Burkhardt et al. (2013) [49] 42 NA 95.2 NA
Starke et al. (2013) [50] 61 28 95.1 8.6
Wagenmakers et al. (2013) [51] 86 71 72.1 16.1
Huan et al. (2014) [52] 84 46 70.2 28.8
Potts et al. (2014) [53] 91 12 58.2 NA
Kuo et al. (2015) [54] 40 41 80 9.4
Chandler et al. (2016) [56] 275 80 79.6 16.9
Sarkar et al. (2016) [58] 64 20 73.4 8.5
Shirvani et al. (2016) [59] 96 44 94.8 23.1
Witek et al. (2016) [60] 40 29 80 NA
Cebula et al. (2017) [61] 230 22 79.1 9.9
Donofrio et al. (2017) [57] 142 NA 80.3 NA

ª 2019 The Association for the Publication of the Journal of Internal Medicine 529
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

Table 1 (Continued )

Number of Follow-up (mean,


Study (first author, year) patients median or range, months) Remission rate (%) Recurrence rate (%)
Masopust et al. (2017) [62] 41 6-34 87.8 NA
Shin et al. (2017) [55] 50 37 80 22.5
Feng et al. (2018) [63] 341 12-36 78.9 2.4
Saini et al. (2019) [64] 60 40 72 18

NA, not available.


Studies include case series of ≥ 40 patients, published between 1999 and 2019, and are listed chronologically.

patients with persistent or recurrent CD will be medical therapy before RT to ensure that hyper-
subsequently discussed and include radiation cortisolism can be medically controlled before RT is
therapy to the sella, generally administered in administered [8].
conjunction with medical therapy and bilateral
adrenalectomy. Adverse effects of RT include anterior hypopitu-
itarism (affecting 20-60% of patients at 5 years and
~85% of patients at 15 years after RT), optic
Radiation therapy
neuropathy (1–2%) and other cranial neuropathies
Radiation therapy to the sella is often considered to (1%) [75, 90, 91]. Rare complications include the
be second-line treatment in patients who have development of secondary tumours (including
either failed TSS (i.e. those with persistent or meningiomas or sarcomas), stroke and temporal
recurrent hypercortisolism postoperatively) or, lobe necrosis, all of which have been observed in
rarely, those who are not candidates for pituitary patients who underwent conventional fractionated
surgery [73]. RT [92]. Whilst it is anticipated that stereotactic RT
may be safer with regard to the risk of these rare
Radiation therapy can be delivered as conventional complications, this remains to be demonstrated.
fractionated photon-based RT, typically adminis-
tered 5 days a week over 5–6 weeks, to a total dose
Medical therapy
of 45–54 Gy [73]. However, RT is increasingly being
administered as one of several stereotactic treat- The role of medical therapy is generally adjunctive
ment modalities, including photon-based (includ- in the management of CD. Medical therapy is
ing Gamma knifeTM or linear accelerator-based, currently not considered as definitive treatment
such as ‘CyberknifeTM’) or proton beam RT options. [8]. Most commonly, medications are used to
Using computer-assisted techniques and precise, control hypercortisolism in patients who have
positioning frames or helmets, stereotactic RT is failed TSS and have opted for RT as definitive
accurately focused on the treatment target, whilst treatment. In these patients, it is advisable to begin
minimizing radiation exposure of normal tissues medical therapy before RT is administered in order
[74]. It can be delivered in a single fraction to assure the effectiveness of medical strategy in
(‘radiosurgery’) to patients with smaller tumours controlling hypercortisolism whilst awaiting the
that are distant from the optic apparatus at a salutary effects of RT [8]. In some cases, medical
(margin) dose ranging between 12 and 35 Gy [73]. therapy is administered to patients who have
Based on historical comparisons, it appears that severe manifestations of CD and/or severe hyper-
radiosurgery may lead to endocrine remission cortisolism in anticipation of definitive therapy (e.g.
sooner than conventional fractionated radiation patients with sepsis, sinusitis or psychosis, who
therapy [75]. However, regardless of the treatment will be undergoing TSS as soon as they are med-
modality, RT leads to tumour control in 83–100% ically stable). In a minority of cases, medical
of patients (Table 2) [76–90]. In contrast, endocrine therapy is used in patients whose tumour location
remission occurs in 28–84% of treated patients is uncertain or those who decline surgery, as may
after a variable time interval, which may extend to rarely be the case.
many years. These patients require medical ther-
apy to control hypercortisolism until the salutary There are currently three groups of medications
effects of RT occur. It is advisable to initiate used in patients with CD (Table 3), which are

530 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

Table 2. Outcomes of radiation therapy in patients with Cushing’s disease

Follow-up
(mean or Endocrine Endocrine Tumour
Number of median, Treatment (Margin) remission recurrence control
Study (first author, year) patients months) modality dose (Gy) rate (%) rate (%) (%)
Sheehan et al. (2000) [77] 43 39.1 Gamma knife 20 63 6.9 100
Jane et al. (2003) [78] 45 NA Gamma knife 15 73 4 NA
Devin et al. (2004) [79] 35 7.5 Linear 14.7 49 11 91
accelerator
Minniti et al. (2007) [76] 40 108 Fractionated 45 84 0 93
conventional
RT
Jagannathan et al. (2007) [80] 90 41.3 Gamma knife 23 54 20 95
Castinetti et al. (2007) [81] 40 94 Gamma knife 29.5 42.5 NA NA
Petit et al. (2008) [82] 33 62 Proton beam 20 55 0 NA
Aghi et al. (2008) [84] 31 43 Proton beam 20 58 3 NA
Wan et al. (2009) [83] 68 67.3 Gamma knife 21.9 27.9 NA 89.7
Losa et al. (2010) [85] 49 48 Gamma knife 25 53 NA NA
Sheehan et al. (2011) [86] 82 31 Gamma knife 24 54 NA NA
Sheehan et al. (2013) [87] 96 48 Gamma knife 22 78 15.6 98
Wattson et al. (2014) [88] 79 47 Proton beam 20 67 1 99
Wilson et al. (2014) [89] 36 66 Linear 20 36.1 3 83
accelerator
Mehta et al. (2017) [90] 278 67 Gamma knife 23.7 80 18 NA

NA, not available; RT, radiation therapy.


Studies include case series of ≥ 30 patients, published between 1999 and 2019, and are listed chronologically.

conveniently categorized depending on their site of is the only agent available for intravenous use and
action, including steroidogenesis inhibitors (in- has been administered ‘off label’ to control serious
hibiting adrenal steroidogenesis), centrally acting hypercortisolism rapidly in severely ill patients.
agents (controlling excessive ACTH secretion) and Several other agents are at different stages of
glucocorticoid receptor (GR) antagonists (inhibiting development, as will be discussed.
cortisol action) [93].
Steroidogenesis inhibitors are generally titrated
towards normalizing 24-h urine free cortisol
Steroidogenesis inhibitors
(UFC) levels; however, escape from their salutary
Steroidogenesis inhibitors act on one or several effects may occur over time. In addition, hypoad-
enzymes involved in adrenal steroidogenesis to renalism may result as an extension of their
inhibit cortisol synthesis [8]. Ketoconazole and pharmacologic action, which is treatable with
metyrapone are currently used for this purpose, temporary interruption of the medication and/or
though they are prescribed ‘off label’ in the treat- dose reduction. As an alternative, a ‘block and
ment of CD in the United States. Both agents are replace’ strategy can be employed, whereby
licensed by the European Medicines Agency (EMA) endogenous glucocorticoid secretion is completely
for the treatment of hypercortisolism. Mitotane is blocked with a steroidogenesis inhibitor, whilst the
primarily administered to patients with adrenocor- patient receives glucocorticoid replacement. This
tical carcinoma but has been also used to treat CD latter strategy can be particularly helpful in
in some countries; it is seldom prescribed to patients with cyclic (intermittent) CD. However,
patients with CD in the United States. Etomidate particular caution is required if a ‘block and

ª 2019 The Association for the Publication of the Journal of Internal Medicine 531
Journal of Internal Medicine, 2019, 286; 526–541
532
Table 3. Currently available medical therapies for patients with Cushing’s disease

Mechanism Effectiveness
Agent of action Typical dose range (% cases) Adverse effects Comments
Ketoconazole Steroidogenesis 200-600 mg po bid-tid 49%a (escape Nausea, headache, skin rash, Regular monitoring of liver
inhibitor may occur) pruritus, transaminitis, liver failure chemistries is advised;
potential for drug–drug
interactions; gastric acid
required for absorption
Metyrapone Steroidogenesis 250-1000 mg po qid 76%b/ 43%a Nausea, dizziness, hypertension, Successfully used during

Journal of Internal Medicine, 2019, 286; 526–541


inhibitor (escape may oedema, hypokalaemia, hirsutism, pregnancy
occur) acne
Mitotane Steroidogenesis 0.5-3.0 g po tid Up to 85%a Gastrointestinal, genitourinary, Used primarily in
inhibitor haematologic, metabolic, adrenocortical carcinoma;
neurologic, ophthalmic and skin delayed onset of action;
toxicities teratogenic

ª 2019 The Association for the Publication of the Journal of Internal Medicine
1 b
Etomidate Steroidogenesis 0.03 mg kg iv (bolus), Up to 100% Sedation, vomiting, myoclonus, Monitoring in intensive care
inhibitor followed by (short term) dystonia unit is advised
1 1
0.1–0.3 mg kg h
Cabergoline Centrally acting 0.5–7.0 mg po weekly 30–40%a Nausea, vomiting, orthostasis, Effective in doses that are
(escape may headache, vivid dreams, psychiatric generally higher than those
occur) manifestations used in hyperprolactinaemia
Pasireotide Centrally acting 0.3-0.9 mg sc bid 21%a Diarrhoea, dyspepsia, nausea, Glucose monitoring is advised
cholelithiasis,
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

hyperglycaemia/diabetes mellitus,
hypoadrenalism, bradycardia
Pasireotide LAR Centrally acting 10–30 mg im 40%a Same as pasireotide Same as pasireotide
every 4 weeks
Mifepristone Glucocorticoid 300-1200 mg po daily 60%c/ 38%d/ Hypoadrenalism, hypokalaemia, Dose titration based on clinical
receptor 87%e elevated blood pressure, end-points only; potential for
antagonist endometrial thickening/vaginal drug–drug interactions;
bleeding abortifacient

bid: twice daily; im: intramuscularly; LAR: long-acting release; po: by mouth; qid: four times daily; sc: subcutaneously; tid: three times daily; iv:
intravenously.
a
Based on normalization of 24-h urine free cortisol. bBased on normalization of serum cortisol. cBased on improvement in glycaemia. dBased on
improvement in blood pressure. eBased on improvement in global clinical status.
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

replace’ regimen is used in order to avoid hyper- normalization in 43% of cases and control of serum
cortisolism as a consequence of inadequate sup- cortisol levels in 76% of treated patients [99]. As a
pression of endogenous cortisol secretion and/or result of the enzymatic blockade, metyrapone
overreplacement with exogenous glucocorticoid. administration leads to increased ACTH secretion
by the pituitary when given to patients with CD. It
Ketoconazole is an imidazole with antifungal prop- also produces an accumulation of steroid precur-
erties that inhibits several enzymes involved in sors, some of which have mineralocorticoid or
adrenal steroidogenesis, thus inhibiting cortisol androgenic activity, thus accounting for several of
synthesis. Ketoconazole administration has been its adverse effects, including hypertension, oedema
reported to result in 24-h UFC normalization in up and hypokalaemia, or hirsutism and acne, respec-
to 75% of patients [94, 95]. In a more recent study tively. Blood pressure, serum potassium and
of 200 patients, ketoconazole therapy led to 24-h testosterone (the latter in women) levels ought to
UFC normalization in 49% of patients [96]. There be monitored in treated patients. Additional possi-
were improvements in blood pressure, potassium ble adverse events include nausea, dyspepsia and
and glucose levels in patients receiving ketocona- dizziness. Metyrapone has been used to control
zole. Its oral absorption requires gastric acid. hypercortisolism during pregnancy [100, 101];
Consequently, oral bioavailability is lower in however, it is not specifically labelled for use
patients with hypochlorhydria or those receiving during gestation.
proton pump inhibitors or H2 receptor antagonists.
Adverse effects associated with ketoconazole ther- Mitotane (o, p’ – DDD) is chemically related to an
apy include dyspepsia, transaminitis, headache, older insecticide and inhibits several enzymes
pruritus and skin rash. Hypogonadism and gynae- involved in adrenal steroidogenesis. It is also
comastia may occur in men as a result of inhibition adrenolytic in higher doses administered long term
of testosterone biosynthesis. Ketoconazole may and may lead to permanent hypoadrenalism. Mito-
interfere with the masculinization of a male foetus tane has a slow onset of action as a consequence of
and should be avoided during pregnancy. its gradual accumulation over several weeks.
Hydrocortisone replacement is advisable in patients
Asymptomatic transaminitis may occur in about treated with mitotane; higher than usual glucocor-
18% of patients and is generally reversible with ticoid replacement doses are recommended since
ketoconazole dose reduction but may require treat- the drug leads to elevated cortisol-binding globulin
ment discontinuation (if severe or symptomatic). Of (CBG) levels and accelerates glucocorticoid clear-
particular concern, fulminant hepatic failure has ance [102]. Mitotane is primarily used in patients
been reported and has estimated to occur in about with adrenocortical carcinoma [103, 104]. In addi-
1 out of 15 000 treated patients [97]. The Food and tion, it has been administered to patients with CD in
Drug Administration (FDA) has inserted a ‘black some countries. In this latter context, it can control
box warning’ into the ketoconazole label, informing hypercortisolism in up to ~85% of patients [105,
prescribers of the risk of severe hepatotoxicity and 106]. Mitotane therapy has been associated with
recommending regular monitoring of liver enzymes gastrointestinal, hepatic, genitourinary, haemato-
in patients treated with ketoconazole. This medi- logic, metabolic, skin, ophthalmic and neurologic
cation has substantial potential for drug–drug toxicities. It is extensively stored in adipose tissue
interactions with agents metabolized by the cyto- and is a potent teratogen. Owing to its prolonged
chrome P450 3A4 system, necessitating caution in half-life, it can take several years for the drug to
reviewing all the medications that patients receive completely dissipate after its discontinuation. As a
in order to avoid significant interactions. corollary, it is advisable to defer pregnancy for up to
5 years after the medication is stopped.
Metyrapone primarily inhibits 11 beta-hydroxy-
lase, the terminal enzyme involved in cortisol Etomidate is a parenterally administered anaes-
synthesis. It is rapidly effective after oral adminis- thetic agent, which additionally inhibits 11 beta-
tration, which is helpful when prompt control of hydroxylase, even in subhypnotic doses. It has been
hypercortisolism is especially desirable in an out- used to control severe, life-threatening hypercorti-
patient setting. It has been reported to control solism and is particularly helpful as ‘bridge therapy’
hypercortisolism in up to 75% of patients [98]. In a to a definitive intervention [107, 108]. Careful
recent study of 195 patients with CS (including 115 monitoring is required to avoid excessive sedation
with CD), metyrapone therapy led to 24-h UFC in patients on etomidate therapy. Additional

ª 2019 The Association for the Publication of the Journal of Internal Medicine 533
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

adverse events associated with etomidate use in cortisol secretion. These agents may also afford
include vomiting, dystonia and myoclonus. tumour control in some cases. Cabergoline, pasir-
eotide and pasireotide long-acting release (LAR) are
Combination therapy has been reported in case currently the only agents available for use in CD; of
series, including ketoconazole plus metyrapone these, cabergoline is prescribed ‘off label’ to
with or without mitotane [109]. Such combinations patients with CD. There are also several drugs in
can be particularly helpful in patients with severe development as will be subsequently detailed.
hypercortisolism.
Cabergoline is a dopamine agonist with relative
Several agents are in development, including osilo- selectivity for the D2 receptor isoform [118]. This
drostat, levoketoconazole, ATR-101 and abi- agent is licensed for use in patients with hyper-
raterone acetate, all of which act as prolactinaemia and prolactin-secreting adenomas.
steroidogenesis inhibitors. Osilodrostat inhibits Its effectiveness in CD is predicated by the pres-
11 beta-hydroxylase and aldosterone synthase. ence of dopamine receptors on tumorous corti-
Osilodrostat administration led to 24-h UFC nor- cotrophs in most tumours. Cabergoline
malization in approximately 80% of patients in administration leads to 24-h UFC normalization
phase II extension studies [110]. Phase 3 studies of in approximately 30-40% of patients with CD [119–
osilodrostat are either in progress or completed 122]. However, escape from its beneficial effects
(but the results have not been published yet). may occur over time [122]. Of note, the effective
Hypoadrenalism, headache, nausea, diarrhoea, dose range in patients with CD (1.0–
dizziness, mineralocorticoid (oedema, hyperten- 7.0 mg week 1) is generally higher than the typical
sion, hypokalaemia) and hyperandrogenic (hir- dose range prescribed to patients with hyperpro-
sutism, acne) adverse events have been reported. lactinaemia (0.5–2.0 mg week 1). Common
Levoketoconazole is an enantiomer (2S, 4R) of adverse events associated with cabergoline use
ketoconazole that is more potent in inhibiting include nausea, vomiting and orthostatic dizzi-
several steroidogenic enzymes than its enantiomer ness, which often improve over time and can be
(2R, 4S) [111, 112]. Based on theoretical consider- minimized (in some cases) by taking the medication
ations, it has been proposed that levoketoconazole at bedtime with a snack. Other possible adverse
may be less likely to result in hepatotoxicity than events include headache, nasal congestion, consti-
ketoconazole. A phase 3 study of levoketoconazole pation, digital vasospasm, vivid dreams, night-
has been completed but the results have not been mares, anxiety and depression. It is advisable to
published yet. ATR-101 inhibits acyl coenzyme A: avoid cabergoline administration in patients with
cholesterol acyltransferase 1 (ACAT1) and is being psychosis and use caution when prescribing it to
studied as potential therapy in CD [113]. Abi- patients with depression. Impulse control disor-
raterone acetate inhibits 17 alpha-hydroxylase and ders have been reported in some hyperprolacti-
17, 20 lyase. It has been shown to have antitumour naemic patients treated with cabergoline [123–
activity in patients with disseminated prostate 126]. In addition, high-dose cabergoline therapy
carcinoma [114, 115]. A case report of a patient used to treat Parkinson’s disease has been associ-
with adrenocortical carcinoma who showed a bio- ated with cardiac valvulopathy [127, 128]. This
chemical response to abiraterone acetate has been phenomenon is likely a consequence of serotonin
published [116]. A study of abiraterone acetate in (5HT2B) receptor activation in the endocardium,
adrenocortical carcinoma is under way; however, leading to valvular fibrosis. However, most studies
there are currently no published data on the suggest that cabergoline therapy (at least in typical
efficacy of abiraterone acetate in CD. Other agents doses used to treat hyperprolactinaemia) does not
in early stages of development include a mono- appear to be associated with significant cardiac
clonal antibody (ALD1613) that neutralizes ACTH valvulopathy [129].
and peptides acting as antagonists of the melano-
cortin 2 receptor, which aim at inhibiting ACTH Pasireotide and pasireotide LAR activate multiple
action [111, 117]. somatostatin receptor (SSTR) isoforms, including
SSTR1, SSTR2, SSTR3 and SSTR5. It is thought
that their effectiveness is dependent on activation
Centrally acting agents
of SSTR2 and SSTR5, which are expressed in the
Centrally acting agents inhibit ACTH secretion majority of corticotroph adenomas. Pasireotide
from tumorous corticotrophs, leading to a decrease therapy led to 24-hr UFC normalization in

534 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

approximately 21% of 162 patients with CD [130, increased recycling of several signalling molecules,
131]. Pasireotide may also control nadir cortisol including the EGFR, to the plasma membrane. In
levels, as measured by late-night salivary cortisol turn, EGFR has been shown to stimulate ACTH
[132]. In another study of 150 patients with CD, secretion. Whether gefitinib is particularly effective
pasireotide LAR administration normalized 24-h in tumours harbouring USP8 gene mutations
UFC in about 40% of treated patients [133]. remains to be established. A phase 2 study of
Improvement in clinical status, including body gefitinib in patients with CD and somatic USP8
weight, blood pressure and lipids, has been mutations is under way. Retinoic acid inhibits
reported in patients treated with these agents. A ACTH secretion and tumour growth in preclinical
decrease in tumour size may also occur in response studies [139]. In small studies, isotretinoin, a 13
to pasireotide therapy [131]. Both agents have been cis retinoic acid isomer, controlled hypercorti-
approved by the FDA and the EMA for use in solism in about 25% of patients [140, 141]. R-
patients with CD who have hypercortisolism per- roscovitine inhibits cyclin-dependent kinase 2 and
sisting or recurring after TSS or are not candidates cyclin E and has decreased ACTH secretion and
for pituitary surgery. Both pasireotide and pasireo- tumour growth in animal studies [142, 143]. A
tide LAR share several adverse effects with first- phase 2 study of r-roscovitine in CD is in progress.
generation somatostatin receptor agonists (oc- Silibinin binds to heat shock protein 90, leading to
treotide and lanreotide), including diarrhoea, dys- its dissociation from the GR [144]. Both in vitro and
pepsia, nausea, cholelithiasis and transaminitis. animal studies are promising [144], but pertinent
Hypoadrenalism, hair loss, vitamin B12 deficiency data in humans are currently not available.
and asymptomatic sinus bradycardia may also
develop. Hyperglycaemia and diabetes mellitus
Glucocorticoid receptor antagonists
occur frequently amongst patients treated with
pasireotide or pasireotide LAR (70–80% of cases). Glucocorticoid receptor antagonists inhibit cortisol
In these patients, insulin secretion is inhibited actions mediated through its cognate receptor but
both directly and indirectly (via inhibition of have no effects on mineralocorticoid receptor (MR)
incretin peptides that are expressed in the gas- activation by cortisol. Mifepristone is currently the
trointestinal tract and normally stimulate food- only agent available for use in CD [145]. Mifepri-
dependent insulin secretion) [134]. Metformin, stone is approved by the FDA and the EMA for use
incretin-based therapies and insulin have been in hyperglycaemic patients with CS (regardless of
proposed as treatments of hyperglycaemia and aetiology) who have failed surgery or are not
diabetes mellitus in patients treated with pasireo- surgical candidates. In a study of 50 patients with
tide or pasireotide LAR. CS, most of whom had CD, mifepristone therapy
led to an improvement in glycaemia amongst 60%
Combination therapy can be effective in patients of 29 patients who had diabetes mellitus or hyper-
with severe hypercortisolism, including pasireotide glycaemia at study entry, and a significant
in combination with cabergoline and ketoconazole decrease in blood pressure amongst 38% of 21
[135], but more data are needed to fully establish patients with hypertension at baseline [146]. An
the role, risks and benefits of such therapy. improvement in global clinical status was observed
in 87% of subjects [146]. Cortisol and ACTH levels
Several agents that are centrally acting to suppress significantly increase in patients with CD under-
ACTH secretion or others that aim at decreasing going mifepristone therapy in the absence of clear-
ACTH action are in development, including gefi- cut evidence of tumour progression [147]. There-
tinib, retinoic acid, silibinin and r-roscovitine. fore, monitoring of serum cortisol, 24-h UFC and
Gefitinib is a tyrosine kinase inhibitor that pre- LNSC levels is of no therapeutic value in treated
vents signalling mediated through the epidermal patients, who require close clinical monitoring to
growth factor receptor (EGFR) and is being studied establish effectiveness and detect the possible
as a potential therapy in CD [136]. Recent studies emergence of hypoadrenalism. Of note, patients
have identified the presence of somatic, activating who develop adrenal insufficiency on mifepristone
mutations of the ubiquitin-specific protease 8 therapy do not have low serum cortisol levels; the
(USP8) gene in 30-60% of corticotroph adenomas diagnosis must be made clinically. Symptomatic
[137, 138]. These mutations lead to increased patients with hypoadrenalism may require at least
deubiquitination of proteins targeted for protea- temporary interruption of treatment and can be
some-mediated degradation, thus allowing for rescued by dexamethasone administration in

ª 2019 The Association for the Publication of the Journal of Internal Medicine 535
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

higher doses over several days. Adverse effects BA also leads to permanent primary adrenal insuf-
associated with mifepristone therapy include those ficiency and the consequent requirement for lifelong
arising from unopposed activation of the MR by glucocorticoid and mineralocorticoid replacement.
endogenous cortisol, including hypertension and In addition, BA has been associated with the sub-
hypokalaemia. Potassium levels require careful sequent development of corticotroph tumour pro-
monitoring in treated patients, who may require gression (in about 50% of patients undergoing BA),
large doses of potassium supplementation and/or which may progress to Nelson’s syndrome (in 8-20%
spironolactone in order to maintain normokalae- of patients undergoing BA) [154, 155]. In these
mia. Mifepristone also serves as a progesterone cases, tumour growth may cause mass effect and
receptor antagonist and may therefore cause progressive elevation in ACTH levels, leading to skin
endometrial thickening, metrorrhagia and will ter- hyperpigmentation [12, 155]. Pituitary surgery and
minate pregnancy [146]. However, precancerous RT are treatment options in patients who develop
endometria hyperplasia has not been reported in Nelson’s syndrome [151, 156, 157]. In addition,
patients on mifepristone therapy. Fatigue, head- pasireotide LAR and cabergoline have been reported
ache, nausea and reversible abnormalities in to decrease plasma ACTH levels and tumour size in
serum lipids and thyrotropin levels have been also isolated cases with Nelson’s syndrome [158, 159].
reported [146]. Mifepristone inhibits several hep-
atic enzymes, accounting for its significant poten-
Summary
tial for drug–drug interactions with other agents
metabolized through the same enzymatic path- The management of patients with CD can be
ways, so it is important to review all medications complex and often requires careful coordination
taken by patients before using mifepristone. between experts from neurosurgery, endocrinol-
ogy, radiation oncology and neuro-oncology. Whilst
Relacorilant is an investigational GR antagonist TSS is the mainstay of treatment for CD, radiation
that does not interact with the progesterone recep- therapy with interim medical therapy and bilateral
tor and is therefore anticipated to lack adverse adrenalectomy are important treatment options in
effects on the endometrium [148]. Relacorilant is patients with persistent or recurrent CD after TSS.
currently being studied as a potential therapy in The prognosis of patients with CD has improved
CS/CD. In a phase 2 study of 32 patients with CS over time and is more favourable in patients who
(including CD), improvements in blood pressure achieve remission [7–9, 160]. However, some stud-
and glucose levels were reported [148, 149]. A ies suggest that the mortality risk of patients with
phase 3 study of relacorilant is in progress. CD remains elevated above that of the general
population, even if they are in remission [6, 161]. It
is anticipated that further progress in our under-
Bilateral adrenalectomy
standing of the molecular pathogenesis of corti-
Bilateral adrenalectomy (BA) is currently consid- cotroph pituitary tumours may culminate in the
ered as a treatment option for patients who have development of novel, more efficacious therapies
had persistent or recurrent hypercortisolism after for this potentially serious condition.
TSS. Of note, BA leads to rapid control of hyper-
cortisolism and can be particularly helpful in
Conflict of interest statement
patients with severe, symptomatic hypercorti-
solism who do not respond adequately to medical NAT has received institution-directed research
therapy. It should be also noted that BA may be support from Novartis and Ipsen. BMKB has
elected upon by patients eager for prompt resolu- received institution-directed research support from
tion of hypercortisolism or those who are con- Millendo Therapeutics, Novartis and Strongbridge
cerned about potential adverse events associated Biopharma and served as an occasional consultant
with RT, including women who are planning to to Novartis and Strongbridge Biopharma.
conceive.

Currently, BA is almost always performed laparo-


References
scopically with low morbidity [150–153]. It leads to
permanent resolution of hypercortisolism in the 1 Cushing H. The basophil adenomas of the pituitary body and
vast majority of cases, the exception being the rare their clinical manifestations (pituitary basophilism). Bull
patients with ectopic adrenal rest tissue. However, Johns Hopkins Hosp 1932; 50: 137–95.

536 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

2 Cushing H. The basophil adenomas of the pituitary body. 21 Blevins LS Jr, Christy JH, Khajavi M, Tindall GT. Outcomes
Ann R Coll Surg Engl 1969; 44: 180–1. of therapy for Cushing’s disease due to adrenocorticotropin-
3 Nieman LK, Biller BM, Findling JW et al. The diagnosis of secreting pituitary macroadenomas. J Clin Endocrinol Metab
Cushing’s syndrome: an Endocrine Society Clinical Practice 1998; 83: 63–7.
Guideline. J Clin Endocrinol Metab 2008; 93: 1526–40. 22 Pikkarainen L, Sane T, Reunanen A. The survival and well-
4 Lindholm J, Juul S, Jorgensen JO et al. Incidence and late being of patients treated for Cushing’s syndrome. J Intern
prognosis of Cushing’s syndrome: a population-based study. Med 1999; 245: 463–8.
J Clin Endocrinol Metab 2001; 86: 117–23. 23 Kurosaki M, Luedecke DK, Knappe UJ, Flitsch J, Saeger W.
5 Tjornstrand A, Gunnarsson K, Evert M et al. The incidence The value of intraoperative cytology during transsphenoidal
rate of pituitary adenomas in western Sweden for the period surgery for ACTH-secreting microadenoma. Acta Neurochir
2001-2011. Eur J Endocrinol 2014; 171: 519–26. (Wien) 2000; 142: 865–70.
6 Ragnarsson O, Olsson DS, Papakokkinou E et al. Overall 24 Vallette-Kasic S, Dufour H, Mugnier M et al. Markers of
and disease-specific mortality in patients with Cushing tumor invasion are major predictive factors for the long-term
disease: A Swedish nationwide study. J Clin Endocrinol outcome of corticotroph microadenomas treated by
Metab 2019; 104: 2375–84. transsphenoidal adenomectomy. Eur J Endocrinol 2000;
7 Lacroix A, Feelders RA, Stratakis CA, Nieman LK. Cushing’s 143: 761–8.
syndrome. Lancet 2015; 386: 913–27. 25 Barbetta L, Dall’Asta C, Tomei G, Locatelli M, Giovanelli M,
8 Nieman LK, Biller BM, Findling JW et al. Treatment of Ambrosi B. Assessment of cure and recurrence after pitu-
Cushing’s syndrome: an endocrine society clinical practice itary surgery for Cushing’s disease. Acta Neurochir (Wien)
guideline. J Clin Endocrinol Metab 2015; 100: 2807–31. 2001; 143: 477–81; discussion 81-2.
9 Swearingen B, Biller BM, Barker FG 2nd et al. Long-term 26 Chee GH, Mathias DB, James RA, Kendall-Taylor P.
mortality after transsphenoidal surgery for Cushing disease. Transsphenoidal pituitary surgery in Cushing’s disease:
Ann Intern Med 1999; 130: 821–4. can we predict outcome? Clin Endocrinol (Oxf) 2001; 54:
10 Swearingen B, Barker FG 2nd, Zervas NT. The management 617–26.
of pituitary adenomas: the MGH experience. Clin Neurosurg 27 Imaki T, Tsushima T, Hizuka N et al. Postoperative plasma
1999; 45: 48–56. cortisol levels predict long-term outcome in patients with
11 Tritos NA, Biller BM, Swearingen B. Management of Cushing Cushing’s disease and determine which patients should be
disease. Nat Rev Endocrinol 2011; 7: 279–89. treated with pituitary irradiation after surgery. Endocr J
12 Tritos NA, Schaefer PW, Stein TD. Case records of the 2001; 48: 53–62.
Massachusetts General Hospital. Case 40-2011. A 52-year- 28 Shimon I, Ram Z, Cohen ZR, Hadani M. Transsphenoidal
old man with weakness, infections, and enlarged adrenal surgery for Cushing’s disease: endocrinological follow-up
glands. N Engl J Med 2011; 365: 2520–30. monitoring of 82 patients. Neurosurgery 2002; 51: 57–61;
13 Sheth SA, Bourne SK, Tritos NA, Swearingen B. Neurosur- discussion -2.
gical treatment of Cushing disease. Neurosurg Clin N Am 29 Yap LB, Turner HE, Adams CB, Wass JA. Undetectable
2012; 23: 639–51. postoperative cortisol does not always predict long-term
14 Swearingen B. Update on pituitary surgery. J Clin Endocrinol remission in Cushing’s disease: a single centre audit. Clin
Metab 2012; 97: 1073–81. Endocrinol (Oxf) 2002; 56: 25–31.
15 Qiao N, Swearingen B, Hedley-Whyte ET, Tritos NA. The 30 Chen JC, Amar AP, Choi S, Singer P, Couldwell WT, Weiss
utility of intraoperative cytological smear and frozen section MH. Transsphenoidal microsurgical treatment of Cushing
in the surgical management of patients with cushing’s disease: postoperative assessment of surgical efficacy by
disease due to pituitary microadenomas. Endocr Pathol application of an overnight low-dose dexamethasone sup-
2019; 30: 180–88. pression test. J Neurosurg 2003; 98: 967–73.
16 Barker FG 2nd, Klibanski A, Swearingen B. Transsphe- 31 Flitsch J, Knappe UJ, Ludecke DK. The use of postoperative
noidal surgery for pituitary tumors in the United States, ACTH levels as a marker for successful transsphenoidal
1996-2000: mortality, morbidity, and the effects of hospi- microsurgery in Cushing’s disease. Zentralbl Neurochir
tal and surgeon volume. J Clin Endocrinol Metab 2003; 88: 2003; 64: 6–11.
4709–19. 32 Hammer GD, Tyrrell JB, Lamborn KR et al. Transsphenoidal
17 Trainer PJ, Lawrie HS, Verhelst J et al. Transsphenoidal microsurgery for Cushing’s disease: initial outcome and
resection in Cushing’s disease: undetectable serum cortisol long-term results. J Clin Endocrinol Metab 2004; 89: 6348–
as the definition of successful treatment. Clin Endocrinol 57.
(Oxf) 1993; 38: 73–8. 33 Mortini P, Losa M, Barzaghi R, Boari N, Giovanelli M. Results
18 Esposito F, Dusick JR, Cohan P et al. Clinical review: Early of transsphenoidal surgery in a large series of patients with
morning cortisol levels as a predictor of remission after pituitary adenoma. Neurosurgery 2005; 56: 1222–33; dis-
transsphenoidal surgery for Cushing’s disease. J Clin cussion 33.
Endocrinol Metab 2006; 91: 7–13. 34 Frank G, Pasquini E, Farneti G et al. The endoscopic versus
19 Amlashi FG, Swearingen B, Faje AT et al. Accuracy of late- the traditional approach in pituitary surgery. Neuroen-
night salivary cortisol in evaluating postoperative remission docrinology 2006; 83: 240–8.
and recurrence in cushing’s disease. J Clin Endocrinol Metab 35 Shah NS, Goel AH, Nagpal RD, Menon PS. Cushing’s
2015; 100: 3770–7. disease: management outcome in a tertiary care centre. J
20 Valassi E, Biller BM, Swearingen B et al. Delayed remission Assoc Physicians India 2006; 54: 919–22.
after transsphenoidal surgery in patients with Cushing’s 36 Acebes JJ, Martino J, Masuet C, Montanya E, Soler J. Early
disease. J Clin Endocrinol Metab 2010; 95: 601–10. post-operative ACTH and cortisol as predictors of remission

ª 2019 The Association for the Publication of the Journal of Internal Medicine 537
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

in Cushing’s disease. Acta Neurochir (Wien) 2007; 149: 471– resonance imaging in the preoperative evaluation of Cush-
7; discussion 7-9. ing’s disease. J Neurooncol 2014; 116: 593–600.
37 Rollin G, Ferreira NP, Czepielewski MA. Prospective evalu- 54 Kuo CH, Yen YS, Wu JC, Chen YC, Huang WC, Cheng H.
ation of transsphenoidal pituitary surgery in 108 patients Primary endoscopic transnasal transsphenoidal surgery for
with Cushing’s disease. Arq Bras Endocrinol Metabol 2007; magnetic resonance image-positive Cushing disease: Out-
51: 1355–61. comes of a Series over 14 Years. World Neurosurg 2015; 84:
38 Hofmann BM, Hlavac M, Martinez R, Buchfelder M, Muller 772–9.
OA, Fahlbusch R. Long-term results after microsurgery for 55 Shin SS, Gardner PA, Ng J et al. Endoscopic endonasal
Cushing disease: experience with 426 primary operations approach for adrenocorticotropic hormone-secreting pitu-
over 35 years. J Neurosurg 2008; 108: 9–18. itary adenomas: outcomes and analysis of remission rates
39 Jehle S, Walsh JE, Freda PU, Post KD. Selective use of and tumor biochemical activity with respect to tumor inva-
bilateral inferior petrosal sinus sampling in patients with siveness. World Neurosurg 2017; 102: 651–8.e1.
adrenocorticotropin-dependent Cushing’s syndrome prior to 56 Chandler WF, Barkan AL, Hollon T et al. Outcome of
transsphenoidal surgery. J Clin Endocrinol Metab 2008; 93: transsphenoidal surgery for cushing disease: a single-center
4624–32. experience over 32 years. Neurosurgery 2016; 78: 216–23.
40 Patil CG, Veeravagu A, Prevedello DM, Katznelson L, Vance 57 Donofrio CA, Losa M, Gemma M, Giudice L, Barzaghi LR,
ML, Laws Jr ER. Outcomes after repeat transsphenoidal Mortini P. Safety of transsphenoidal microsurgical approach
surgery for recurrent Cushing’s disease. Neurosurgery 2008; in patients with an ACTH-secreting pituitary adenoma.
63: 266–70; discussion 70-1. Endocrine 2017; 58: 303–11.
41 Jagannathan J, Smith R, DeVroom HL et al. Outcome of 58 Sarkar S, Rajaratnam S, Chacko G, Mani S, Hesargatta AS,
using the histological pseudocapsule as a surgical capsule in Chacko AG. Pure endoscopic transsphenoidal surgery for
Cushing disease. J Neurosurg 2009; 111: 531–9. functional pituitary adenomas: outcomes with Cushing’s
42 Alwani RA, de Herder WW, van Aken MO et al. Biochemical disease. Acta Neurochir (Wien) 2016; 158: 77–86; discus-
predictors of outcome of pituitary surgery for Cushing’s sion.
disease. Neuroendocrinology 2010; 91: 169–78. 59 Shirvani M, Motiei-Langroudi R, Sadeghian H. Outcome of
43 Ammini AC, Bhattacharya S, Sahoo JP et al. Cushing’s microscopic transsphenoidal surgery in cushing disease:
disease: results of treatment and factors affecting outcome. a case series of 96 patients. World Neurosurg 2016; 87:
Hormones (Athens) 2011; 10: 222–9. 170–5.
44 Ciric I, Zhao JC, Du H et al. Transsphenoidal surgery for 60 Witek P, Zielinski G, Szamotulska K, Witek J, Kaminski G.
Cushing disease: experience with 136 patients. Neuro- Cushing‘s disease: Fibrinogen and D-dimer levels fail to
surgery 2012; 70: 70–80; discussion -1. normalize despite early postoperative remission - a prospec-
45 Honegger J, Schmalisch K, Beuschlein F et al. Contempo- tive, controlled study. Endokrynol Pol 2016; 67: 283–91.
rary microsurgical concept for the treatment of Cushing’s 61 Cebula H, Baussart B, Villa C et al. Efficacy of endoscopic
disease: endocrine outcome in 83 consecutive patients. Clin endonasal transsphenoidal surgery for Cushing’s disease in
Endocrinol (Oxf) 2012; 76: 560–7. 230 patients with positive and negative MRI. Acta Neurochir
46 Alexandraki KI, Kaltsas GA, Isidori AM et al. Long-term (Wien) 2017; 159: 1227–36.
remission and recurrence rates in Cushing’s disease: pre- 62 Masopust V, Netuka D, Benes V et al. Magnetic resonance
dictive factors in a single-centre study. Eur J Endocrinol imaging and histology correlation in Cushing’s disease.
2013; 168: 639–48. Neurol Neurochir Pol 2017; 51: 45–52.
47 Barbot M, Albiger N, Koutroumpi S et al. Predicting late 63 Feng M, Liu Z, Liu X et al. Diagnosis and outcomes of 341
recurrence in surgically treated patients with Cushing’s patients with Cushing’s Disease following transsphenoid
disease. Clin Endocrinol (Oxf) 2013; 79: 394–401. surgery: a single-center experience. World Neurosurg 2018;
48 Berker M, Isikay I, Berker D, Bayraktar M, Gurlek A. Early 109: e75–80.
promising results for the endoscopic surgical treatment of 64 Saini S, Kanwar J, Gupta S, Behari S, Bhatia E, Yadav S.
Cushing’s disease. Neurosurg Rev 2014; 37: 105–14. Long-term outcome of trans-sphenoidal surgery for Cush-
49 Burkhardt T, Schmidt NO, Vettorazzi E, Aberle J, Mengel M, ing’s disease in Indian patients. Acta Neurochir (Wien) 2019;
Flitsch J. DHEA(S)–a novel marker in Cushing’s disease. 161: 119–27.
Acta Neurochir (Wien) 2013; 155: 479–84; discussion 84. 65 Invitti C, Pecori Giraldi F, de Martin M, Cavagnini F.
50 Starke RM, Reames DL, Chen CJ, Laws ER, Jane Jr JA. Diagnosis and management of Cushing’s syndrome: results
Endoscopic transsphenoidal surgery for cushing disease: of an Italian multicentre study. Study group of the Italian
techniques, outcomes, and predictors of remission. Neuro- society of endocrinology on the pathophysiology of the
surgery 2013; 72: 240–7; discussion 7. hypothalamic-pituitary-adrenal axis. J Clin Endocrinol
51 Wagenmakers MA, Boogaarts HD, Roerink SH et al. Endo- Metab 1999; 84: 440–8.
scopic transsphenoidal pituitary surgery: a good and safe 66 Bochicchio D, Losa M, Buchfelder M. Factors influencing the
primary treatment option for Cushing’s disease, even in case immediate and late outcome of Cushing’s disease treated by
of macroadenomas or invasive adenomas. Eur J Endocrinol transsphenoidal surgery: a retrospective study by the Euro-
2013; 169: 329–37. pean Cushing’s Disease Survey Group. J Clin Endocrinol
52 Huan C, Lu C, Xu GM, Qu X, Qu YM. Retrospective analysis Metab 1995; 80: 3114–20.
of cushing’s disease with or without hyperprolactinemia. Int 67 Rees DA, Hanna FW, Davies JS, Mills RG, Vafidis J, Scanlon
J Endocrinol 2014; 2014: 919704. MF. Long-term follow-up results of transsphenoidal surgery
53 Potts MB, Shah JK, Molinaro AM et al. Cavernous and for Cushing’s disease in a single centre using strict criteria
inferior petrosal sinus sampling and dynamic magnetic for remission. Clin Endocrinol (Oxf) 2002; 56: 541–51.

538 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

68 Broersen LHA, Biermasz NR, van Furth WR et al. Endo- 86 Sheehan JP, Pouratian N, Steiner L, Laws ER, Vance ML.
scopic vs. microscopic transsphenoidal surgery for Cush- Gamma Knife surgery for pituitary adenomas: factors related
ing’s disease: a systematic review and meta-analysis. to radiological and endocrine outcomes. J Neurosurg 2011;
Pituitary 2018; 21: 524–34. 114: 303–9.
69 Roelfsema F, Biermasz NR, Pereira AM. Clinical factors 87 Sheehan JP, Xu Z, Salvetti DJ, Schmitt PJ, Vance ML.
involved in the recurrence of pituitary adenomas after Results of gamma knife surgery for Cushing’s disease. J
surgical remission: a structured review and meta-analysis. Neurosurg 2013; 119: 1486–92.
Pituitary 2012; 15: 71–83. 88 Wattson DA, Tanguturi SK, Spiegel DY et al. Outcomes of
70 Estrada J, Garcia-Uria J, Lamas C et al. The complete proton therapy for patients with functional pituitary adeno-
normalization of the adrenocortical function as the criterion mas. Int J Radiat Oncol Biol Phys 2014; 90: 532–9.
of cure after transsphenoidal surgery for Cushing’s disease. 89 Wilson PJ, Williams JR, Smee RI. Cushing’s disease: a single
J Clin Endocrinol Metab 2001; 86: 5695–9. centre’s experience using the linear accelerator (LINAC) for
71 Patil CG, Prevedello DM, Lad SP et al. Late recurrences of stereotactic radiosurgery and fractionated stereotactic radio-
Cushing’s disease after initial successful transsphenoidal therapy. J Clin Neurosci 2014; 21: 100–6.
surgery. J Clin Endocrinol Metab 2008; 93: 358–62. 90 Mehta GU, Ding D, Patibandla MR et al. Stereotactic radio-
72 Ram Z, Nieman LK, Cutler GB Jr, Chrousos GP, Doppman surgery for cushing disease: results of an international,
JL, Oldfield EH. Early repeat surgery for persistent Cush- multicenter study. J Clin Endocrinol Metab 2017; 102: 4284–
ing’s disease. J Neurosurg 1994; 80: 37–45. 91.
73 Loeffler JS, Shih HA. Radiation therapy in the management 91 Brada M, Rajan B, Traish D et al. The long-term efficacy
of pituitary adenomas. J Clin Endocrinol Metab 2011; 96: of conservative surgery and radiotherapy in the control
1992–2003. of pituitary adenomas. Clin Endocrinol (Oxf) 1993; 38:
74 Pashtan I, Oh KS, Loeffler JS. Radiation therapy in the 571–8.
management of pituitary adenomas. Handb Clin Neurol 92 Brada M, Burchell L, Ashley S, Traish D. The incidence of
2014; 124: 317–24. cerebrovascular accidents in patients with pituitary ade-
75 Sheehan JP, Niranjan A, Sheehan JM et al. Stereotactic noma. Int J Radiat Oncol Biol Phys 1999; 45: 693–8.
radiosurgery for pituitary adenomas: an intermediate review 93 Broersen LHA, Jha M, Biermasz NR, Pereira AM, Dekkers
of its safety, efficacy, and role in the neurosurgical treatment OM. Effectiveness of medical treatment for Cushing’s syn-
armamentarium. J Neurosurg 2005; 102: 678–91. drome: a systematic review and meta-analysis. Pituitary
76 Minniti G, Osti M, Jaffrain-Rea ML, Esposito V, Cantore G, 2018; 21: 631–41.
Maurizi Enrici R. Long-term follow-up results of postopera- 94 Sonino N, Boscaro M, Paoletta A, Mantero F, Ziliotto D.
tive radiation therapy for Cushing’s disease. J Neurooncol Ketoconazole treatment in Cushing’s syndrome: experience
2007; 84: 79–84. in 34 patients. Clin Endocrinol (Oxf) 1991; 35: 347–52.
77 Sheehan JM, Vance ML, Sheehan JP, Ellegala DB, Laws ER 95 Tabarin A, Navarranne A, Guerin J, Corcuff JB, Parneix M,
Jr. Radiosurgery for Cushing’s disease after failed Roger P. Use of ketoconazole in the treatment of Cushing’s
transsphenoidal surgery. J Neurosurg 2000; 93: 738–42. disease and ectopic ACTH syndrome. Clin Endocrinol (Oxf)
78 Jane JA Jr, Vance ML, Woodburn CJ, Laws ER Jr. Stereo- 1991; 34: 63–9.
tactic radiosurgery for hypersecreting pituitary tumors: part 96 Castinetti F, Guignat L, Giraud P et al. Ketoconazole in
of a multimodality approach. Neurosurg Focus 2003; 14: Cushing’s disease: is it worth a try? J Clin Endocrinol Metab
e12. 2014; 99: 1623–30.
79 Devin JK, Allen GS, Cmelak AJ, Duggan DM, Blevins LS. The 97 McCance DR, Ritchie CM, Sheridan B, Atkinson AB. Acute
efficacy of linear accelerator radiosurgery in the manage- hypoadrenalism and hepatotoxicity after treatment with
ment of patients with Cushing’s disease. Stereotact Funct ketoconazole. Lancet 1987; 1: 573.
Neurosurg 2004; 82: 254–62. 98 Verhelst JA, Trainer PJ, Howlett TA et al. Short and long-
80 Jagannathan J, Sheehan JP, Pouratian N, Laws ER, Steiner term responses to metyrapone in the medical management of
L, Vance ML. Gamma Knife surgery for Cushing’s disease. J 91 patients with Cushing’s syndrome. Clin Endocrinol (Oxf)
Neurosurg 2007; 106: 980–7. 1991; 35: 169–78.
81 Castinetti F, Nagai M, Dufour H et al. Gamma knife radio- 99 Daniel E, Aylwin S, Mustafa O et al. Effectiveness of
surgery is a successful adjunctive treatment in Cushing’s metyrapone in treating Cushing’s Syndrome: a retrospective
disease. Eur J Endocrinol 2007; 156: 91–8. multicenter study in 195 patients. J Clin Endocrinol Metab
82 Petit JH, Biller BM, Yock TI et al. Proton stereotactic 2015; 100: 4146–54.
radiotherapy for persistent adrenocorticotropin-producing 100 Gormley MJ, Hadden DR, Kennedy TL, Montgomery DA,
adenomas. J Clin Endocrinol Metab 2008; 93: 393–9. Murnaghan GA, Sheridan B. Cushing’s syndrome in preg-
83 Wan H, Chihiro O, Yuan S. MASEP gamma knife radio- nancy–treatment with metyrapone. Clin Endocrinol (Oxf)
surgery for secretory pituitary adenomas: experience in 347 1982; 16: 283–93.
consecutive cases. J Exp Clin Cancer Res 2009; 28: 36. 101 Lindsay JR, Jonklaas J, Oldfield EH, Nieman LK. Cushing’s
84 Aghi MK, Petit J, Chapman P et al. Management of recurrent syndrome during pregnancy: personal experience and
and refractory Cushing’s disease with reoperation and/or review of the literature. J Clin Endocrinol Metab 2005; 90:
proton beam radiosurgery. Clin Neurosurg 2008; 55: 141–4. 3077–83.
85 Losa M, Picozzi P, Redaelli MG, Laurenzi A, Mortini P. 102 Robinson BG, Hales IB, Henniker AJ et al. The effect of o, p’-
Pituitary radiotherapy for Cushing’s disease. Neuroen- DDD on adrenal steroid replacement therapy requirements.
docrinology 2010; 92(Suppl 1): 107–10. Clin Endocrinol (Oxf) 1987; 27: 437–44.

ª 2019 The Association for the Publication of the Journal of Internal Medicine 539
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

103 Terzolo M, Angeli A, Fassnacht M et al. Adjuvant mitotane long-term treatment of Cushing’s disease. Eur J Endocrinol
treatment for adrenocortical carcinoma. N Engl J Med 2007; 2010; 163: 709–16.
356: 2372–80. 121 Petrossians P, Thonnard AS, Beckers A. Medical treatment
104 Mauclere-Denost S, Leboulleux S, Borget I et al. High-dose in Cushing’s syndrome: dopamine agonists and cabergoline.
mitotane strategy in adrenocortical carcinoma: prospective Neuroendocrinology 2010; 92(Suppl 1): 116–19.
analysis of plasma mitotane measurement during the first 122 Pivonello R, De Martino MC, Cappabianca P et al. The
3 months of follow-up. Eur J Endocrinol 2012; 166: 261–8. medical treatment of Cushing’s disease: effectiveness of
105 Luton JP, Mahoudeau JA, Bouchard P et al. Treatment of chronic treatment with the dopamine agonist cabergoline in
Cushing’s disease by O, p’DDD. Survey of 62 cases. N Engl J patients unsuccessfully treated by surgery. J Clin Endocrinol
Med 1979; 300: 459–64. Metab 2009; 94: 223–30.
106 Baudry C, Coste J, Bou Khalil R et al. Efficiency and 123 Bancos I, Nannenga MR, Bostwick JM, Silber MH, Erickson
tolerance of mitotane in Cushing’s disease in 76 patients D, Nippoldt TB. Impulse control disorders in patients with
from a single center. Eur J Endocrinol 2012; 167: 473–81. dopamine agonist-treated prolactinomas and nonfunction-
107 Preda VA, Sen J, Karavitaki N, Grossman AB. Etomidate in ing pituitary adenomas: a case-control study. Clin Endocri-
the management of hypercortisolaemia in Cushing’s syn- nol (Oxf) 2014; 80: 863–8.
drome: a review. Eur J Endocrinol 2012; 167: 137–43. 124 Bancos I, Nippoldt TB, Erickson D. Hypersexuality in men
108 Carroll TB, Peppard WJ, Herrmann DJ et al. Continuous with prolactinomas treated with dopamine agonists. Endo-
etomidate infusion for the management of severe Cushing crine 2017; 56: 456–7.
syndrome: validation of a standard protocol. J Endocr Soc 125 Barake M, Evins AE, Stoeckel L et al. Investigation of
2019; 3: 1–12. impulsivity in patients on dopamine agonist therapy for
109 Kamenicky P, Droumaguet C, Salenave S et al. Mitotane, hyperprolactinemia: a pilot study. Pituitary 2014; 17: 150–
metyrapone, and ketoconazole combination therapy as an 6.
alternative to rescue adrenalectomy for severe ACTH-depen- 126 Barake M, Klibanski A, Tritos NA. Management of endocrine
dent Cushing’s syndrome. J Clin Endocrinol Metab 2011; 96: disease: Impulse control disorders in patients with hyper-
2796–804. polactinemia treated with dopamine agonists: how much
110 Fleseriu M, Pivonello R, Young J et al. Osilodrostat, a potent should we worry? Eur J Endocrinol 2018; 179: R287–96.
oral 11beta-hydroxylase inhibitor: 22-week, prospective, 127 Schade R, Andersohn F, Suissa S, Haverkamp W, Garbe E.
Phase II study in Cushing’s disease. Pituitary 2016; 19: Dopamine agonists and the risk of cardiac-valve regurgita-
138–48. tion. N Engl J Med 2007; 356: 29–38.
111 Cuevas-Ramos D, Lim DST, Fleseriu M. Update on medical 128 Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli
treatment for Cushing’s disease. Clin Diabetes Endocrinol G. Valvular heart disease and the use of dopamine agonists
2016; 2: 16. for Parkinson’s disease. N Engl J Med 2007; 356: 39–46.
112 Auchus RJ, Wu Y, Liu J. 2S, 4R-ketoconazole is the relevant 129 Valassi E, Klibanski A, Biller BM. Clinical Review: Potential
enantiomer of ketoconazole for cortisol synthesis inhibition: cardiac valve effects of dopamine agonists in hyperpro-
steroidogenic P450 inhibition involves multiple mecha- lactinemia. J Clin Endocrinol Metab 2010; 95: 1025–33.
nisms. Endocr Rev 2018; 39. 130 Boscaro M, Bertherat J, Findling J et al. Extended treatment
113 Langlois DK, Fritz MC, Schall WD et al. ATR-101, a selective of Cushing’s disease with pasireotide: results from a 2-year,
ACAT1 inhibitor, decreases ACTH-stimulated cortisol con- Phase II study. Pituitary 2014; 17: 320–6.
centrations in dogs with naturally occurring Cushing’s 131 Colao A, Petersenn S, Newell-Price J et al. A 12-month
syndrome. BMC Endocr Disord 2018; 18: 24. phase 3 study of pasireotide in Cushing’s disease. N Engl J
114 Berruti A, Pia A, Terzolo M. Abiraterone and increased Med 2012; 366: 914–24.
survival in metastatic prostate cancer. N Engl J Med 2011; 132 Findling JW, Fleseriu M, Newell-Price J et al. Late-night
365: 766; author reply 7-8. salivary cortisol may be valuable for assessing treatment
115 Gartrell BA, Saad F. Abiraterone in the management of response in patients with Cushing’s disease: 12-month,
castration-resistant prostate cancer prior to chemotherapy. Phase III pasireotide study. Endocrine 2016; 54: 516–23.
Ther Adv Urol 2015; 7: 194–202. 133 Lacroix A, Gu F, Gallardo W et al. Efficacy and safety of
116 Claps M, Lazzari B, Grisanti S et al. Management of severe once-monthly pasireotide in Cushing’s disease: a 12 month
Cushing syndrome induced by adrenocortical carcinoma clinical trial. Lancet Diabetes Endocrinol 2018; 6: 17–26.
with abiraterone acetate: a case report. AACE Clinical Case 134 Henry RR, Ciaraldi TP, Armstrong D, Burke P, Ligueros-
Reports 2016; 2: e337–41. Saylan M, Mudaliar S. Hyperglycemia associated with
117 Feldhaus AL, Anderson K, Dutzar B et al. ALD1613, a novel pasireotide: results from a mechanistic study in healthy
long-acting monoclonal antibody to control ACTH-driven volunteers. J Clin Endocrinol Metab 2013; 98: 3446–53.
pharmacology. Endocrinology 2017; 158: 1–8. 135 Feelders RA, de Bruin C, Pereira AM et al. Pasireotide alone
118 Klibanski A. Clinical practice. Prolactinomas. N Engl J Med or with cabergoline and ketoconazole in Cushing’s disease.
2010; 362: 1219–26. N Engl J Med 2010; 362: 1846–8.
119 Burman P, Eden-Engstrom B, Ekman B, Karlsson FA, 136 Fukuoka H, Cooper O, Ben-Shlomo A et al. EGFR as a
Schwarcz E, Wahlberg J. Limited value of cabergoline in therapeutic target for human, canine, and mouse ACTH-
Cushing’s disease: a prospective study of a 6-week secreting pituitary adenomas. J Clin Invest 2011; 121:
treatment in 20 patients. Eur J Endocrinol 2016; 174: 4712–21.
17–24. 137 Reincke M, Sbiera S, Hayakawa A et al. Mutations in the
120 Godbout A, Manavela M, Danilowicz K, Beauregard H, deubiquitinase gene USP8 cause Cushing’s disease. Nat
Bruno OD, Lacroix A. Cabergoline monotherapy in the Genet 2015; 47: 31–8.

540 ª 2019 The Association for the Publication of the Journal of Internal Medicine
Journal of Internal Medicine, 2019, 286; 526–541
Treatment of Cushing’s disease / N. A. Tritos & B. M. K. Biller

138 Theodoropoulou M, Reincke M, Fassnacht M, Komada M. 151 Nagesser SK, van Seters AP, Kievit J, Hermans J, Krans HM,
Decoding the genetic basis of Cushing’s disease: USP8 in the van de Velde CJ. Long-term results of total adrenalectomy
spotlight. Eur J Endocrinol 2015; 173: M73–83. for Cushing’s disease. World J Surg 2000; 24: 108–13.
139 Castillo V, Giacomini D, Paez-Pereda M et al. Retinoic acid 152 Smith PW, Turza KC, Carter CO, Vance ML, Laws ER, Hanks
as a novel medical therapy for Cushing’s disease in dogs. JB. Bilateral adrenalectomy for refractory Cushing disease:
Endocrinology 2006; 147: 4438–44. a safe and definitive therapy. J Am Coll Surg 2009; 208:
140 Pecori Giraldi F, Ambrogio AG, Andrioli M et al. Potential 1059–64.
role for retinoic acid in patients with Cushing’s disease. J 153 Vella A, Thompson GB, Grant CS, van Heerden JA, Farley
Clin Endocrinol Metab 2012; 97: 3577–83. DR, Young WF Jr. Laparoscopic adrenalectomy for adreno-
141 Vilar L, Albuquerque JL, Lyra R et al. The Role of Isotreti- corticotropin-dependent Cushing’s syndrome. J Clin Endo-
noin Therapy for Cushing’s Disease: Results of a Prospective crinol Metab 2001; 86: 1596–9.
Study. Int J Endocrinol 2016; 2016: 8173182. 154 Assie G, Bahurel H, Coste J et al. Corticotroph tumor
142 Liu NA, Araki T, Cuevas-Ramos D et al. Cyclin E-mediated progression after adrenalectomy in Cushing’s Disease: A
human proopiomelanocortin regulation as a therapeutic reappraisal of Nelson’s Syndrome. J Clin Endocrinol Metab
target for cushing disease. J Clin Endocrinol Metab 2015; 2007; 92: 172–9.
100: 2557–64. 155 Nelson DH, Meakin JW, Dealy JB Jr, Matson DD, Emerson
143 Liu NA, Jiang H, Ben-Shlomo A et al. Targeting zebrafish K Jr, Thorn GW. ACTH-producing tumor of the pituitary
and murine pituitary corticotroph tumors with a cyclin- gland. N Engl J Med 1958; 259: 161–4.
dependent kinase (CDK) inhibitor. Proc Natl Acad Sci U S A 156 Wilson PJ, Williams JR, Smee RI. Nelson’s syndrome: single
2011; 108: 8414–19. centre experience using the linear accelerator (LINAC) for
144 Riebold M, Kozany C, Freiburger L et al. A C-terminal HSP90 stereotactic radiosurgery and fractionated stereotactic
inhibitor restores glucocorticoid sensitivity and relieves a radiotherapy. J Clin Neurosci 2014; 21: 1520–4.
mouse allograft model of Cushing disease. Nat Med 2015; 157 De Tommasi C, Vance ML, Okonkwo DO, Diallo A, Laws ER
21: 276–80. Jr. Surgical management of adrenocorticotropic hormone-
145 Castinetti F, Fassnacht M, Johanssen S et al. Merits and secreting macroadenomas: outcome and challenges in
pitfalls of mifepristone in Cushing’s syndrome. Eur J patients with Cushing’s disease or Nelson’s syndrome. J
Endocrinol 2009; 160: 1003–10. Neurosurg 2005; 103: 825–30.
146 Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart 158 Katznelson L. Sustained improvements in plasma ACTH and
DE, Gross C. Mifepristone, a glucocorticoid receptor antag- clinical status in a patient with Nelson’s syndrome treated
onist, produces clinical and metabolic benefits in patients with pasireotide LAR, a multireceptor somatostatin analog. J
with Cushing’s syndrome. J Clin Endocrinol Metab 2012; 97: Clin Endocrinol Metab 2013; 98: 1803–7.
2039–49. 159 Pivonello R, Faggiano A, Di Salle F, Filippella M, Lombardi G,
147 Fleseriu M, Findling JW, Koch CA, Schlaffer SM, Buchfelder Colao A. Complete remission of Nelson’s syndrome after 1-
M, Gross C. Changes in plasma ACTH levels and corti- year treatment with cabergoline. J Endocrinol Invest 1999;
cotroph tumor size in patients with Cushing’s disease 22: 860–5.
during long-term treatment with the glucocorticoid receptor 160 Clayton RN, Raskauskiene D, Reulen RC, Jones PW. Mor-
antagonist mifepristone. J Clin Endocrinol Metab 2014; 99: tality and morbidity in Cushing’s disease over 50 years in
3718–27. Stoke-on-Trent, UK: audit and meta-analysis of literature. J
148 Hunt H, Donaldson K, Strem M et al. Assessment of safety, Clin Endocrinol Metab 2011; 96: 632–42.
tolerability, pharmacokinetics, and pharmacological effect 161 Clayton RN, Jones PW, Reulen RC et al. Mortality in patients
of orally administered CORT125134: an adaptive, double- with Cushing’s disease more than 10 years after remission:
blind, randomized, placebo-controlled phase 1 clinical a multicentre, multinational, retrospective cohort study.
study. Clin Pharmacol Drug Dev 2018; 7: 408–21. Lancet Diabetes Endocrinol 2016; 4: 569–76.
149 Moraitis A, Agrawal N, Bancos I et al. Open label phase 2
study to assess safety and efficacy of relacorilant Correspondence: Nicholas A. Tritos MD, DSc, Neuroendocrine and
(CORT125134), a selective cortisol modulator, in the treat- Pituitary Tumor Clinical Center and Neuroendocrine Unit, Mas-
ment of endogenous hypercortisolism. Endocr Pract 2018; sachusetts General Hospital, 100 Blossom Street # 140, Boston,
24: 300–1. MA 02114, USA. (fax: + 1 617 726 1241; e-mail: ntritos@mgh.har-
150 Balla A, Ortenzi M, Palmieri L et al. Laparoscopic bilateral vard.edu).
anterior transperitoneal adrenalectomy: 24 years experi-
ence. Surg Endosc 2019. https://doi.org/10.1007/
s00464-019-06665-6. [Epub ahead of print].

ª 2019 The Association for the Publication of the Journal of Internal Medicine 541
Journal of Internal Medicine, 2019, 286; 526–541

You might also like