throbber
18-Dang-Endo51/8 26/11/07 11:15 Page 1339
`
`Pharmacological Management of Cushing’s Syndrome:
`An Update
`
`atualização
`
`CUONG NGUYEN DANG
`PETER TRAINER
`
`Christie Hospital,
`Manchester, UK.
`
`ABSTRACT
`
`The treatment of choice for Cushing’s syndrome remains surgical. The role
`for medical therapy is twofold. Firstly it is used to control hyper-
`cortisolaemia prior to surgery to optimize patient’s preoperative state and
`secondly, it is used where surgery has failed and radiotherapy has not
`taken effect. The main drugs used inhibit steroidogenesis and include
`metyrapone, ketoconazole, and mitotane. Drugs targeting the hypo-
`thalamic-pituitary axis have been investigated but their roles in clinical
`practice remain limited although PPAR-γ agonist and somatostatin ana-
`logue som-230 (pasireotide) need further investigation. The only drug
`acting at the periphery targeting the glucocorticoid receptor remains
`Mifepristone (RU486). The management of Cushing syndrome may well
`involve combination therapy acting at different pathways of hypercor-
`tisolaemia but monitoring of therapy will remain a challenge. (Arq Bras
`Endocrinol Metab 2007;51/8:1339-1348)
`
`Keywords: Cushing’s syndrome; Drug therapy; Steroidogenesis inhibitor;
`Hypothalamic-pituitary modulator
`
`RESUMO
`
`Manejo Farmacológico da Síndrome de Cushing: Uma Atualização.
`O tratamento de escolha para a síndrome de Cushing ainda é a
`cirurgia. O papel da terapia medicamentosa é duplo: ele é usado para
`controlar o hipercortisolismo antes da cirurgia e otimizar o estado
`pré-operatório do paciente e, adicionalmente, quando ocorre falha
`cirúrgica e a radioterapia ainda não se mostrou efetiva. Os principais
`medicamentos são empregados para inibir a esteroidogênese e
`incluem: metirapona, cetoconazol e mitotano. Medicamentos visando
`o eixo hipotálamo-hipofisário têm sido investigados, mas seu papel
`na prática clínica permanece limitado, embora o agonista PPAR-γ e
`análogo de somatostatina, som-230 (pasireotídeo), requeira estudos
`adicionais. A única droga que age perifericamente no receptor glico-
`corticóide é a mifepristona (RU486). O manejo da síndrome de Cu-
`shing deve envolver uma combinação terapêutica atuando em
`diferentes vias da hipercortisolemia, mas o monitoramento dessa te-
`rapia ainda permanece um desafio. (Arq Bras Endocrinol Metab
`2007;51/8:1339-1348)
`
`Descritores: Síndrome de Cushing; Terapia médica; Inibidores da
`esteroidogênese; Moduladores hipotálamo-hipofisários
`
`Recebido em 01/10/07
`Aceito em 08/10/07
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`1339
`
`1
`
`TEVA1032
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1340
`
`Pharmacological Management of CS
`Dang & Trainer
`
`CUSHING’S IS ARARE DISEASE and therefore of min-
`
`imal interest to the pharmaceutical industry and
`hence for many years there were few developments.
`However in recent times there has been renewed inter-
`est in whether agents marketed for other conditions
`may have a role to play in the medical management of
`Cushing’s syndrome. This review will endeavour to
`assess the place of the ‘new’ agents alongside the
`longer established agents.
`The definitive management for Cushing’s syn-
`drome is surgical excision of the underlying cause of
`the hypercortisolaemia, with the exception of ACTH-
`independent bilateral macronodular hyperplasia where
`pharmacological treatment directed against the aber-
`rant receptor can be effective (1). However, in many
`patients with Cushing’s syndrome there is a role for
`medical therapy in certain specific circumstances. It is
`common practice to prepare patients for surgery by
`lowering circulating cortisol levels to reverse the meta-
`bolic consequence of cortisol excess and by implica-
`tion reduce the complications of surgery. This clearly
`depends on the interval to surgery and disease severi-
`ty. As any clinician dealing with Cushing’s syndrome is
`aware establishing the precise aetiology is a challenge
`and it is not always possible to make a definitive diag-
`nosis at first investigation, and in such cases medical
`therapy can be used as a stop gap to control signs and
`symptoms and thereby allow time for re-investigation.
`In patients not cured by surgery or in patients with
`metastatic disease medical therapy can be used to con-
`trol manifestations of the disease. Pituitary radiothera-
`py is extremely effective at controlling hypercortiso-
`laemia but can take several years to have its full effect
`and medical therapy is often required in the interim
`(see figure 1).
`Medical therapy can be separated into agents
`that inhibit adrenal steroidgenesis and those that mod-
`ulate pituitary ACTH release. Currently in clinical
`practice, the most effective, reliable and widely use
`agents are those that inhibit steroidgenesis.
`A major challenge of medical therapy is the
`monitoring of its effectiveness. Urinary free cortisol
`(UFC) measurement is widely used but has several
`major limitations and is intrinsically a poor solution to
`the problem of disease monitoring. Only a small pro-
`portion of cortisol is excreted unaltered in urine and
`UFC immunoassays to varying extent detect biologi-
`cally inactive cortisol metabolites, which may be raised
`in patients treated with agents such as metyrapone.
`UFC has the additional disadvantages of relying on
`complete collection and of being unable to detect
`over-treatment induced hypoadrenalism.
`
`Although more labour intensive, measurement
`of serum cortisol is a more appropriate means of
`assessing disease activity. The best validated technique
`is calculation of a mean serum cortisol from multiple
`measurements taken during a single day. Studies com-
`paring isotopically calculated cortisol production rates
`
`Figura 1. Mean plasma ACTH levels (a) and serum cortisol
`levels (b) during short-term metyrapone therapy in 53
`patients with Cushing’s syndrome. The bars represent the
`median values. ACTH ng/L x 0.225 = pmol/l. [Courtesy of Ver-
`helst JA, Trainer PJ, Howlett TA, et al. Short- and long-term
`responses to metyrapone in the medical management of 91
`patients with Cushing’s syndrome. Clin Endocrinology (Oxf)
`1991;35:169-78]
`
`1340
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`2
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1341
`
`to serum levels indicate that a mean serum cortisol in
`the range 150–300 nmol/l equates to a normal corti-
`sol production rate, and this should be the target of
`medical therapy (2).
`The cyclical nature of Cushing’s syndrome in
`some patients means that even after disease control has
`been achieved regular treatment monitoring is
`required.
`
`STEROIDOGENESIS INHIBITION
`
`These agents are the most consistently effective means
`of controlling cortisol secretion.
`
`Metyrapone
`In the era before it was possible to measure plasma
`ACTH, the metyrapone test was used to investigate
`suspected Cushing’s syndrome and hypoadrenalism
`but its use now is exclusively therapeutic (3,4). It acts
`primarily on the final step in cortisol synthesis namely
`the conversion of 11-deoxycortisol to cortisol and
`therefore results in a dramatic increase in circulating
`11-deoxycortisol levels, which can cross-react in serum
`and urine cortisol immunoassays. This cross-reactivity
`may result in spuriously elevated cortisol levels and a
`failure to appreciate that a patient is over-treated and
`hypoadrenal.
`Metyrapone is the most potent, short-acting
`inhibitor of cortisol synthesis with a rapid onset of
`action. Serum cortisol levels fall within four hours of
`an initial dose and care is required to avoid over-treat-
`ment. The routine starting dose is 250 mg three times
`per day with reassessment of cortisol levels 72 hours
`later and dose titration as appropriate until a mean
`cortisol level of between 150 and 300 nmol/l is
`achieved. In patients with severe hypercortisolaemia
`up to 8 gm per day in 3–4 divided doses may be nec-
`essary. Most patients tolerate the drug without diffi-
`culty as long as hypoadrenalism is avoided. Nausea,
`anorexia and abdominal pain can occur but usually
`this is a sign of over-treatment. The major limitation
`of metyrapone is in women as the accumulation of
`cortisol precursors results in elevated androgens,
`which frequently is manifest as hirsutism and acne.
`Although mineralocorticoid precursors levels are ele-
`vated, hypokalaemia, hypertension and oedema are
`not problems, presumably because of the benefits of
`lower circulating cortisol levels (5,6). In patients with
`pituitary-dependent Cushing’s disease, ACTH levels
`rise but there is no evidence that this results in tachy-
`phylaxis (5,7).
`
`Pharmacological Management of CS
`Dang & Trainer
`
`Ketoconazole
`Ketoconazole is an imidazole derivative developed as
`an oral antifungal agent that inhibits cholesterol, sex
`steroid and cortisol synthesis by acting on the
`11β-hydroxylase and C17-20 lyase enzymes (8-11). It
`is the most frequently used agent in the treatment of
`Cushing’s syndrome with the starting dose being 200
`mg twice daily increasing as necessary to 1200 mg/day
`in four divided doses (12,13). In contrast to
`metyrapone it can take several weeks to see the full
`benefit of a dose adjustment and there is less risk of
`over-treatment and hypoadrenalism. With time it is
`effective at controlling the symptoms of Cushing’s
`syndrome and in women its antiandrogenic properties
`are a virtue but in men, gynaecomastia and reduced
`libido have been reported. The most common side
`effects are gastrointestinal upset and skin rashes but
`liver enzyme dysfunction can occur in up to 10% of
`cases, which rarely has proceeded to acute liver failure
`and fatality (14-17). Ketoconazole has the added ben-
`efit of reducing the total cholesterol and LDL choles-
`terol (18).
`Metyrapone and ketoconazole can be very suc-
`cessfully co-administered as the former controls corti-
`sol secretion while waiting for the slower onset of
`action of the latter agent, which in turn lowers andro-
`gens and thus negates one of the major limitations of
`the former.
`
`Mitotane
`Mitotane reduces cortisol production by blocking
`cholesterol side-chain cleavage and 11β-hydroxylase
`(19-21). It was introduced in 1960 for the treatment
`of adrenal carcinoma and subsequently used for the
`treatment of benign causes of Cushing’s syndrome.
`The onset of mitotane action is slow with sustained
`action maintained after discontinuation in up to a
`third of patients (22). When used to control serum
`cortisol levels in benign disease, mitotane is initiated
`at a dose of 0.5–1 gm per day which is increased grad-
`ually by 0.5–1 gm every few weeks to minimise side
`effects. Adverse effects such as nausea, anorexia and
`diarrhoea are common with doses of 2 gm per day
`and almost universal at doses greater than 4 gm per
`day (23). Adrenal insufficiency and neurological side
`effects including abnormal gait, dizziness, vertigo,
`confusion and problem of language expression are
`often seen at higher dose (22). Abnormal liver
`enzymes, hypercholesterolaemia, skin rash, hyporuri-
`caemia, gynaecomastia in male and prolonged bleed-
`ing time are also well recognized (24,25). Changes in
`hormone binding globulins may result in total hor-
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`1341
`
`3
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1342
`
`Pharmacological Management of CS
`Dang & Trainer
`
`mone measurement being unreliable during treat-
`ment and thus caution is required when interpreting
`serum cortisol levels (26,27). Mitotane increases the
`metabolic clearance of exogenously administered
`steroid and the replacement dose of steroid is
`increased by about a third (28). In order to minimise
`side effects mitotane dose should be gradually titrated
`up, taken with meals or at bedtime with food. Chang-
`ing the schedule to once daily or alternate day may
`help with gastrointestinal problems. If side effects are
`severe mitotane can be stopped for a week and restart-
`ed at a lower dose. Mitotane may induce spontaneous
`abortion and is a teratogen. Its effect may persist for
`a number of months after discontinuation and so a
`female patient should avoid pregnancy for up to five
`years after stopping the drug (29).
`
`Aminoglutethimide
`Aminoglutethimide, which was introduced in 1959 as
`an anticonvulsant, has also been used in the treatment
`of breast cancer and was noticed to induce adrenal
`insufficiency. It inhibits the side-chain cleavage of cho-
`lesterol to pregnenolone and therefore inhibits corti-
`sol, oestrogen and aldosterone production and addi-
`tionally inhibits 11β-hydroxylase, 18-hydroxylase and
`aromatase activity
`(30,31).
`Initially aminog-
`lutethimide decreases cortisol production in Cushing’s
`syndrome but appears to be less effective in treating
`Cushing’s disease (32). The suggested mechanism
`may be an increase in ACTH overcoming the enzy-
`matic blockade or it may be induction of hepatic
`enzyme accelerating aminoglutethimide metabolism
`(33,34). Adverse effects such as lethargy, dizziness,
`ataxia and rashes are common on initiation and limit
`its use although they do resolve with time (32,35).
`There are better agents for controlling hypercortiso-
`laemia and aminoglutethimide does not have a place in
`the modern treatment of Cushing’s syndrome (36).
`
`Trilostane
`Trilostane is a competitive inhibitor of 3β-hydroxys-
`teroid dehydrogenase, which is an essential enzyme in
`the synthesis of cortisol, aldosterone and androstene-
`dione. It is an effective inhibitor of steroid synthesis in
`vitro but in man the results have been disappointing
`(37). However, it is used in veterinary practice as it is
`very effective in controlling pituitary-dependent Cush-
`ing’s in dogs (38). The maximum daily dose is 1,440
`mg and patients may experience side effects such as
`abdominal discomfort, diarrhoea and paraesthesia.
`Trilostane has largely fallen out of clinical use but the
`very fact that it is so effective in dogs may mean it jus-
`tifies reconsideration in man.
`
`Etomidate
`Etomidate is a parenteral anaesthetic agent which
`when first introduced was associated with excessive
`mortality in patients in intensive care which was ulti-
`mately explained by the recognition it lowered circu-
`lating cortisol levels by inhibiting 11β-hydroxylase,
`17-hydroxylase, c17-20 lyase as well as cholesterol side
`chain at cleavage (39-41). A number of case reports
`have shown etomidate at 2.5 mg/hour to be effective
`at correcting hypercortisolaemia in seriously ill patients
`with ectopic ACTH production (42-44). Etomidate’s
`use is limited by the need to be given intravenously
`but it has a place in acutely sick patients unable to be
`treated orally where rapid correction of hypercortiso-
`laemia may be life saving.
`
`HYPOTHALAMIC-PITUITARY
`NEUROMODULATORY AGENTS
`
`Pituitary ACTH secretion is regulated by a number of
`neurotransmitters including catecholamines, sero-
`tonin, acetylcholine, GABA and peptides. In Cush-
`
`Ketoconazole
`
`Table 1. Agents inhibiting steroidogenesis in clinical use.
`Agent
`Dose
`Properties
`Metyrapone
`750–8000
`Hypoadrenalism
`mg daily
`Side effects: nausea, abdominal pain, hirsutism, acne
`400–1200
`Slow onset of action
`mg daily
`Side effects: gastro-intestinal upset, rashes, abnormal
`LFT, gynaecomastia & reduced libido in men
`Gradual dose titration, taken with meal
`Side effects: gastro-intestinal upset, neurological
`disturbances, abnormal LFT, hypercholesterolaemia
`Avoid pregnancy up to five years after stopping the drug
`
`Mitotane
`
`500–8000
`mg daily
`
`1342
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`4
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1343
`
`ing’s disease the pituitary tumour still remains partial-
`ly responsive to hypothalamic stimuli, illustrated by
`responsiveness to exogenous CRH and dexametha-
`sone. Reports exist advocating the virtues of various
`agents but, to-date, none have gained widespread
`acceptance. However recent data have renewed inter-
`est in the possibility of treating Cushing’s disease with
`centrally acting drugs that modulate through
`dopamine, somatostatin and PPAR receptors function.
`
`Dopamine agonists
`Bromocriptine is a dopamine agonist which has been
`widely used in the treatment of hyperprolactinaemia
`and acromegaly. It is unclear if the action in lowering
`ACTH secretion by bromocriptine is via CRH or
`directly on the pituitary (45-47). A single dose of
`bromocriptine will cause a fall in ACTH in half of the
`patients with Cushing’s disease but unfortunately this
`effect is not maintained in the long term (47,48).
`There are reports that suggest with high dose
`bromocriptine (40 mg/day) there may be clinical
`improvement in up to 50% of patients but others have
`found response rate of only 1–2% in the long term
`(49,50). Potential side effects of bromocriptine
`include nasal congestion, nausea, postural hypoten-
`sion, headaches and hallucination.
`The use of cabergoline in the management of
`Cushing’s disease remains anecdotal. In mixed pitu-
`itary tumour secreting prolactin and ACTH with florid
`clinical signs of Cushing’s disease treatment with
`cabergoline resulted not only in the normalisation of
`prolactin but also clinical and biochemical resolution
`of the features of Cushing’s (51). It has also been use
`to control Cushing’s disease in failed pituitary surgery
`(52,53). Recently there has been renewed interest in
`cabergoline with the publication by Pivonello et al. of
`a case of lung carcinoid with Cushing’s syndrome
`treated with a combination of lanreotide and cabergo-
`line successfully normalising plasma ACTH and UFC
`levels (54). In a study of six patients with ACTH-
`secreting neuroendocrine tumours, dopamine D2
`receptors were expressed in five patients on immuno-
`histochemistry and treatment with cabergoline 3.5
`mg/week for six months normalised UFC in two
`patients although one patient later did have treatment
`escape (55). Case reports of cabergoline use in Nel-
`son’s syndrome have been more encouraging. Casulari
`et al. reported a case of Nelson’s syndrome with failed
`treatment on cyproheptadine (12 mg/day) and
`bromocriptine (7.5 mg/day) but cabergoline (0.5 mg
`twice weekly) normalised the ACTH plasma levels and
`induced complete resolution of the pituitary adenoma
`
`Pharmacological Management of CS
`Dang & Trainer
`
`on MRI (56). There has also been a case report of
`cabergoline (1.5 mg/week) treatment of Nelson’s
`syndrome for six years with normalisation of ACTH
`levels and stable residual pituitary tumour (57).
`The role of dopamine agonists in the manage-
`ment of Cushing’s disease remains limited to the occa-
`sional patient and long-term evidence of efficacy is
`very poor but interest in their use remains unabated.
`The available data are case based anecdote, there is a
`need for a controlled study before treatment can be
`recommended.
`
`PPAR-γ receptor agonists
`In 2002, the nuclear hormone receptor, peroxisome
`proliferator-activated receptor-γ (PPAR-γ) was identi-
`fied in ACTH-secreting pituitary tumour (58). In an
`in vivo experiment, innoculating mice with corti-
`cotroph AtT20 tumour cells, treating with extremely
`high dose of rosiglitazone (150 mg/kg/day) prevent-
`ed the development of tumours. In mice with already
`established corticotroph tumours, rosiglitazone treat-
`ment decreased tumour volume in 75% of cases and
`prevented signs of hypercortisolaemia in all cases, with
`75% reduction in ACTH level and 96% reduction in
`cortisol levels (58). These observations caused great
`interest but are yet to impact on clinical practice.
`In a study of two patients with pituitary-depen-
`dent Cushing’s syndrome treated with rosiglitazone 8
`mg daily for 33 and 20 days (the second patient was
`also taking metyrapone 1 gm/day), 24 hours UFC fell
`in both patients although only in the patient co-treat-
`ed with metyrapone did it reach statistical significance
`(59). In a second study of ten patients, four prior to
`surgery, four following relapse after surgery and two
`immediately after failed surgery treated with 4–16 mg
`of rosiglitazone for 1–8 months (median 3 months),
`there was no consistent reduction in urinary free corti-
`sol, plasma ACTH or serum cortisol levels (60). Side
`effects reported included oedema, weight increase,
`somnolence and increased hirsutism. In one of the larg-
`er studies, fourteen patients with active Cushing’s dis-
`ease (seven untreated and seven post unsuccessful
`transsphenoidal pituitary surgery) were treated with
`8–16 mg of rosiglitazone for 1–7 months (61). In six
`patients, plasma ACTH, serum cortisol and 24 hours
`UFC were lowered but only UFC reached significance.
`Two of the six patients also noted clinical improvement
`on follow up at seven months. No clinical side effects
`were noted but one patient developed hypercholes-
`terolaemia. In a study of seven patients with Nelson’s
`syndrome who took 8 mg of rosiglitazone for 12
`weeks, no significant fall in ACTH was seen (62). Sim-
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`1343
`
`5
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1344
`
`Pharmacological Management of CS
`Dang & Trainer
`
`ilarly in another study of six patients with Nelson’s syn-
`drome given rosiglitazone 12 mg per day for 14 weeks,
`there was no fall in ACTH levels (63).
`Although most studies used rosiglitazone in
`treating Cushing’s disease, pioglitazone has also been
`tried. In a study of five patients with Cushing’s disease
`treated with pioglitazone 45 mg for 30 days, no alter-
`ation in 24 hours UFC, or ACTH and cortisol
`responses to CRH administration was seen (64).
`Currently the success of PPAR agonists in treat-
`ing Cushing’s disease remains disappointing, failing to
`reproduce the success seen in the in vitro and mouse
`model. However with the small number of patients
`and short duration of treatment, further studies are
`still needed. The discrepancy between the in vitro and
`human experience may reflect the differences in the
`order of magnitude in the dose of rosiglitazone.
`
`Somatostatin analogues
`Octreotide, an analogue of somatostatin, has been
`used extensively to treat neuroendocrine tumours and
`acromegaly. In the 1990s five subtypes of somatostatin
`receptors were identified with expression of somato-
`statin receptor subtypes in mammalian corticotrophs
`being variable (65).
`In one study all five subtype somatostatin recep-
`tors were co-localised in rat pituitary cells expressing
`ACTH (66). Yet in another study only 38% of corti-
`cotrophs expressed somatostatin receptor subtype 5
`(sst5) and 3% expresses somatostatin receptor subtype
`2 (sst2) (67). While in contrast Smith et al. found a
`predominance of sst2 rather than sst5 (68). It is gener-
`ally accepted that sst2 and sst5 are involved in the reg-
`ulation of growth hormone, prolactin and TSH (69).
`In vitro studies suggested that normal cortico-
`troph only responds to somatostatin with inhibition of
`ACTH release if the cells have been cultured in gluco-
`corticoid free medium (70-74). In agreement with this is
`the finding that ACTH secretion in normal individuals is
`not affected by infusion of somatostatin or octreotide
`but is affected in patients with Addison’s disease (75,76).
`Initial reports did show that somatostatin infusion
`decreases plasma ACTH level by between 40% to 70% in
`patients with Nelson’s syndrome (77). However, subse-
`quent studies in Nelson’s syndrome have been less
`impressive and most patients with Cushing’s disease have
`failed to respond (78-82). The chronic treatment of rat
`pituitary tumour cells and mouse corticotroph cells with
`glucocorticoid results in decreased binding of somato-
`statin (83). In cultured human corticotroph, adenoma
`cells pre-treated with hydrocortisone resulted in abolition
`of octreotide-induced inhibition of basal and CRH
`
`induced ACTH release (82). The lack of clinical efficacy
`of octreotide may be due to the down regulation of so-
`matostatin receptors by glucocorticoids. In fact in the
`mouse, sst2 gene promoter sequence is the only somato-
`statin receptor shown to be directly transcriptionally reg-
`ulated by glucocorticoids (84,85). There has been specu-
`lation that octreotide may have a role in treating ectopic
`ACTH producing tumours or in Cushing’s disease in
`combination with ketoconazole but the available eviden-
`ce is unconvincing that it has any role in Cushing’s dis-
`ease (86,87).
`There is renewed interest in somatostatin ana-
`logues in Cushing’s disease because of encouraging
`data emerging from early studies with SOM-230
`(pasireotide, Novartis Pharmaceuticals UK Ltd). It is a
`new somatostatin analogue with affinity to all the
`somatostatin receptor subtypes but with 40 fold high-
`er affinity for sst5 than octreotide (88-90).
`Compared to octreotide, SOM-230 is more
`potent at suppressing ACTH release and at inhibiting
`CRH-induced ACTH release in corticotroph tumour
`cells (91-93). Dexamethasone (10 nM) pre-treatment
`of mouse corticotroph cells fails to suppress SOM-230
`inhibition of CRH-induced ACTH release whereas the
`suppressive effect of octreotide is blocked (92).
`The preliminary results of an open label, single
`arm phase 2 study of fourteen patients with persistent or
`recurrent Cushing’s disease treated with pasireotide 600
`µg subcutaneously twice daily for fifteen days, were
`reported as an abstract at ENDO 2006 (94). Pasireotide
`normalised UFC in 3 patients (21%) and in a further 7
`patients there was at least 40% reduction in UFC com-
`pared to baseline. There was significant improvement in
`symptoms including weight loss, facial rubor, abdomi-
`nal obesity, fatigue and proximal weakness in over 40%
`of patients. The drug was well tolerated but common si-
`de effects were mild to moderated gastrointestinal
`upset, injection site reaction and a transient increase in
`fasting blood glucose with one pre-existing diabetes
`mellitus patient stopping treatment early. Although the
`results of this preliminary study are encouraging the
`final results are awaited and further studies will be requi-
`red to confirm these results.
`
`Cyproheptadine
`Cyproheptadine is a non-selective histamine and sero-
`tonin antagonist. In a small series, at a dose of 24 mg/
`day, it was effective at reducing ACTH in three patients
`with Cushing’s disease (95). There is disagreement on
`whether cyproheptadine acts either directly on the pitu-
`itary or through the inhibition of CRH (96-99). It is
`rarely effective and has no place in current practice. Its
`main side effect is sedation.
`
`1344
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`6
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1345
`
`Ritanserin
`Ritanserin is a specific 5-HT2 antagonist which has
`been used in a few patients but its effects do not
`appear to be sustained in most patients (100,101).
`
`Sodium valproate
`Sodium valproate is mainly used as an anti-epileptic
`agent. Evidence for its effectiveness in treating Cush-
`ing’s disease remains conflicting. There are reports
`suggesting it is successful at suppressing ACTH at a
`daily dose of 600 mg, but more recent data have failed
`to demonstrate benefit either as primary therapy or
`after failed pituitary surgery (102,103). However, it
`may have a role as add on therapy to metyrapone at a
`daily dose of 1–2 g (104,105).
`
`Retinoic acid
`Since the 1980’s retinoic acid derivatives are widely
`used by dermatologists in the treatment of acne and
`psoriasis as well as in certain malignancy such as acute
`promyelocytic leukaemia (106,107). Retinoic acid is a
`ligand for Nur77/Nurr1 receptor which is involved in
`the physiological stimulation of ACTH by CRH
`(108). Retinoic acid inhibits cell proliferation and
`induces cell death in ACTH secreting tumours but not
`in normal pituitary cells. In the adrenal cortex it
`inhibits corticosterone secretion and cell proliferation,
`while in a mouse model, it blocks tumour growth and
`reduces circulating ACTH and cortisol. The dose was
`10 mg/kg which is within the dose range in human
`cancer therapy (108). Studies in rodents and dogs
`models of Cushing’s disease have been successful but
`there is now a need for studies in human (108,109).
`
`Agents blocking cortisol action
`Mifepristone (RU486) is a potent antagonist of the
`glucocorticoid and progesterone receptors (110). In
`man mifepristone blocks glucocorticoid action result-
`ing in negative feedback at the hypothalamic-pituitary
`level leading to a rise in ACTH, arginine-vasopressin
`and therefore cortisol (111). Mifepristone, at doses of
`
`Pharmacological Management of CS
`Dang & Trainer
`
`up to 20 mg/kg, has been successfully used to treat a
`small number of patients with ectopic ACTH syn-
`drome and there is every reason to believe that it could
`be successfully used in all patients if it were not for the
`problem of monitoring therapy (112). As a receptor
`antagonist it does not lower circulating cortisol levels,
`which in fact rise, and therefore it is very difficult to
`dose titrate and judge effectiveness. The GH receptor
`antagonist pegvisomant has gained widespread accep-
`tance as a treatment for acromegaly because its effec-
`tiveness can be judged by monitoring IGF-1. Unfor-
`tunately, the HPA axis lack a marker analogous to
`IGF-I. Even with short term use, a number of patients
`did develop symptoms of hypoadrenalism, which is
`problematic as there is no effective method of moni-
`toring over treatment (113). There has also been
`report of a case of mifepristone causing severe
`hypokalaemia that is attributed to excess cortisol acti-
`vation of mineralcorticoid receptor which responded
`to spironolactone therapy (114). With caution,
`mifepristone may have a role in the treatment of Cush-
`ing’s syndrome and could be first line treatment if a
`biochemical measure of disease were identified (115).
`
`CONCLUSIONS
`
`A number of drugs have been used in the management
`of Cushing’s syndrome. Regardless of the aetiology,
`steroid biosynthesis remains the most effective and wide-
`ly used agent. The preferred treatments are metyrapone
`or ketoconazole as monotherapy, or in combination.
`Careful monitoring of therapy is important as all agents
`have the potential of causing hypoadrenalism.
`Currently drugs acting on the hypothalamic-
`pituitary pathways have been less successful in clinical
`practice and their role is likely to be limited to add on
`therapy on an individual basis. However, with the
`identification of new receptors and development of
`agent blocking these receptors, there remains the hope
`that they may still prove to be useful in the future.
`
`Table 2. Potential agents for treating Cushing’s syndrome.
`Receptors
`Agents
`Properties
`Dopamine
`Cabergoline
`Poor long term results but renewed interest
`Bromocriptine
`especially in treating Nelson’s syndrome
`Rosiglitazone
`In vitro success not reproduced in clinical practice
`Pioglitazone
`SOM-230
`(pasireotide)
`Retinoic acid
`Nu77/Nurr1
`Glucocorticoid Mifepristone
`
`PPAR-γ
`
`Somatostatin
`
`Phase 2 study show promising results
`
`Evaluated in mouse model
`Successfully utilised in a small number of patients
`Inability to monitor treatment limits usefulness
`
`Arq Bras Endocrinol Metab 2007;51/8
`
`1345
`
`7
`
`

`

`18-Dang-Endo51/8 26/11/07 11:15 Page 1346
`
`Pharmacological Management of CS
`Dang & Trainer
`
`REFERENCES
`
`1. Christopoulos S, Bourdeau I, Lacroix A. Aberrant expression
`of hormone receptors in adrenal Cushing’s syndrome. Pitu-
`itary 2004;7:225-35.
`2. Trainer PJ, Eastment C, Grossman AB, Wheeler MJ, Perry L,
`Besser GM. The relationship between cortisol production rate
`and serial serum cortisol estimation in patients on medical
`therapy for Cushing’s syndrome. Clin Endocrinol (Oxf)
`1993;39:441-3.
`3. Newell-Price J, Grossman AB. The differential diagnosis of
`Cushing’s syndrome. Ann Endocrinol (Paris) 2001;62:173-9.
`4. Liddle GW, Estepe HL, Kendall JWJ, Williams WCJ, Townes
`A. Clinical application of a new test of pituitary reserve. J
`Clin Endocrinol Metab 1959;19:875-94.
`5. Verhelst JA, Trainer PJ, Howlett TA, Perry L, Rees LH, Gross-
`man AB, et al. Short and long-term responses to metyrapone
`in the medical management of 91 patients with Cushing’s
`syndrome. Clin Endocrinol (Oxf) 1991;35:169-78.
`6. Connell JM, Cordiner J, Davies DL, Fraser R, Frier BM,
`McPherson SG. Pregnancy complicated by Cushing’s syn-
`drome: potential hazard of metyrapone therapy. Case report.
`Br J Obstet Gynaecol 1985;92:1192-5.
`7. Orth DN. Metyrapone is useful only as adjunctive therapy in
`Cushing’s disease. Ann Intern Med 1978;89:128-30.
`8. Engelhardt D, Dorr G, Jaspers C, Knorr D. Ketoconazole
`blocks cortisol secretion in man by inhibition of adrenal 11
`beta-hydroxylase. Klin Wochenschr 1985;63:607-12.
`9. Oelkers W, Bahr V, Hensen J, Pickartz H. Primary adrenocor-
`tical micronodular adenomatosis causing Cushing’s syn-
`drome. Effects of ketoconazole on steroid production and in
`vitro performance of adrenal cells. Acta Endocrinol
`(Copenh) 1986;113:370-7.
`10. Sonino N. The endocrine effects of ketoconazole. J
`Endocrinol Invest 1986;9:341-7.
`11. Weber MM, Luppa P, Engelhardt D. Inhibition of human
`adrenal androgen secretion by ketoconazole. Klin Wochen-
`schr 1989;67:707-12.
`12. Angeli A, Frairia R. Ketoconazole therapy in Cushing’s dis-
`ease. Lancet 1985;1:821.
`13. Sonino N, Boscaro M, Paoletta A, Mantero F, Zilio

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket