throbber
This material may be protected by Copyright law (Title 17 U5. Code
`
`Mifepristone for Management of Cushing’s Syndrome
`
`Farah H. Morgan, and Marc J. Laufgraben
`
`Cushing’s syndrome is a debilitating endocrine disorder caused by elevated
`circulating glucocorticoid levels. Although uncommon, Cushing’s syndrome is
`associated with significant morbidity necessitating rapid reversal of hypercorti-
`solemia. Primary therapy for most patients with Cushing’s syndrome is surgi-
`cal, but many patients will require additional treatments with radiation or
`drugs. Although several options for drug therapy exist, few are readily available
`and all have dose-limiting adverse effects. Mifepristone (RU 486), a first-in-
`Class glucocorticoid receptor antagonist, was approved by the United States
`Food and Drug Administration in 2012 for use in Cushing’s syndrome to con-
`trol hyperglycemia in patients who are not surgical candidates or have not
`achieved remission from surgery. The drug is approved for oral once-daily
`administration. In its pivotal trial, 60% of patients responded to mifepristone
`with significant improvements in glycemic control and 38% had a reduction in
`diastolic blood pressure. The most common adverse events were nausea, fati-
`gue, headache, endometrial hyperplasia, and hypokalemia. Adrenal
`insuffi-
`ciency occurred in fewer than 5% of patients. The recommended starting
`dosage of mifepristone is 300 mg/day. The dosage may be increased every 2—
`4 weeks up to a maximum of 1200 mg/day, although it should not exceed
`20 mg/kg/day. Significant drug—drug interactions exist due to mifepristone’s
`effects on a number of cytochrome P450 enzymes. Despite its limitations, mife-
`pristone is a welcome addition and an appropriate alternative to the available
`drug therapy for Cushing‘s syndrome.
`Key Words: mifepristone, glucocorticoid receptor antagonist, Cushing’s syn-
`drome, Cushing’s disease, RU486.
`(Pharmacotherapy 2013;33(3):319-—329)
`
`Cushing’s syndrome is a rare but debilitating
`endocrine disorder caused by excess circulat-
`ing glucocorticoids. The excess glucocorticoids
`result from increased glucocorticoid production in
`the adrenal gland secondary to adrenal stimulation
`
`From the Division of Endocrinology, Diabetes and Metab-
`olism, Department of Medicine, Cooper Medical School of
`Rowan University, Camden, Ncwjersey (both authors).
`For questions or comments, contact Marc ]. faufgrabcn,
`Divisron' Head, DIViSion .Of Endocrinology, Diabetes and
`Metabolism, Cooper Medical School of Rowan UniverSity,
`Cooper University Hospital, 3 Cooper Plaza Suite 220,
`Camden, NJ
`08103;
`c-mail:
`laufgraben-marc@cooper
`healthedu.
`
`1
`
`or a primary adrenal tumor. For most forms of
`Cushing’s syndrome, the initial treatment is surgi-
`cal. However, a substantial proportion of patients
`will not be cured by surgery. Second-line therapy
`can include additional surgery, radiation, or phar-
`macologic agents. Previously available drugs have
`primarily been inhibitors of adrenal steroid syn-
`thesw’ and .[lle use Of these agents has beep limited
`by aVallablllty and tolerabillty.
`leeprlstone, a
`first-in-class glucocorticoid receptor antagonist,
`was appI‘OVCd by the United States Food and Drug
`Administration (FDA) in 2012 for use in patients
`.
`,
`_
`,
`Wlth
`hyperglycemia
`secondary
`‘0 CUShmgS
`Syndrome. With approval of this new agent, prac-
`titioners need a thorough understanding of its
`TEVA1 030
`
`1
`
`TEVA1030
`
`

`

`320
`
`PHARMACOTHERAPY Volume 33, Number 3, 2013
`
`pharmacology, pharmacokinetics, pharmacody-
`namics, clinical efficacy, indications for use, and
`limitations.
`
`Cortisol: Normal Physiology
`
`Secretion of cortisol is maintained by a clas—
`sic endocrine feedback system. Cortisol pro-
`duction occurs in the zona fasciculata cells of
`the adrenal cortex. These cells are stimulated
`by
`adrenocorticotrophic
`hormone
`(ACTH),
`which is secreted by corticotroph cells in the
`anterior pituitary gland. ACTH production is
`stimulated by corticotrophin—releasing hormone
`(CRH) produced in the paraventricular nucleus
`of the hypothalamus. Circulating cortisol
`then
`provides negative feedback to inhibit produc-
`tion of CRH and ACTH. Thus, cortisol dynam-
`ics depend on normal hypothalamic, pituitary,
`and adrenal function—the hypothalamic-pitui—
`tary—adrenal axis
`(Figure 1). Normal cortisol
`levels follow a circadian rhythm with a peak in
`the early morning (7:00—9:00 AM.) and a nadir
`at 11:00 P.M. CRH production is further regu-
`lated by physiologic and emotional stress.1
`Cortisol is necessary to sustain life. It plays a
`role in multiple essential
`functions including
`carbohydrate, protein, and lipid metabolism and
`vascular tone and blood pressure maintenance.
`It
`is also involved in the immune system and
`
`Stress
`
`\
`
`Circadian rhythm
`
`/
`
`>
`
`llypothalamus
`
`CRH
`
`Pituitary
`
`ACTH
`
`Adrenal Cortex
`
`Cortisol
`
`Figure 1. Normal regulation of the hypothalamic-pituitary-
`adrenocortlcal
`axis.
`'
`2 ACTH = andrenocorticotrophic
`hormone; CRH = corticotrophin-releasing hormone.
`
`2
`
`their
`responses to stress. Glucocorticoids exert
`actions mainly through binding at the glucocor-
`ticoid receptor, a member of the thyroid and
`steroid hormone receptor superfamily of nuclear
`transcription factors. As would be expected, the
`glucocorticoid receptor is expressed widely in
`peripheral
`tissues
`and brain regions. Many
`glucocorticoids,
`including cortisol,
`also have
`affinity for the mineralocorticoid receptor. How-
`ever, under normal circumstances,
`the renal
`mineralocorticoid receptor is “protected” from
`cortisol binding by the local activity of type 2
`llB—hydroxysteroid dehydrogenase (1 1 B—HSD),
`which converts cortisol
`to cortisone and does
`not bind to
`the mineralocorticoid receptor.
`Under physiologic circumstances, aldosterone is
`the primary activator of the mineralocorticoid
`receptor;
`its activation promotes sodium reten-
`tion (and therefore maintenance of blood pres—
`sure) and potassium excretionl’ 2
`Deficiency of cortisol results in the signs and
`symptoms of adrenal insufficiency, which can vary
`in severity from fatigue and anorexia to hypoten-
`sion and hypoglycemia to shock and death. Corti-
`sol excess results in Cushing’s syndrome.
`
`Cushing’s Syndrome
`
`the result of excess
`Cushing’s syndrome is
`circulating glucocorticoids. Exogenous, or iatro-
`genic, Cushing’s syndrome is common and typi-
`cally results from the use of supraphysiologic
`doses of glucocorticoids
`to treat pulmonary,
`rheumatologic, hematologic, or other disorders.
`Endogenous Cushing’s syndrome is
`rare and
`results from inappropriate activation of either the
`pituitary gland or adrenal glands,
`leading to
`increased circulating cortisol levels. The majority
`of cases are caused by an ACTH-secreting tumor
`of the pituitary gland (i.e., Cushing’s disease).
`Cushing’s syndrome may also result from ectopic
`secretion of ACTH by neoplasms such as small
`cell lung cancer and carcinoid tumors; rarely, a
`tumor may secrete CRH. Cushing’s syndrome can
`also result from benign, malignant, or hyperplas—
`tic diseases of the adrenal glands that secrete
`cortisol in the absence ofACTH stimulation.3'5
`
`The excess cortisol seen in Cushing’s syn-
`drome results in hypertension, hyperglycemia,
`obesity,
`and a myriad
`of other problems
`(Table 1).5 These complications lead to signifi-
`cant morbidity related to illness and twice the
`mortality rate in patients with Cushing’s syn-
`drome compared to the general population.6
`Diabetes mellitus and hypertension are the most
`
`2
`
`

`

`MIFEPRISTONE FOR CUSHING’S SYNDROME Morgan et al
`Table 1. Signs and Symptoms of Cushing‘s SyndromeS
`Other
`
`321
`
`Symptoms
`
`Depression
`
`Fatigue
`Weight gain
`
`Signs
`
`Obesity (especially
`central obesity)
`Facial plethora
`Moon facies
`
`Conditions
`
`Hypokalemia
`
`Hypertension
`Diabetes
`mellitus
`
`Insomnia
`Muscle weakness
`
`Irritability
`
`Irregular menses
`Back pain
`
`Osteoporosis
`Renal calculi
`
`Easy bruising
`Striae (especially wide
`violaceous striae)
`Proximal myopathy
`Dorsocervical fat pad
`Supraclavicular fat pads
`Edema
`Acne
`Thin skin
`Ilirsutism
`Female balding
`
`important predictors of death.6 Hypertension
`occurs in 80% of patients with Cushing’s syn—
`drome and is thought to be caused by the effect
`of cortisol on both the glucocorticoid receptor
`and the mineralocorticoid receptor."r The excess
`cortisol overwhelms the ability of type 2 HB-
`HSD to convert it to cortisone, and thus cortisol
`has access to and activates the mineralocorti-
`coid receptor.
`Impaired glucose tolerance or
`type 2 diabetes also occurs in 80% of patients as
`a
`result of
`increased insulin resistance and
`impaired
`insulin
`secretion.(7 Glucocorticoids
`increase insulin resistance through actions on
`liver, skeletal muscle, and adipose tissue. The
`net result of increased liver gluconeogenesis and
`decreased glucose uptake in skeletal muscle and
`adipose tissue is hyperglycemia.6 Impaired insu—
`lin secretion also occurs as a result of glucocor-
`ticoids binding to pancreatic B cells, resulting in
`impaired B-cell function.6 This combined effect
`causes hyperglycemia that can be very difficult
`to treat, often requiring escalating doses of insu-
`lin for appropriate management.
`
`Diagnosis of Cushing’s Syndrome
`
`Because of the complexity of the screening
`and diagnostic algorithms for Cushing’s
`syn-
`drome, referral
`to an endocrinologist is appro-
`priate when the disorder is suspected. Accurate
`diagnosis of Cushing’s
`syndrome
`is
`critical
`to avoid unnecessary testing, procedures, and
`expenditures.5' 7
`testing for
`Before
`considering biochemical
`endogenous Cushing’s syndrome, it is important
`to rule out exogenous glucocorticoid exposure
`
`Initial
`causing iatrogenic Cushing’s syndrome.
`testing can include measurement of 24-hour
`urine free cortisol,
`late-night salivary cortisol
`collected at 11:00 P.M. or 12:00 A.M., or early
`morning cortisol after dexamethasone 1 mg the
`previous evening at 11:00 P.M. (overnight dexa-
`methasone l-mg suppression test). When an
`abnormal result is obtained, a physiologic cause
`of hypercortisolemia,
`such as depression or
`other psychiatric illness, alcohol abuse, physical
`stress, malnutrition, or pregnancy, should be
`excludeds’ 8’ 9
`.
`After establishment of the diagnosis of Cushing’s
`syndrome, the endocrinologist must determine the
`source of the hypercortisolemia. A low or unde—
`tectable ACTH level should raise the suspicion for
`an ACTH-independent source, and imaging of the
`adrenal glands should be performed. An elevated
`or
`nonsuppressed
`(“inappropriately
`normal”)
`ACTH level reflects an ACTH-dependent source of
`hypercortisolemia. A combination of noninvasive
`biochemical
`testing (high-dose dexamethasone
`testing or CRH stimulation), pituitary magnetic
`resonance imaging, and, often,
`inferior petrosal
`sinus sampling for ACTH may be necessary to
`determine if the source is an ACTH-secreting
`1
`pituitary adenoma or a nonpituitary tumor with
`ectopic ACTH production.5‘
`
`Treatment Options for Cushing’s Syndrome
`
`Treatment of Cushing’s syndrome is dependent
`on the identified source of the disorder. Patients
`with cortisol-secreting adrenal adenomas are usu-
`ally cured with unilateral adrenalectomy, whereas
`patients with adrenocortical carcinoma often have
`persistent or recurrent disease after surgery due
`to local invasion or metastases. In patients with
`the ectopic ACTH syndrome, initial treatment is
`directed at
`the underlying neoplasm. Medical
`therapies are often needed in patients with persis-
`tent hypercortisolemia.
`In patients with Cushing’s disease (ACTH-
`secreting pituitary adenomas), who make up the
`majority of patients with Cushing’s syndrome,
`the primary treatment modality is transsphenoi-
`dal surgery, which results in remission rates of
`50—80%.3’ ”T14 If there is failure to attain remis-
`sion after initial surgery or if the disease recurs
`later, second-line interventions include repeat
`surgery,
`radiotherapy, bilateral adrenalectomy,
`or pharmacologic therapy?“ 13‘ 15
`Medical therapy for hypercortisolemia is pro-
`vided to patients who are unable to undergo
`surgery because of another illness,
`to patients
`
`3
`
`

`

`,v
`
`therapy
`
`
`
`
`
`endometrialhyperplasra,nausea
`
`Expensive,difficulttomonitor
`
`
`
`Hypokalemia,adrenalinsufficiencv
`
`
`type5receptorsGlucocorticoidreceptorantagonism
`
`322
`
`PHARMACOTHERAPY Volume 33, Number 3, 2013
`
`
`
`Intravenousadministrationonly
`
`Sedation
`
`
`
`
`
`Limitedavailabilityandefficacy
`
`withanotheragentSlowonset
`
`
`
`
`
`
`
`Rash,fever,dizziness,depression
`
`
`
`Druginteractionsduetoinhibitionof CYP3A
`
`
`
`Hepatotoxicity,hypogonadism
`
`
`
`Inhibitionofcortisolsynthesis:11B-hydroxlase,
`
`Comments
`
`
`
`AdverseReactions
`
`MechanismofAction/SiteofAction
`
`DoseRange
`
`200mgb.i.d.—400mg
`
`t.i.d.
`
`Limitedavailability,oftencombined
`
`
`
`
`
`Hypertension,edema,hypokalemia,
`
`
`
`
`
`Gastrointestinaleffects,ataxia,
`
`hirsutism
`
`
`
`17-hydroxylase,andC17,20lyseInhibitionofcortisolsynthesis:
`
`250—1500mgq.i.d.
`
`1IB-hydroxylaseInhibitionofcortisolsynthesis:
`
`500—3000mgt.i.d.
`
`confusion
`
`
`
`IlB—hydroxylaseandcholesterolsidechains
`
`who have failed to achieve remission with other
`treatment modalities, as a bridge to radiotherapy
`or surgery, or as a palliative option. A number
`of potential
`targets exist
`for medical
`therapy,
`including inhibition of steroidogenesis,
`inhibi-
`tion of ACTH secretion, and steroid receptor
`antagonism. The most commonly used agents
`are steroidogenesis inhibitors such as ketoconaz-
`ole, metyrapone, and mitotane. Other agents in
`this class include aminoglutethimide and etomi-
`date (Table 2)}, 13, 16-20
`Steroidogenesis inhibitors are considered adre-
`nal—directed medical
`therapy because they con—
`trol cortisol production by directly decreasing
`adrenal hormone production. Ketoconazole is
`the most commonly used steroidogenesis inhibi-
`tor because of its availability and relatively rapid
`onset of action. Kctoconazole was developed as
`an antifungal drug. It inhibits cortisol synthesis
`by preventing cholesterol Side chain cleavage,
`inhibiting cytochrome P450 enzyme 17,20-lyase,
`and inhibiting llB-hydroxylase,
`the
`enzyme
`involved in the final step of cortisol synthesis.
`Its major limiting adverse effect is elevated liver
`enzyme levels, which occur in up to 10% of
`patients. It can also cause hypogonadism in meg
`because of inhibition of testosterone synthesis.
`’
`19«21
`
`Metyrapone blocks the production of corti-
`sol
`through inhibition of
`IIB-hydroxylase.
`This effectively reduces hypercortisolemia, but
`because metyrapone is
`specific for
`a
`single
`enzyme late in the steroid biosynthesis path-
`way,
`there is often a dramatic rise in steroids
`formed proximal
`to IIB-hydroxylase, particu-
`larly 11—deoxycortisol, a mineralocorticoid that
`causes the frequent adverse effects of hypokale-
`mia, edema, and hypertension. An increase in
`adrenal
`androgens
`can
`cause hirsutism in
`women. In an effort
`to limit
`the accumulation
`of precursor
`steroids, metyrapone
`is often
`used in combination with other medical thera~
`pies such as ketoconazole. It is currently avail-
`able in the United States directly from the
`manufacturer.16' 19’ 20’ 22
`Mitotane is used most often for the manage-
`ment of adrenocortical carcinoma.
`It works
`through the inhibition of multiple enzymes19
`and, unlike other agents,
`is directly cytotoxic
`to adrenocortical cells. Undesirable features of
`
`mitotane are its delayed onset of action and
`dose-limiting
`gastrointestinal
`effects.
`Serious
`neurologic effects, including ataxia, vertigo, and
`confusion, also occur at higher doses. These
`adverse effects limit the tolerability of mitotane,
`
`4
`
`
`
`Table2.DrugTherapyfortheTreatmentofCushing’sSyndrome3‘13’1640
`
`
`
`
`
`
`
`
`
`
`
`IIItrialsLimitedefficacyLimitedefficacyInphase
`
`
`
`
`
`Nausea,hypotension
`
`
`
`Nausea,hypotension
`
`Hyperglycemia
`
`
`
`
`
`IIB-hydroxylaseand18hydroxylaseInhibitionofcortisolsynthesis:
`0.1—0.3mg/kg/hr
`
`ylase,l7hydroxylase,
`
`
`lIb-hvdrox
`
`andC17,20IyseDopamineagonist
`
`
`
`Dopamineagonist
`
`2.5—40mg/day1—2mg/wk
`
`Somatostatinanalog,somatostatin
`
`
`600—900ugbid.
`
`300—1200mg/day
`
`
`
`
`
`Inhibitionofcortisolsynthesisandsidechain
`cleavage:
`
`2000mgqid.
`
`
`
`250mgb.i.d.-—
`
`Aminoglutethimide
`
`Ketoconazole
`
`Drug
`
`Metyrapone
`
`Mitotane
`
`
`
`CYP=cytochromeP450.
`
`BromocriptineCabergolinePasireotide
`
`Mifepristone
`
`Etomidate
`
`4
`
`

`

`MIFEPRISTONE FOR CUSHING’S SYNDROME Morgan et al
`
`323
`
`and it must often be used in combination
`
`with another drug to attain more rapid control
`of hypercortisolemia.3’ 19’ 23
`Aminoglutethimide and etomidate are steroi-
`dogenesis inhibitors that are used infrequently
`due
`to
`their
`limitations. Aminoglutethimide,
`which works by inhibiting the conversion of
`cholesterol
`to pregnenolone,
`is neither particu-
`larly effective as monotherapy nor readily avail-
`able in the United Statesm’
`19 Etomidate, an
`intravenous agent used for anesthesia induction,
`inhibits cholesterol side chain cleavage and 11B-
`hydroxylase.
`It has been used in emergent set-
`tings for the rapid control of hypercortisolemia
`but
`is not practical for routine use due to its
`sedative effects.16' 19’ 24
`
`Drugs that suppress ACTH secretion have
`been investigated for use in the management of
`Cushing’s disease. Among these are dopamine
`agonists and somatostatin analogs. Dopamine
`agonists are potentially attractive agents for the
`treatment of Cushing’s syndrome because of
`the potential
`for decreased prevalence of glu-
`cose intolerance and diabetesf" 13’
`16’
`25 but
`results have been variable and few patients
`with Cushing’s syndrome experienced sustained
`improvement after receiving dopamine agonist
`therapy.]3’ 19’ 20 Bromocriptine causes an acute
`decrease in ACTH, although this effect
`is not
`sustained over time with repeated dosing.
`Octreotide, a somatostatin analog that pre-
`dominantly acts on type 2 somatostatin recep-
`tors,
`is
`largely ineffective in lowering ACTH
`levels.13‘
`16 A newer multiligand somatostatin
`analog, pasireotide (SOM230), has been demon—
`strated to inhibit ACTH release in human corti-
`
`cotroph cells through interaction with type 5
`somatostatin receptorsfi‘
`16’
`26
`Its use has
`resulted in reduced urine free cortisol levels and
`improved features of Cushing’s
`syndrome in
`phase II and III studies, but it appears to have
`the undesirable effect of hyperglycemia,” 27
`possibly caused by direct
`inhibition of insulin
`and incretin hormone secretion.
`
`Mifepristone
`
`Mifepristone (RU486), a derivative of the syn-
`thetic progestin norethindrone, was discovered
`in the 19805 at the French pharmaceutical com-
`pany Roussel-Uclaf as part of a special research
`project
`to
`develOp
`antiglucocorticoid
`com-
`pounds.28 Its antiprogestin effects were quickly
`recognized, and it was developed as an abortifa-
`cient because of its effectiveness in pregnancy
`
`termination, particularly when combined with a
`prostaglandin. Other investigated uses that take
`advantage of its antiprogesterone activity include
`the treatment of meningioma and breast cancer.
`Unfortunately,
`research on mifepristone has
`been hindered by the controversy surrounding
`its use as an abortion pill.28
`Mifepristone is a selective antagonist of the
`progesterone receptor at lower doses and blocks
`the glucocorticoid receptor at higher doses.18
`Mifepristone occupies glucocorticoid receptors
`with an affinity that is 4-fold higher than that of
`dexamethasone and 18-fold higher than that of
`cortisol.28 After binding,
`it
`inhibits transcrip-
`tional activation of the glucocorticoid receptor,
`thereby decreasing the physiologic effects of hy-
`percortisolemia. It blocks both central (negative
`feedback on CRH and ACTH) and peripheral
`actions of cortisol.28 Antagonism of negative
`feedback of cortisol results in increased circulat-
`ing ACTH and cortisol levels.28’ 20 It has little
`affinity for
`the mineralocorticoid receptor and
`estrogen receptors but is a weak antiandrogen.
`Mifepristone is also a weak glucocorticoid ago-
`nist,
`roughly
`1/250th
`of
`that
`of
`cortisol,
`although this weak effect is unlikely to prevent
`adrenal insufficiency.2830
`
`Pharmacolzinetics and Pharmacodynamics
`
`readily absorbed after oral
`Mifepristone is
`ingestion with a bioavailability exceeding 30%.31
`Time to peak plasma concentrations after oral
`administration of a single dose is
`1—2 hours,
`increasing to 1—4 hours with repeated doses.
`Food increases
`the plasma concentrations of
`mifepristone. Mifepristone has three active metab-
`olites, all of which have high affinity and antago_
`nism for the glucocorticoid receptor (~50% of
`that of mifepristone). Cytochrome P450 (CYP)
`3A is involved in the metabolism of mifepristone.
`Two of the known active metabolites are a result
`of demethylation, whereas the third is a result of
`hydroxylation. Mean plasma concentration of
`these metabolites peaks between 2 and 8 hours
`after multiple doses of the drug and eventually
`exceeds that of mifepristone.18 Therefore, drug
`interactions affecting enzyme metabolism may
`affect the degree of antagonism of the glucocorti-
`coid receptor. Time to steady state with repeated
`daily dosing is 2 weeks. Mifepristone has a very
`long elimination half-life of 85 hours
`after
`repeated dosing.18
`exist
`interactions
`Significant
`drug—drug
`because of mifepristonc’s effects on several CYP
`
`5
`
`

`

`324
`
`PHARMACOTHERAPY Volume 33, Number 3, 2013
`
`enzymes. For example, CYP3A is involved in the
`metabolism of mifepristone and mifepristone
`also both inhibits and induces CYP3A. There-
`fore, drugs
`that are metabolized by CYP3A
`should
`be
`avoided
`or
`used With
`caution
`(Table 3). When a once-daily dose of mifepri-
`stone 1200 mg was coadministered with simvast-
`atin 80 mg for 10 days in healthy volunteers,
`there was an 18-fold increase in the maximum
`lasma concentration (Cmax) of simvastatin acid
`and a 7-fold increase in the Cmax of simvastatin,
`significantly increasing the risk of toxicity of
`this drug. 8 Drugs
`that
`inhibit CYP3A can
`increase mifepristone levels, and a dose reduc-
`tion of mifepriStone may be required (Table 3).
`Coadministration 0f mifepristone and CYP3A in-
`ducers has not been studied. Other enzymes
`affected by mifepristone are CYP2C8/2CQ and
`CYP2B6. Drugs metabolized by these pathways
`should be used with caution (Table 4). The
`Cmax of iluvastatin 40 mg was increased 1.76—
`fold when coadministered with mifepristone
`1200 mg.18 Because of the long half-life of mife-
`pristone and time to reach steady state, dosage
`adjustment of drugs with potential
`interactions
`should not occur more frequently than every
`2 weeks. Drugs that are contraindicated for use
`with mifepristone can be safely initiated 2 weeks
`after the discontinuation of mifepristone.18
`Mean exposure (plasma concentration over
`time)
`to mifepristone (evaluated with multiple
`1200-mg doses for 7 days) increased by 31% in
`patients with a creatinine clearance less than
`30 ml/min compared to patients with normal
`renal
`function (creatinine clearance > 60 my
`min). There was large variability among subjects
`
`Table 4. Drugs that Interact with Mifcpristone Through
`
`CYP2C8/2C9 and CYPZB6'“
`
`Drugs Metabolized
`Drugs Metabolized by
`
` CYP2C8/2C9 by CYPZBé
`Use lowest dose and
`Not studied—use lowest
`monitor closely
`dose
`Fluvastatin
`Bupropion
`NSAIDS
`Efavirenz
`Warfarin
`Repaglinide
`CYI’ = cytochrome P450; NSAIDs = nonsteroidal anti-inflamma-
`tory drugs,
`
`in the exposure of mifepristone and its metabo-
`lites. The pharmacokinetics of mifepristone in
`patients with moderate hepatic impairment was
`found to be similar to that in patients with nor—
`mal hepatic function. The pharmacokinetics in
`patients with severe hepatic impairment has not
`been studied.18
`
`Clinical Efficacy
`
`Initial information regarding efficacy of mife—
`pristone for use in Cushing’s syndrome came
`from case reports. In 1985, a patient with Cush-
`ing’s syndrome secondary to ectopic ACTH secre-
`tion was treated with mifepristone.32 The initial
`dosage was 5 mg/kg/day because it was known
`that a dosage of 6 mg/kg/day prevents morning
`adrenal suppression from dexamethasone 1 mg.
`The dosage was increased in 5-mg/kg/day incre-
`ments every 1—2 days to a maximum of 20 mg/kg/
`day. Treatment resulted in resolution of clinical
`effects of Cushing’s syndrome, redistribution of
`fat, and improvement
`in hyperglycemia and
`
`Limit mifepristone to 300 mg/day
`
`Azole antifungals
`Protease inhibitors
`
`Table 3. Sample List of Drugs or Foods that Interact with Mifcpristone Through CYP4503A“l
`CYP3A
`Drugs Metabolized
`CYP3A Inhibitors
`Induccrs
`by CYP3A
`Do not use (has not
`Use alternative drug or administer
`been studied)
`lowest dose and/or decrease frequency
`Rifabutin
`Cyclosporine
`Phenobarbital
`Dihydroergotamine
`Ergotamine
`Fentanyl
`Pimozide
`Quinidine
`Sirolimus
`Tacrolimus
`Simvastatin
`Lovastatin
`
`Macrolides
`Mibefradil
`Nefazodone
`Conivaptan
`Caution—use lowest effective dose of mifepristone
`lmatinib
`Aprepitant
`Ciprolloxacin
`Grapefruit juice
`Nondihydropyridine CCBs
`CCBs = calcium channel blockers; CYP = cytochrome P450.
`
`Phenytoin
`Carbamachine
`St. john’s wort
`Rifampin
`
`6
`
`

`

`MIFEPRISTONE FOR CUSHING’S SYNDROME Morgan et al
`
`325
`
`hypertension. Fasting glucose and 2-hour glucose
`levels after ingestion of glucose 100 g normalized
`with the highest doses.
`In contrast,
`levels of
`ACTH, serum cortisol, and urine free cortisol
`remained
`elevated. Treatment was
`stopped
`because of the limited availability of mifepristone.
`A cases series of 10 patients with Cushing’s
`syndrome treated with mifepristone was pub-
`lished in 1989.28’ 33’ 34 Six of the 10 patients
`who received mifepristone 5—22 mg/kg/day had
`their symptoms of hypercortisolemia alleviated.
`Patients” hypertension improved, with reduction
`in mean blood pressure. Carbohydrate metabo-
`lism also improved, as evidenced by reduction
`in 2-hour plasma glucose levels during oral glu-
`cose tolerance testing. Two of three men com-
`plained of gynecomastia and impotence, whereas
`only two patients developed adrenal
`insuffi-
`ciency.3“1
`Subsequent studies ensued to elucidate the
`drug’s
`antiglucocorticoid effects
`and relative
`safety for long-term treatment. In 1994, mifepri-
`stone 200 mg/day was administered to 10 healthy
`volunteers for 8 consecutive days. The investiga-
`tors found heightened activation of the hypotha-
`lamic-pituitary-adrenal axis with elevations in
`plasma cortisol, urinary cortisol, and ACTH lev-
`els, although there was no evidence of clinical
`symptoms. Adrenocortical reserves seemed to be
`preserved as evidenced by the ability to further
`increase circulating cortisol with ACTH stimula-
`tion.29 In 1995, the antiglucocorticoid effects of
`mifepristone were evaluated in eight healthy
`men.35 This study evaluated subjects during the
`infusion of cortisol after the ingestion of mifepri-
`stone 600 mg or placebo and during the infusion
`of normal saline with placebo or mifepristone.
`The increase in plasma glucose levels seen with
`cortisol
`infusion plus placebo was not demon-
`strated when cortisol was administered after mife-
`pristone ingestion. Although this study involved
`only healthy men and was limited by the small
`number of patients studied, it supported the con-
`cept that mifepristone may suppress the effects of
`hypercortisolemia on glucose.
`In 2009,
`results from a retrospective study
`were published: 20 patients with Cushing’s syn-
`drome due to malignant or benign causes were
`treated with oral mifepristone 400—2000 mg/day
`for 5 days to 24 months.36 The median initiation
`dose was 400 mg with a median maximum dose
`of 600 mg/day. Eight of the 12 patients with
`adrenocortical carcinoma, most of whom had
`failed surgery, cytotoxic chemotherapy, and/or
`mitotane as well as other drug therapy, had
`
`rapid improvement within the first month of ini-
`tiation of mifepristone. Seven
`of
`these
`12
`patients developed hypokalemia, although this
`required cessation of therapy in only 1 patient.
`All three patients with ectopic ACTH secretion
`had improvement in clinical symptoms, and the
`two patients with diabetes
`required a
`rapid
`decrease in their insulin dose. Three of four
`patients with Cushing’s disease had improve—
`ment
`in clinical
`signs of hypercortisolemia;
`levels of ACTH and cortisol increased in all four
`
`treated had bilateral
`patients. The last patient
`adrenal hyperplasia. Hypertension and signs of
`hypercortisolemia improved within 3 months of
`therapy, and hemoglobin AIC (AIC) was reduced
`from 7.1% to 6.4%. Overall,
`the investigators
`found improvement in clinical symptoms in 15
`of the 20 patients along with improvement
`in
`blood glucose levels in 4 of 7 patients with
`hyperglycemia. Serious adverse effects included
`signs of adrenal insufficiency in 3 patients and
`hypokalemia in 11 patients.
`Recently, an open-label, 24-week, multicenter
`clinical study evaluated safety and efficacy of mi-
`fepristone for
`the treatment of Cushing’s syn-
`drome; the results led to the drug’s approval by
`the FDA in 2012.37 A separate open-label exten-
`sion of this trial is ongoing. The study enrolled
`50 patients with continued biochemical and clin-
`ical evidence of hypercortisolemia after
`failed
`multimodality therapy, largely with surgery and/
`or radiotherapy. Endogenous hypercortisolemia
`was defined as elevated urine free cortisol levels
`measured from at least two 24-hour collections
`and elevated late-night salivary cortisol
`levels
`and/or lack of suppression with dexamethasone.
`Inclusion criteria were associated type 2 diabe-
`tes,
`impaired glucose tolerance diagnosed from
`an oral glucose tolerance test, or hypertension
`with at
`least
`two other signs or symptoms of
`Cushing’s syndrome. Exclusion criteria included
`an A1C greater
`than 11%, poorly controlled
`hypertension
`(blood
`pressure > 170/110 mm
`Hg), use of drugs to treat hypercortisolemia
`within 1 month, uncorrected hypokalemia, and
`uncontrolled thyroid disease. Women with evi-
`dence of endometrial hyperplasia, cancer, or pol-
`yps
`and those with an intact uterus who
`required anticoagulation or had hemorrhagic
`disorders were also excluded. Initiation or addi—
`
`impaired glucose
`tions to therapy for diabetes,
`tolerance, depression, hyperlipidemia, or hyper-
`tension were not allowed, with the exception of
`the addition of a mineralocorticoid antagonist
`for the treatment of hypokalemia.
`
`7
`
`

`

`326
`
`PHARMACOTHERAPY Volume 33, Number 3, 2013
`
`Fifty patients were enrolled at 17 centers.
`Forty-three of the patients had Cushing’s dis-
`ease, 4 had ectopic ACTH tumors, and 3 had
`adrenal carcinoma. Patients were separated into
`a diabetes cohort (29 patients) and a hyperten-
`sion cohort (21 patients), and efficacy was eval-
`uated in each cohort. Treatment was initiated
`with mifepristone 300 mg/day, which could be
`increased to 600 mg after
`2 weeks. Further
`increases by 300-mg increments were allowed
`every 4 weeks with a maximum dosage of 900—
`1200 mg/day based on weight. The mean :1: SD
`dose at the final study visit was 732 :l: 366 mg/
`da .
`lln the diabetes cohort, patients were required
`to have received stable antidiabetic regimens
`before enrollment and the regimens could not
`be advanced during the study. The primary end
`oint was a reduction in the area under the
`curve (AUC) for glucose of at least 25% as mea-
`sured with an oral glucose tolerance test. This
`test was chosen because it could be used for
`evaluation of patients with either diabetes or
`impaired glucose tolerance. In the hypertension
`cohort, the primary end point was the change in
`diastolic blood pressure from baseline to week
`24 with response defined as a reduction of at
`least 5 mm Hg. Secondary end points included
`other clinical responses compared to baseline as
`evaluated by an independent data review board
`at weeks 6, 10, 16, and 24. This board evaluated
`glucose homeostasis, blood pressure, lipid levels,
`weight and body composition, clinical appear-
`ance,
`strength,
`and neuropsychological
`and
`quality of life scales using validated methods.
`Thirty-four patients completed the study: 20 in
`the diabetes cohort and 14 in the hypertension
`cohort. Reasons
`for withdrawal were adverse
`events (7), death (2), withdrawn consent (5), and
`other (2). Deaths were related to progression of
`underlying malignancy. A modified intent-to-treat
`analysis was used. All patients who received at
`least 30 days of the study drug were included in
`the analysis. Of the 25 patients in the diabetes
`cohort who received mifepristone for at
`least
`30 days, 60% (15 patients) responded with a 25%
`or greater reduction in glucose AUC during stan-
`dard glucose tolerance testing (p<0.0001). Eigh-
`teen of the patients (72%) in the diabetes cohort
`had at least a 25% reduction in glucose AUC or
`were able to reduce antidiabetic therapy. Mean
`reduction in AlC was
`1.1% from baseline
`(p<0.001), and 6 of 12 patients with an AIC
`greater than 7% at baseline had an AIC of 6% or
`less by the end of the trial. Mean :l: SD fasting 8
`
`from
`reduced
`levels were
`glucose
`plasma
`149 :1: 74.7 mg/dl at baseline to 104.7 i 37.5 mg/
`d1 at 24 weeks (p<0.03). The doses of antidiabetic
`drugs were reduced in 7 of 15 patients. Of the 12
`patients receiving insulin, 5 were able to reduce
`their insulin doses by at least 50%.37
`In the hypertension cohort, 8 (38.1%) of 21
`pat

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