throbber
Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`O R I G I N A L
`
`A R T I C L E
`
`E n d o c r i n e C a r e
`
`Mifepristone, a Glucocorticoid Receptor Antagonist,
`Produces Clinical and Metabolic Benefits in Patients
`with Cushing’s Syndrome
`
`Maria Fleseriu, Beverly M. K. Biller, James W. Findling, Mark E. Molitch,
`David E. Schteingart, and Coleman Gross, on behalf of the SEISMIC Study
`Investigators
`
`Oregon Health & Science University (M.F.), Portland, Oregon 97239; Massachusetts General Hospital
`(B.M.K.B.), Boston, Massachusetts 02114; Medical College of Wisconsin (J.W.F.), Milwaukee, Wisconsin
`53226; Northwestern University Feinberg Medical School (M.E.M.), Chicago, Illinois 60611; University of
`Michigan (D.E.S.), Ann Arbor, Michigan 48109; and Corcept Therapeutics (C.G.), Menlo Park, California 94025
`
`Context: Cushing’s syndrome (CS) is a disorder associated with significant morbidity and mortality
`due to prolonged exposure to high cortisol concentrations.
`
`Objective: Our objective was to evaluate the safety and efficacy of mifepristone, a glucocorticoid
`receptor antagonist, in endogenous CS.
`
`Design and Setting: We conducted a 24-wk multicenter, open-label trial after failed multimodality
`therapy at 14 U.S. academic medical centers and three private research centers.
`
`Participants: Participants included 50 adults with endogenous CS associated with type 2 diabetes
`mellitus/impaired glucose tolerance (C-DM) or a diagnosis of hypertension alone (C-HT).
`
`Intervention: Mifepristone was administered at doses of 300-1200 mg daily.
`
`Main Outcome Measures: We evaluated change in area under the curve for glucose on 2-h oral
`glucose test for C-DM and change in diastolic blood pressure from baseline to wk 24 for C-HT.
`
`Results: In the C-DM cohort, an area under the curve for glucose (AUCglucose) response was seen in 60%
`of patients (P ⬍ 0.0001). Mean ⫾ SD glycated hemoglobin (HbA1c) decreased from 7.43 ⫾ 1.52% to
`6.29 ⫾ 0.99% (P ⬍ 0.001); fasting plasma glucose decreased from 149.0 ⫾ 75.7 mg/dl (8.3 ⫾ 4.1 mmol/
`liter) to 104.7 ⫾ 37.5 mg/dl (5.8 ⫾ 2.1 mmol/liter, P ⬍ 0.03). In C-HT cohort, a diastolic blood pressure
`response was seen in 38% of patients (P ⬍ 0.05). Mean weight change was ⫺5.7 ⫾ 7.4% (P ⬍ 0.001) with
`waist circumference decrease of ⫺6.78 ⫾ 5.8 cm (P ⬍ 0.001) in women and ⫺8.44 ⫾ 5.9 cm (P ⬍ 0.001)
`in men. Overall, 87% (P ⬍ 0.0001) had significant improvement in clinical status. Insulin resistance,
`depression, cognition, and quality of life also improved. Common adverse events were fatigue, nausea,
`headache, low potassium, arthralgia, vomiting, edema, and endometrial thickening in women.
`
`Conclusions: Mifepristone produced significant clinical and metabolic improvement in patients
`with CS with an acceptable risk-benefit profile during 6 months of treatment. (J Clin Endocrinol
`Metab 97: 2039 –2049, 2012)
`
`ISSN Print 0021-972X ISSN Online 1945-7197
`Printed in U.S.A.
`Copyright © 2012 by The Endocrine Society
`doi: 10.1210/jc.2011-3350 Received December 12, 2011. Accepted March 8, 2012.
`First Published Online March 30, 2012
`
`Abbreviations: AE, Adverse event; AI, adrenal insufficiency; AUCglucose, area under the
`curve for glucose; BDI, Beck Depression Inventory; CD, Cushing’s disease; C-DM, patients
`with CS and T2DM/IGT; C-HT, patients with CS and a diagnosis of HTN; CI, confidence
`interval; CS, Cushing’s syndrome; DBP, diastolic blood pressure; DRB, data review board;
`ET, early termination; FPG, fasting plasma glucose; HbA1c, glycated hemoglobin; HDL-C,
`high-density lipoprotein cholesterol; HOMA-IR, homeostatic model assessment of insulin
`resistance; HTN, hypertension; IGT, impaired glucose tolerance; mITT, modified intent-to-
`treat; MRI, magnetic resonance imaging; oGTT, oral glucose tolerance test; T2DM, type 2
`diabetes mellitus.
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`jcem.endojournals.org
`
`2039
`
`1
`
`TEVA1029
`
`

`

`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`2040
`
`Fleseriu et al.
`
`Mifepristone and Cushing’s Syndrome
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`Cushing’s syndrome (CS), is a serious endocrine disor-
`
`der that may be caused by a pituitary [Cushing’s
`disease (CD)] or nonpituitary (ectopic) ACTH-secreting
`tumor or by an adrenal neoplasm. If inadequately treated,
`CS is associated with a 3.8- to 5.0-fold higher mortality
`than the general population (1–3). Regardless of cause,
`surgery is usually the treatment of choice; however,
`complete removal of the neoplasm may not be possible
`(4, 5). Adjunctive radiotherapy for CD may take years
`to control excess cortisol (6). Laparoscopic bilateral
`adrenalectomy represents another treatment option.
`No medical treatments were approved by the U.S. Food
`and Drug Administration for CS when the study was
`conducted, but off-label use of several medications is
`common, including dopamine agonists, somatostatin
`analogs, and the adrenal steroidogenesis inhibitors (ke-
`toconazole, metyrapone, mitotane, and etomidate) (4,
`7). Ketoconazole and mitotane are effective in many
`patients, but in CD, doses may need progressive in-
`creases due to escape from cortisol blockade. The tol-
`erability of these drugs, especially at higher doses, limits
`their use in some patients (8, 9).
`(11␤-[P-(dimethylamino)phenyl]-17␤-
`Mifepristone
`hydroxy-17-(1-propynyl)estra-4,9-dien-3-one) is a pro-
`gesterone receptor antagonist that has glucocorticoid re-
`ceptor antagonist activity at higher concentrations, with
`more than three times the binding affinity for the gluco-
`corticoid receptor than dexamethasone (10, 11). It does
`not bind to the mineralocorticoid receptor (9). Case re-
`ports and small retrospective studies of mifepristone treat-
`ment in CS document improvements in abnormal glucose
`metabolism, psychiatric symptoms, and the somatic
`changes associated with CS; hypokalemia was the most
`commonly reported side effect (9, 12–25). Based on these
`preliminary findings, an open-label, prospective, multi-
`center, 6-month study of the safety and efficacy of mife-
`
`pristone was conducted in patients with endogenous CS
`refractory to other therapies.
`
`Patients and Methods
`
`Patients
`Adults with confirmed endogenous CS who had associated
`type 2 diabetes mellitus (T2DM), impaired glucose tolerance
`(IGT), or a diagnosis of hypertension (HTN) were enrolled (Fig.
`1). Endogenous hypercortisolism was defined as elevated urinary
`free cortisol on at least two 24-h collections and elevated late-
`night salivary cortisol and/or lack of suppression with dexameth-
`asone. T2DM was defined as a fasting plasma glucose (FPG) of
`at least 126 mg/dl (ⱖ7.0 mmol/liter) on two measurements or a
`2-h plasma glucose of at least 200 mg/dl (ⱖ11.1 mmol/liter) after
`a 75-g oral glucose tolerance test (oGTT), and IGT was defined
`as 2-h oGTT glucose value of 140 –199 mg/dl (7.8 –11.0 mmol/
`liter). HTN was defined as systolic blood pressure over 140 mm
`Hg and/or diastolic blood pressure (DBP) over 90 mm Hg or
`pharmacologically treated HTN.
`At least two of the following signs or symptoms of Cushing’s
`were also necessary for inclusion: Cushingoid appearance (moon
`facies, dorsocervical fat pad, and plethora), increased body
`weight or central obesity, proximal muscle weakness, low bone
`mineral density (T score ⬍ ⫺1.0), psychiatric symptoms, and
`skin changes (hirsutism, violaceous striae, or acne).
`Patients were excluded for poorly controlled diabetes mellitus
`[glycated hemoglobin (HbA1c) ⱖ 11%], poorly controlled HTN
`(⬎170/110 mm Hg), use of drugs to treat hypercortisolism within
`1 month of baseline (mitotane for adrenal carcinoma was allowed
`ifonstabledose ⱖ1monthbeforeentry),uncorrectedhypokalemia,
`or uncontrolled hypothyroidism or hyperthyroidism; also excluded
`were women with a uterus who required anticoagulants or had
`hemorrhagic disorders, endometrial hyperplasia, carcinoma, or
`polyps. Increases or additions of antihyperglycemic medications
`during the study were not permitted for patients with T2DM/IGT.
`For patients with HTN, increases or additions of antihypertensive
`medications were not permitted with the exception of mineralo-
`corticoid receptor antagonists, which were allowed for treating hy-
`pokalemia, a known side effect of mifepristone (9). Changes in or
`initiation of antidepressant or lipid-lowering medications were not
`allowed.
`
`The study was approved by the insti-
`tutional review board at each center and
`was registered with www.clinicaltrials.
`gov (NCT00569582). All patients pro-
`vided written informed consent.
`
`Design
`This was a 24-wk, open-label, multicenter
`study of mifepristone administered as a single
`daily oral dose. Treatment began at 300 mg/d;
`if no significant clinical improvement was
`noted by the investigator, doses could be in-
`creased to 600 mg/d on d 14, 900 mg/d at wk
`6, and 1200 mg/d at wk 10. Dose interruption
`and reduction were specified in the protocol
`for the following adverse events (AEs): adre-
`nal insufficiency (AI), severe hypokalemia,
`
`FIG. 1. Enrollment: ITT/safety population.
`
`2
`
`

`

`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`jcem.endojournals.org
`
`2041
`
`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`and vaginal bleeding. Temporary glucocorticoid rescue for sus-
`pected AI was also allowed.
`
`Assessments
`The primary endpoint for patients with CS and T2DM/IGT
`(C-DM cohort) was the change in area under the curve for glucose
`(AUCglucose) on oGTT from baseline to wk 24. Response was de-
`fined as at least a 25% decrease in AUCglucose, an amount consid-
`ered clinically meaningful improvement in glucose control (26).
`AUCglucose was used because both patients with T2DM and pa-
`tients with IGT were enrolled, and HbA1c and FPG would not be
`uniformlyapplicable.Inpatientsreceivingmedicationsfordiabetes,
`administration occurred before the oGTT (other than short-acting
`insulin and glucagon-like peptide-1 analogs). The primary endpoint
`for patients with CS and a diagnosis of HTN (C-HT cohort) was the
`change in DBP from baseline to wk 24; response was defined as DBP
`decrease of at least 5 mm Hg (mean of two sequential readings).
`Patients with both T2DM/IGT and HTN were included only in the
`C-DM cohort.
`Key secondary endpoints included clinical response graded by
`an independent data review board (DRB) at wk 6, 10, 16, and 24
`compared with baseline. The DRB consisted of three CS experts
`who evaluated the following assessments: glucose homeostasis,
`blood pressure, lipids, weight and body composition change, clin-
`ical appearance (acne, hirsutism, striae, and Cushingoid appear-
`ance) (27, 28) as rated by the investigators, strength, and neuro-
`psychological [Beck Depression Inventory (BDI)-II and Trail
`Making Test] (29–31) and quality of life [Short-Form 36 Health
`Survey version 2 (SF-36)] (32) parameters. The DRB also reviewed
`standardized photographs of 34 consenting patients. Visit number
`after baseline and mifepristone dose were blinded. Each DRB mem-
`ber categorized patient overall status at follow-up visits as worse
`(⫺1), unchanged (0), or having clinically significant improvement
`(⫹1) from baseline. If the reviewers’ median score was ⫹1, the
`patient was considered to have clinical improvement.
`Blood, urine, and saliva samples were analyzed by a central
`laboratory (Quest Diagnostics, Collegeville, PA). AUCglucose and
`AUCinsulin were determined using the linear trapezoidal rule;
`homeostatic model assessment of insulin resistance (HOMA-IR)
`was calculated (33). Urinary and salivary cortisol levels were
`assayed with liquid chromatography tandem mass spectrometry
`[normal ranges, respectively, are 2– 42.4 ␮g/24 h (5.5–117
`nmol/24 h) and ⱕ0.09 ␮g/dl (2.5 nmol/liter)]; serum cortisol
`[normal range is 4 –22 ␮g/dl (110 – 607 nmol/24 h)], and ACTH
`(normal range is 5–27 pg/ml (1.1–5.9 pmol/liter) for females and
`7–50 pg/ml (1.5–11 pmol/liter) for males] were measured with
`immunochemiluminometric assay.
`AEs were reviewed every visit, and patients were monitored
`with vital signs, physical exams, and blood tests; transvaginal
`ultrasounds were conducted at baseline, wk 24 [or early termi-
`nation (ET)], and 6 wks after last dose. Pituitary magnetic res-
`onance imaging (MRI) was performed at screening and at wk 10
`and 24 (or ET) in patients with CD. Body composition was mea-
`sured using dual-energy x-ray absorptiometry at baseline and wk
`24 or ET using Hologic (Bedford, MA) or GE Lunar (Madison,
`WI) instruments; results were submitted to a central reading site
`for quality control and analysis.
`
`Statistics
`Patients who took at least one dose of study medication com-
`prised the safety population (n ⫽ 50). A modified intent-to-treat
`
`(mITT) population (patients who received ⱖ30 d of study med-
`ication) was used for analyses of efficacy (n ⫽ 46). The completer
`population included participants who completed through wk 24
`and were at least 80% compliant with study medication (n ⫽ 33).
`Because there was no placebo group in this study, a responder
`analysis was conducted. Responder rates were tested against an
`a priori threshold of 20%, which was chosen based on very low
`spontaneous response rates in this patient population (⬍5%)
`(34). The null hypothesis was to be rejected if the lower bound
`of the one-sided binomial 95% confidence interval (CI) of re-
`sponder rates was over 20%. Because mifepristone blocks rather
`than lowers cortisol, alternative quantitative endpoints (other
`than cortisol) were assigned at study entry based on inclusion in
`either C-DM or C-HT cohorts. Two abnormal oGTTs were re-
`quired for inclusion in the C-DM group; patients with a diagnosis
`of HTN and without T2DM/IGT were included in the C-HT
`group. For statistical analysis, response was defined as at least
`25% reduction in AUCglucose for C-DM patients or at least 5 mm
`Hg reduction in DBP in C-HT patients comparing baseline with
`wk 24/ET. For patients who did not complete the study or have
`an ET visit, the last available data were used. ANOVA and t tests
`were used for analyses of other endpoints. Nonparametric sta-
`tistical testing was employed for nonnormally distributed data.
`Change in oGTT curves over the course of the study was modeled
`by a hierarchical linear mixed model that took into consideration
`the correlation within subjects. SAS statistical software versions
`9.1.3 and 9.2 (Cary, NC) were used. Data are shown as mean ⫾
`SD unless otherwise stated.
`
`Results
`
`Patients
`From January 2008 to January 2011, 50 patients with CS
`were enrolled at 17 U.S. centers; 34 completed the study.
`Forty-three patients had a pituitary source of CS (42 with
`unsuccessful pituitary surgery, 18 with pituitary radiation,
`and one without previous surgery), four had ectopic ACTH
`secretion, and three had adrenal cortical carcinoma. Baseline
`characteristics are detailed in Tables 1 and 2. The mean
`dose ⫾ SD at the final study visit was 732 ⫾ 366 mg/d. Twen-
`ty-two subjects received the maximum dose of 1200 mg/d.
`Dose interruptions occurred in 42% of patients with median
`duration of 2 d (range 1–39 d). There were 18 dose reduc-
`tions in 12 patients; reductions occurred most commonly in
`300-mg decrements (317 ⫾ 114 mg).
`
`Primary efficacy analyses
`
`Patients with T2DM/IGT
`In the C-DM mITT population, AUCglucose decreased
`by at least 25% on oGTT in 15 of 25 (60%) patients from
`baseline to wk 24/ET (95% CI lower bound 42%, P ⬍
`0.0001) with a median decrease of 36% [30330.0 mg/
`dl䡠120 min (1683.3 mmol/liter䡠120 min) to 20655.0 mg/
`dl䡠120 min (1146.4 mmol/liter䡠120 min)] as well as com-
`parable reductions in plasma glucose levels (Fig. 2 and
`
`3
`
`

`

`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`2042
`
`Fleseriu et al.
`
`Mifepristone and Cushing’s Syndrome
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`TABLE 1. Demographics and body measurements at baseline (ITT/safety population)
`
`Characteristic
`Sex 关n (%)兴
`Male
`Female
`Race 关n (%)兴
`Black or African-American
`White
`Ethnicity 关n (%)兴
`Hispanic or Latino
`Not Hispanic or Latino
`Age (yr)
`Mean ⫾ SD
`Median
`Range
`Height (cm)
`Mean ⫾ SD
`Median
`Range
`Weight (kg)
`Mean ⫾ SD
`Median
`Range
`BMI (kg/m2)
`Mean ⫾ SD
`Median
`Range
`Waist circumference, cm
`Mean ⫾ SD
`Median
`Range
`Etiology of CS
`CD 关n (%)兴
`Ectopic ACTH 关n (%)兴
`Adrenal cancer 关n (%)兴
`
`C-DM (n ⴝ 29)
`
`C-HT (n ⴝ 21)
`
`Overall (n ⴝ 50)
`
`7 (24.1)
`22 (75.9)
`
`6 (20.7)
`23 (79.3)
`
`2 (6.9)
`27 (93.1)
`
`44.4 ⫾ 13.71
`41.0
`26 –71
`
`168 ⫾ 12.11
`168
`143.5–190.5
`
`105 (33.54)
`102
`61.3–198.7
`
`37.4 (11.18)
`35.1
`24.1– 66.4
`
`124 (21.73)
`120
`97.9 –178.4
`
`24 (82.8)
`3 (10.3)
`2 (6.9)
`
`8 (38.1)
`13 (61.9)
`
`2 (9.5)
`19 (90.5)
`
`2 (9.5)
`19 (90.5)
`
`46.7 ⫾ 8.83
`46.0
`26 – 67
`
`166 ⫾ 8.84
`163
`154.0 –185.4
`
`91.4 (21.10)
`88.2
`62.7–150.5
`
`33.4 (7.44)
`31.8
`24.5–53.6
`
`111 (15.77)
`104
`88.5–153.5
`
`19 (90.5)
`1 (4.8)
`1 (4.8)
`
`15 (30.0)
`35 (70.0)
`
`8 (16.0)
`42 (84.0)
`
`4 (8.0)
`46 (92.0)
`
`45.4 ⫾ 11.85
`45.0
`26 –71
`
`167 ⫾ 10.81
`166
`143.5–190.5
`
`99.5 (29.55)
`92.4
`61.3–198.7
`
`35.7 (9.90)
`33.5
`24.1– 66.4
`
`119 (20.31)
`115
`88.5–178.4
`
`43 (86.0)
`4 (8.0)
`3 (6.0)
`
`The C-DM group included subjects with T2DM and/or IGT at screening and d 1 asdetermined by two or more abnormal oGTT. The C-HT group
`included subjects with a diagnosis of HTN at screening but without T2DM and/or IGT.
`in DBP (95% CI lower bound 21%, P ⬍ 0.05; Table 3).
`Table 3). Similar reductions in AUCglucose were observed
`in the C-DM ITT and completer populations. The most
`Four patients (two responders) received spironolactone
`common doses among responders at wk 24/ET were 600
`during the study; one nonresponder was on spironolac-
`mg (40%) and 1200 mg (40%), followed by 300 mg
`tone at entry and remained on a stable dose throughout the
`(13.3%) and 900 mg (6.7%). In exploratory analyses we
`study.
`found no relationship between the incremental change in
`dose from baseline and AUCglucose (see Supplemental Fig.
`1, published on The Endocrine Society’s Journals Online
`web site at http://jcem.endojournals.org).
`
`Secondary endpoints
`
`Patients with HTN
`In the C-HT mITT cohort, eight of 21) patients (38.1%
`achieved the primary endpoint of at least 5 mm Hg decline
`
`TABLE 2. Biochemistry at baseline (ITT/safety population)
`
`Clinical improvement
`Theoverallclinicalimprovementresponserateasassessed
`by the DRB in the mITT population was 87% (95% CI lower
`bound 76%, P ⬍ 0.0001); response rates were similar in the
`C-DM and C-HT cohorts (Table 3). Thirty-three patients
`
`Biochemistry
`ACTH (pg/ml)
`24 h UFC (␮g/24 h)
`Serum cortisol (␮g/dl)
`Late-night salivary cortisol (␮g/dl)
`
`CD
`
`Ectopic ACTH
`
`Adrenal cancer
`
`Overall
`
`63 (51)
`139 (137)
`21.2 (6.0)
`0.29 (0.29)
`
`153 (140.3)
`2471 (3266)
`42.6 (14.3)
`1.90 (2.26)
`
`812 (559)
`37.4 (15.4)
`1.02 (0.58)
`
`66 (66)
`366 (1049)
`23.9 (10.0)
`0.47 (0.83)
`
`To convert values of ACTH to picomoles per liter, multiply by 0.22; urinary free cortisol (UFC) to nanomoles per 24 h, multiply by 2.759; cortisol to
`nanomoles per liter, multiply by 27.59.
`
`4
`
`

`

`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`jcem.endojournals.org
`
`2043
`
`FIG. 2. Changes in glycemic parameters. A, Significant decreases in AUCglucose were observed in the C-DM cohort from baseline to each
`subsequent visit including wk 24/ET (P ⬍ 0.001). Data are shown as mean ⫾ SD. B, Significant decreases were also seen in plasma and fasting
`plasma glucose (P ⫽ 0.03), as measured by oGTT from baseline to wk 24. The oGTT response curves at each visit were statistically different
`compared with baseline. Mean data are shown. To convert glucose values to millimoles per liter, multiply by 0.0555.
`
`(72%) had a median score of ⫹1 at wk 24 or ET. Eleven
`patients by wk 6 and another six patients by wk 10 had a
`median score of ⫹1 with responses maintained throughout
`the remainder of the study (Initial clinical improvement re-
`sponse by dose and visit are shown in Supplemental Fig. 2).
`Three patients had a nonsustained improvement (median
`score of ⫹1 decreased to 0 at wk 24 or ET). One patient was
`rated as being worse at the final visit (early termination at wk
`10) than at baseline.
`
`Other glucose-related endpoints
`FPG decreased from 149.0 ⫾ 74.7 mg/dl (8.3 ⫾ 4.1
`mmol/liter) at baseline to 104.7 ⫾ 37.5 mg/dl (5.8 ⫾ 2.1
`mmol/liter) at wk 24 (P ⬍ 0.03). Antidiabetic medica-
`tions were reduced in seven of 15 patients. Of 12 pa-
`tients taking insulin, five reduced their daily dose by at
`least half. Eighteen of 25 C-DM patients (72%) had at
`least a 25% reduction from baseline in AUCglucose or a
`reduction in antidiabetic medication (95% CI ⫽ 50.6 –
`
`TABLE 3. Summary of responder analyses (mITT population)
`
`Responder
`关n (%)兴
`
`Nonresponder
`关n (%)兴
`
`Lower bound one-sided
`95% exact binomial CI (%)
`
`Statistics (mITT population)
`C-DM (n ⫽ 25)
`Participants with or without a 25% reduction
`from baseline in AUCglucose at wk 24/ET
`C-HT (n ⫽ 21)
`Participants who had ⱖ5 mm Hg reduction
`from baseline in DBP at wk 24/ET
`C-HT and C-DM with HTN at screening (n ⫽ 40)
`Participants who had ⱖ5 mm Hg reduction
`from baseline in DBP at wk 24/ET
`Participants who had a reduction in
`antihypertensive medications at wk 24/ET
`Participants who had either ⱖ5 mm Hg reduction
`from baseline in DBP or had a reduction in
`antihypertensive medications at wk 24/ET
`Median clinical improvement score of ⫹1
`at any reviewed visitb
`Combined cohorts (n ⫽ 46)
`C-DM (n ⫽ 25)
`C-HT (n ⫽ 21)
`a 95% CI ⫽ 36.1– 68.5.
`b For overall clinical improvement (median DRB score ⫹1) at any reviewed visit, the null hypothesis was to be rejected in favor of the alternative if
`the lower limit of the 95% exact one-sided binomial CI for the responder rate was at least 30%.
`
`41.7
`
`20.6
`
`P value
`
`⬍0.0001
`
`⬍0.05
`
`15 (60)
`
`10 (40)
`
`8 (38.1)
`
`13 (61.9)
`
`17 (42.5)
`
`23 (57.5)
`
`11 (27.5)
`
`29 (72.5)
`
`21 (52.5)a
`
`19 (47.5)
`
`40 (87.0)
`23 (92.0)
`17 (81.0)
`
`6 (13.0)
`2 (8.0)
`4 (19.0)
`
`75.9
`76.9
`61.6
`
`⬍0.0001
`
`5
`
`

`

`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`2044
`
`Fleseriu et al.
`
`Mifepristone and Cushing’s Syndrome
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`87.9%). The mean baseline HbA1c of 7.43 ⫾ 1.52% in
`the C-DM group decreased to 6.29 ⫾ 0.99% at wk
`24/ET (P ⬍ 0.001) (Fig. 3A). Twelve C-DM patients had
`an HbA1c over 7% at baseline (mean 8.53 ⫾ 1.11%);
`of these, nine achieved an HbA1c below 7%, including
`six reaching an HbA1c of 6% or below. C-DM and
`C-HT patients were insulin resistant and demonstrated
`rapid and significant
`improvements in AUCinsulin,
`which continued throughout
`the study (Fig. 3B);
`HOMA-IR demonstrated improvements in insulin sen-
`sitivity (Fig. 3C).
`
`Weight and body composition
`In the mITT population (n ⫽ 46), mean ⫾ SD body
`weight change from baseline (99.5 kg) to wk 24/ET was
`⫺5.7 ⫾ 7.4% (P ⬍ 0.001) (Fig. 4A). Twenty-four patients
`lost at least 5% of their baseline weight, 12 of whom lost
`
`at least 10%; 10 patients gained an average of 3.6 ⫾ 3.9%.
`Waist circumference decreased by ⫺6.8 ⫾ 5.8 cm (P ⬍
`0.001) in women and ⫺8.4 ⫾ 5.9 cm in men (P ⬍ 0.001)
`(Fig. 4B). Mean percent total body fat declined by 3.6% by
`wk 24 (P ⬍ 0.001). Absolute fat mass declined by 13.9%
`(P ⬍ 0.001) for the total body, 15.6% (P ⬍ 0.001) for the
`trunk, and 17.1% (P ⬍ 0.001) for the abdominal region
`(Fig. 4C).
`
`DBP and antihypertensive medications (C-HT and
`C-DM with HTN)
`In addition to the 21 C-HT patients, 19 C-DM patients
`had a diagnosis of HTN at study entry; 42.5% (17 of 40)
`of these had a reduction in DBP of at least 5 mm Hg at wk
`24/ET compared with baseline, and 27.5% had reductions
`in antihypertensive medications (50% of patients with a
`diagnosis of HTN were taking at least two antihyperten-
`
`FIG. 3. Changes in glucose-related outcomes. A, HbA1c significantly decreased from baseline to wk 24/ET (P ⬍ 0.001); B and C, a significant
`reduction in AUCinsulin (B) and significant improvements in HOMA-IR (C) were also observed. Data are shown as mean ⫾ SD. To convert insulin
`values to picomoles per liter, multiply by 6.945.
`
`6
`
`

`

`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`jcem.endojournals.org
`
`2045
`
`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`FIG. 4. Changes in weight and body composition. Results demonstrated a significant reduction in body weight from baseline to wk 24/ET (P ⬍
`0.001) (A), significant decreases in overall waist circumference for females and males (P ⬍ 0.001) (B), and improvements in body composition (C).
`Data are shown as mean ⫾ SE.
`
`sive medications at baseline). Overall, 52.5% (95% CI ⫽
`36.13– 68.49%) had either a response in DBP or a reduc-
`tion in antihypertensive medications (Table 3). However,
`there were no significant differences in mean systolic
`blood pressure and DBP from baseline to wk 24/ET among
`C-HT patients (129.5 ⫾ 16.3/82.9 ⫾ 11.4 vs. 129.9 ⫾
`19.0/82.8 ⫾ 13.2 mm Hg) or in C-DM patients with a
`diagnosis of HTN (137.7 ⫾ 24.0/86.4 ⫾ 15.3 vs. 132.2 ⫾
`16.7/82.4 ⫾ 13.2 mm Hg). Eight of 12 patients with DBP
`of at least 90 mm Hg at study entry had a reduction of at
`least 5 mm Hg (median decline ⫺14 mm Hg, range ⫺26.5
`to ⫺5.5 mm Hg); only one (C-DM patient) of the eight
`received additional antihypertensive therapy. AEs of in-
`creased blood pressure were reported in 12 patients, nine
`(75%) of whom had evidence of mineralocorticoid recep-
`
`tor activation (edema, hypokalemia, and/or need for spi-
`ronolactone to control hypokalemia).
`
`Mood, cognition, and quality of life
`Median BDI-II depression scores improved in the mITT
`population (baseline 14.5, range 0 – 49; wk 24/ET 9.5,
`range 0 –36; P ⬍ 0.001). For patients with at least mild
`depression at baseline (BDI-II ⱖ 14, n ⫽ 24), median
`BDI-II scores improved from 23 (range 14 – 49) to 12
`(range 0 –34) (P ⬍ 0.001). Cognition scores were mea-
`sured by the Trail Making Test at wk 24/ET; there were
`improvements in both Trail A (median decrease of 4.0 sec,
`P ⬍ 0.01) and Trail B (median decrease of 12 sec, P ⬍
`0.01). Quality of life improved at wk 24/ET as measured
`by SF-36 mental composite scores (mean 40.0 ⫾ 14.5 vs.
`
`7
`
`

`

`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`2046
`
`Fleseriu et al.
`
`Mifepristone and Cushing’s Syndrome
`
`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`45.4 ⫾ 12.5, P ⫽ 0.01) and physical composite scores
`(mean 34.9 ⫾ 11.0 vs. 39.1 ⫾ 10.8, P ⫽ 0.02).
`
`Hormone and pituitary MRI scan changes
`During mifepristone treatment, 72% of the 43 patients
`with CD had at least a 2-fold increase in ACTH, cortisol,
`or both; 28% had smaller increases. These changes were
`observed early (by d 14), plateaued from wk 10 –24, and
`declined to baseline levels at the follow-up visit 6 wk after
`discontinuation of mifepristone. Increases in ACTH of at
`least 2-fold were observed in 62.8% of patients; 33.6%
`had lesser increases, and 4.7% had no change. Late-night
`salivary cortisol increased 7.92-fold (1.43) at wk 16, and
`urinary free cortisol increased 7.70-fold (15.29) at wk 24/
`ET. At the 6-wk follow-up visit, ACTH and cortisol (se-
`rum and urine) declined to near baseline levels. Patients
`with ectopic ACTH secretion did not demonstrate in-
`creases in ACTH and cortisol in response to mifepristone.
`Pituitary MRIs were obtained in 41 CD patients; 17 had
`visible tumors, 10 of which were macroadenomas, and the
`remaining 24 did not have demonstrable tumors after sur-
`gery. MRIs were stable at wk 10 and 24 in all cases except
`one. This patient had an aggressive pituitary tumor at
`baseline that was increased in size at wk 10, leading to
`treatment discontinuation.
`
`Safety
`Overall, AEs were reported in 88% of patients during
`mifepristone treatment, most commonly nausea (48%),
`fatigue (48%), headache (44%), decreased blood potas-
`sium (34%), arthralgia (30%), vomiting (26%), periph-
`eral edema (26%), HTN (24%), dizziness (22%), de-
`creased appetite (20%), and endometrial
`thickening
`(20%). The majority of AEs were considered mild or mod-
`erate. Seven patients discontinued mifepristone because of
`an AE; fatigue was the only cause of discontinuation for
`more than one patient (n ⫽ 2). Interruptions or reductions
`in mifepristone due to AEs, most commonly nausea (n ⫽
`6), occurred in 40% of patients; there were interruptions
`or reductions for protocol-specified events in four subjects
`(two for AI, one for severe hypokalemia, and one for vag-
`inal bleeding). After dose interruption or reduction before
`wk 10, there were increases in dose in one of four and two
`of five patients, respectively; after wk 10, dose escalation
`did not occur after an interruption for an AE except in a
`single patient. Four patients experienced progression of
`preexisting metastatic malignancy that resulted in death.
`AI was reported in two patients. One occurred during
`an infection and responded to withdrawal of mifepristone;
`the other resolved with mifepristone withdrawal and
`dexamethasone administration (6 –9 mg by mouth daily
`for 6 d). Neither episode was associated with hypoglyce-
`
`mia or hypotension, and mifepristone was restarted at a
`lower dose. Analysis of AEs and concomitant medications
`identified five other instances of two or more symptoms
`possibly consistent with AI during which glucocorticoids
`were administered. Dexamethasone doses for these epi-
`sodes ranged from 2– 8 mg daily in tapering amounts for
`1–12 d. Vaginal bleeding was observed during the study in
`five premenopausal women. Prolonged metrorrhagia was
`observed in two of them after discontinuing mifepristone.
`Endometrial thickening was reported as an AE in 10
`women. Three women underwent dilatation and curettage
`for unresolved endometrial thickening.
`Twenty-two patients had a serum potassium level less
`than 3.5 mEq/liter (⬍3.5 mmol/liter), but only three ex-
`perienced severe hypokalemia [ⱕ2.5 mEq/liter (ⱕ2.5
`mmol/liter)] during mifepristone treatment, including one
`serious AE [potassium 2.1 mEq/liter (2.1 mmol/liter)]. Hy-
`pokalemia occurred in patients with both ACTH-depen-
`dent and independent CS. Four (one adrenal cancer and
`three ectopic ACTH) of seven patients with nonpituitary
`CS experienced hypokalemia during treatment. Hypoka-
`lemia was often associated with alkalosis and edema and
`generally responded to potassium replacement (10 – 420
`mEq daily); all nonpituitary CS patients received potas-
`sium supplementation. Overall, spironolactone (50 – 400
`mg daily) was used by 14 patients; it was started or in-
`creased in 11 patients for hypokalemia while taking mife-
`pristone, including one patient with adrenal cancer and
`two patients with ectopic ACTH secretion. Reversible de-
`creases in high-density lipoprotein cholesterol (HDL-C)
`and increases in TSH were observed. The mean change in
`HDL-C from baseline [62.3 ⫾ 27.8 mg/dl (1.61 ⫾ 0.72
`mmol/liter)] to wk 24/ET was ⫺14.2 ⫾ 11.9 mg/dl (0.37 ⫾
`0.31 mmol/liter) (P ⬍ 0.001); there were small declines in
`low-density lipoprotein cholesterol and triglycerides that
`were not statistically significant. Eight patients had unde-
`tectable TSH at baseline; of the remaining 42 patients,
`eight had increases in TSH above normal (three with
`TSH ⬎ 10 ␮U/liter, one with TSH of 32 ␮U/liter). Six
`weeks after mifepristone discontinuation, both HDL-C
`and thyroid function tests reverted to baseline levels.
`
`Discussion
`
`Cushing’s syndrome is a complex endocrine condition
`with serious sequelae, including cardiovascular mortality,
`fractures, proximal myopathy, insulin-resistant hypergly-
`cemia, and neuropsychiatric and neurocognitive disorders
`(35, 36). Transsphenoidal pituitary surgery with adenoma
`resection is initially successful in 65–90% of patients with
`ACTH-secreting microadenomas when performed by ex-
`
`8
`
`

`

`J Clin Endocrinol Metab, June 2012, 97(6):2039 –2049
`
`jcem.endojournals.org
`
`2047
`
`Downloaded from https://academic.oup.com/jcem/article-abstract/97/6/2039/2536684 by National Library of Medicine user on 13 March 2019
`
`pert surgeons, but approximately 20 –25% have persistent
`hypercortisolism or recurrence postoperatively; cure rates
`are lower and recurrence rates are higher for macroad-
`enomas (4). Morbidity and mortality in patients with CD
`are related to cortisol excess and rarely to the ACTH-
`secreting pituitary tumor mass. When surgery fails to re-
`verse hypercortisolemia, medical treatment can suppress
`cortisol overproduction and improve clinical manifesta-
`tions. Bilateral adrenalectomy promptly resolves hypercor-
`tiso

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