ArticlePDF Available

Relacorilant, a Selective Glucocorticoid Receptor Modulator, Induces Clinical Improvements in Patients With Cushing Syndrome: Results From A Prospective, Open-Label Phase 2 Study

Frontiers
Frontiers in Endocrinology
Authors:

Abstract and Figures

Introduction/Purpose Relacorilant is a selective glucocorticoid receptor modulator (SGRM) with no progesterone receptor activity. We evaluated the efficacy and safety of relacorilant in patients with endogenous Cushing syndrome (CS). Materials and Methods A single-arm, open-label, phase 2, dose-finding study with 2 dose groups (NCT02804750, https://clinicaltrials.gov/ct2/show/NCT02804750) was conducted at 19 sites in the U.S. and Europe. Low-dose relacorilant (100-200 mg/d; n = 17) was administered for 12 weeks or high-dose relacorilant (250-400 mg/d; n = 18) for 16 weeks; doses were up-titrated by 50 mg every 4 weeks. Outcome measures included proportion of patients with clinically meaningful changes in hypertension and/or hyperglycemia from baseline to last observed visit. For patients with hypertension, clinical response was defined as a ≥5-mmHg decrease in mean systolic or diastolic blood pressure, measured by a standardized and validated 24-h ABPM. For patients with hyperglycemia, clinical response was defined ad-hoc as ≥0.5% decrease in HbA1c, normalization or ≥50-mg/dL decrease in 2-h plasma glucose value on oral glucose tolerance test, or decrease in daily insulin (≥25%) or sulfonylurea dose (≥50%). Results 35 adults with CS and hypertension and/or hyperglycemia (impaired glucose tolerance or type 2 diabetes mellitus) were enrolled, of which 34 (24 women/10 men) received treatment and had postbaseline data. In the low-dose group, 5/12 patients (41.7%) with hypertension and 2/13 patients (15.4%) with hyperglycemia achieved response. In the high-dose group, 7/11 patients (63.6%) with hypertension and 6/12 patients (50%) with hyperglycemia achieved response. Common (≥20%) adverse events included back pain, headache, peripheral edema, nausea, pain at extremities, diarrhea, and dizziness. No drug-induced vaginal bleeding or hypokalemia occurred. Conclusions The SGRM relacorilant provided clinical benefit to patients with CS without undesirable antiprogesterone effects or drug-induced hypokalemia.
Content may be subject to copyright.
Relacorilant, a Selective
Glucocorticoid Receptor Modulator,
Induces Clinical Improvements in
Patients With Cushing Syndrome:
Results From A Prospective,
Open-Label Phase 2 Study
Rosario Pivonello
1
, Irina Bancos
2
, Richard A. Feelders
3
, Atil Y. Kargi
4
, Janice M. Kerr
5
,
Murray B. Gordon
6
, Cary N. Mariash
7
, Massimo Terzolo
8
, Noel Ellison
9
and Andreas G. Moraitis
10
*
1
Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy,
2
Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester,
MN, United States,
3
Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam,
Netherlands,
4
Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Miami, Miami, FL,
United States,
5
Department of Endocrinology, Division of Endocrinology, Metabolism and Diabetes, University of Colorado
Denver, Aurora, CO, United States,
6
Allegheny Neuroendocrinology Center, Allegheny General Hospital, Pittsburgh, PA,
United States,
7
Methodist Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States,
8
Department of Clinical and Biological Sciences, Internal Medicine 1 San Luigi Gonzaga Hospital, University of Turin,
Orbassano, Italy,
9
Biostatistics, Trialwise, Inc, Houston, TX, United States,
10
Drug Research and Development, Corcept
Therapeutics, Menlo Park, CA, United States
Introduction/Purpose: Relacorilant is a selective glucocorticoid receptor modulator
(SGRM) with no progesterone receptor activity. We evaluated the efcacy and safety of
relacorilant in patients with endogenous Cushing syndrome (CS).
Materials and Methods: A single-arm, open-label, phase 2, dose-nding study with 2
dose groups (NCT02804750, https://clinicaltrials.gov/ct2/show/NCT02804750) was
conducted at 19 sites in the U.S. and Europe. Low-dose relacorilant (100-200 mg/d;
n = 17) was administered for 12 weeks or high-dose relacorilant (250-400 mg/d; n = 18)
for 16 weeks; doses were up-titrated by 50 mg every 4 weeks. Outcome measures
included proportion of patients with clinically meaningful changes in hypertension and/or
hyperglycemia from baseline to last observed visit. For patients with hypertension, clinical
response was dened as a 5-mmHg decrease in mean systolic or diastolic blood
pressure, measured by a standardized and validated 24-h ABPM. For patients with
hyperglycemia, clinical response was dened ad-hoc as 0.5% decrease in HbA1c,
normalization or 50-mg/dL decrease in 2-h plasma glucose value on oral glucose
tolerance test, or decrease in daily insulin (25%) or sulfonylurea dose (50%).
Results: 35 adults with CS and hypertension and/or hyperglycemia (impaired glucose
tolerance or type 2 diabetes mellitus) were enrolled, of which 34 (24 women/10 men)
received treatment and had postbaseline data. In the low-dose group, 5/12 patients
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628651
Edited by:
Corin Badiu,
Carol Davila University of Medicine
and Pharmacy, Romania
Reviewed by:
Annamaria Anita Livia Colao,
University of Naples Federico II, Italy
George Mastorakos,
National and Kapodistrian University
of Athens, Greece
*Correspondence:
Andreas G. Moraitis
amoraitis@corcept.com
This article was submitted to
Pituitary Endocrinology,
a section of the journal
Frontiers in Endocrinology
Specialty section:
Received: 01 February 2021
Accepted: 25 June 2021
Published: 14 July 2021
Citation:
Pivonello R, Bancos I, Feelders RA,
Kargi AY, Kerr JM, Gordon MB,
Mariash CN, Terzolo M, Ellison N
and Moraitis AG (2021) Relacorilant, a
Selective Glucocorticoid Receptor
Modulator, Induces Clinical
Improvements in Patients
With Cushing Syndrome:
Results From A Prospective,
Open-Label Phase 2 Study.
Front. Endocrinol. 12:662865.
doi: 10.3389/fendo.2021.662865
CLINICAL TRIAL
published: 14 July 2021
doi: 10.3389/fendo.2021.662865
(41.7%) with hypertension and 2/13 patients (15.4%) with hyperglycemia achieved
response. In the high-dose group, 7/11 patients (63.6%) with hypertension and 6/12
patients (50%) with hyperglycemia achieved response. Common (20%) adverse events
included back pain, headache, peripheral edema, nausea, pain at extremities, diarrhea,
and dizziness. No drug-induced vaginal bleeding or hypokalemia occurred.
Conclusions: The SGRM relacorilant provided clinical benet to patients with CS without
undesirable antiprogesterone effects or drug-induced hypokalemia.
Keywords: clinical trial, cortisol, Cushing syndrome, glucocorticoid, hypercortisolism, hyperglycemia,
hypertension, relacorilant
INTRODUCTION
Endogenous hypercortisolism (Cushing syndrome [CS]) is a
complex, multisystem endocrine disorder characterized by
cortisol excess and is frequently associated with hypertension
and hyperglycemia (including impaired glucose tolerance/type 2
diabetes mellitus [IGT/T2DM]) (1,2). CS, especially if untreated,
is associated with increased cardiovascular-related mortality and
multiple morbidities beyond hypertension and hyperglycemia,
including visceral obesity, dyslipidemia, liver steatosis,
osteoporosis, hypercoagulopathy, susceptibility to infection,
neuropsychiatric disorders, and reproductive and sexual
disturbances (211).
Medical therapies, including pituitary-targeting agents,
steroid synthesis inhibitors, and glucocorticoid receptor [GR]
antagonists, are a treatment option for patients who are not
candidates for surgery, and for patients with persistent or
recurrent hypercortisolism after surgery who are unsuitable
for, or unwilling to undergo, additional surgical procedures
(1215).
The competitive GR and progesterone antagonist mifepristone
was approved by the Food and Drug Administration in 2012 to
control hyperglycemia in patients with endogenous CS who have
IGT or T2DM. The clinical benet of mifepristone in patients with
CS was shown in an open-label, phase 3 trial, SEISMIC (Study of
the Efcacy and Safety of Mifepristone in the Treatment of
Endogenous Cushing Syndrome) (16). Serum cortisol and
ACTH levels may rise in response to GR antagonism with
mifepristone (15,17,18). In patients with CS, further increases
in cortisol in turn can lead to stimulation of the mineralocorticoid
receptor, resulting in adverse events such as hypertension and
hypokalemia (15,18,19). Because of its mechanism of action,
clinical and metabolic parameters, rather than cortisol levels, must
be monitored in patients during mifepristone treatment for
assessment of efcacy (18).
Relacorilant (CORT125134, Corcept Therapeutics, Menlo
Park, CA) is an investigational, highly selective GR modulator
that competitively antagonizes cortisol activity (20). Unlike
mifepristone, relacorilant does not bind to the progesterone
receptor (20). Given its binding afnity prole, it was
hypothesized that relacorilant would provide patients with
endogenous CS the treatment benets of cortisol modulation,
but without the unwanted effects of progesterone receptor
antagonism (e.g., induction of abortion, progesterone receptor
modulator-associated endometrial changes and irregular vaginal
bleeding in women).
This dose-nding study was designed to assess the efcacy
and safety of relacorilant in patients with endogenous
hypercortisolism due to either excess ACTH secretion (from a
pituitary or other ectopic tumor) or autonomous adrenal cortisol
secretion, as well as to inform phase 3 study design. Because GR
antagonism lowers cortisol activity, but not cortisol levels (16),
an improvement in clinical manifestations of hypercortisolism
were evaluated as assessments of efcacy with relacorilant. This
study assessed the impact of reduced cortisol activity on blood
pressure and parameters of blood glucose control following
treatment with relacorilant in patients with CS. Additional
secondary and exploratory outcome measures were also assessed.
MATERIALS AND METHODS
Study Design
This phase 2, multicenter, single-arm, open-label, dose-nding
study (NCT02804750) assessed the efcacy and safety of
relacorilant using 2 dose groups: a low-dose group and a high-
dose group. The dose in each group was increased by 50 mg every
4 weeks (Figure 1). The low-dose group received a starting
relacorilant dose of 100 mg/d, followed by 150 mg/d and then
200 mg/d. The high-dose group began with a starting relacorilant
dose of 250 mg/d, followed by 300 mg/d, 350 mg/d, and 400 mg/
d. Dose reductions were permitted at the discretion of the
investigator for safety/tolerability. Enrolment in the high-dose
Abbreviations: ABPM, ambulatory blood pressure monitoring; ACTH,
adrenocorticotropic hormone; ALT, alanine aminotransferase; aPTT, activated
partial thromboplastin time; AST, aspartate aminotransferase; AUC, area under
the curve; BDI-II, Beck Depression Inventory; BMI, body mass index; CD,
Cushing disease; CI, condence interval; CS, Cushing syndrome; DBP, diastolic
blood pressure; DST, dexamethasone suppression test; ET, early termination; GR,
glucocorticoid receptor; HbA1c, glycated hemoglobin; HOMA-IR, homeostatic
model assessment of insulin resistance; IGT, impaired glucose tolerance; LNSC,
late-night salivary cortisol; NAFLD, nonalcoholic fatty liver disease; oGTT, oral
glucose tolerance test; QoL, quality of life; SBP, systolic blood pressure; SD,
standard deviation; SGRM, selective glucocorticoid receptor modulator; TEAE,
treatment-emergent adverse event; T2DM, type 2 diabetes mellitus; UFC, urinary
free cortisol; ULN, upper limit of normal.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628652
group began after completion of enrollment of the low-
dose group.
The study was approved by the institutional review board at
each study center and conducted in accordance with the World
Medical Association Declaration of Helsinki and the
International Council on Harmonisation Good Clinical
Practice guidelines. All patients provided written informed
consent. The study was conducted at 19 centers in the United
States, Italy, United Kingdom, Hungary, and The Netherlands
between February 2017 and September 2018.
Patients
Adult patients aged 18 to 80 years with a diagnosis of
endogenous CS and requiring medical treatment (i.e., those for
whom surgery or radiation is contraindicated or has been
refused) were eligible for the study. These patients must have
met at least 2 of the following biochemical criteria based on the
Endocrine Society Guidelines (Nieman 2008): (1) a 24-h urinary
free cortisol (UFC) above the upper limit of normal (ULN) (50
µg/24 h [138 nmol/d]) on at least 2 collections, (2) a late-night
salivary cortisol (LNSC) value above ULN (10-11 PM normal
range, 0.09 µg/dL [2.5 nmol/L]) on at least 2 collections, or (3) a
lack of cortisol suppression (>1.8 µg/dL serum cortisol [49.7
nmol/L]) on either the overnight 1-mg or 48-h 2-mg
dexamethasone suppression test (DST) (1,21). In addition,
patients were also required to have at least 2 clinical signs or
symptoms of hypercortisolism: a Cushingoid appearance (moon
facies, dorsocervical fat pad, and/or facial plethora), increased
body weight or central obesity, proximal muscle weakness, low
bone mass (dual energy X-ray absorptiometry T < -1.0),
psychiatric symptoms (including depression or psychosis), easy
bruising, or skin changes (hirsutism, violaceous striae, and/or
acne) (1).
Patients with adrenal adenomas were also eligible if they met
the following criteria for autonomous cortisol secretion based on
the European Society of Endocrinology/European Network for
the Study of Adrenal Tumors Guidelines (Fassnacht 2016):
unilateral or bilateral adrenal disease, lack of cortisol
suppression (>5 µg/dL [>138 nmol/L] serum cortisol) on either
1-mg or 48-h 2-mg DST, low or suppressed ACTH (<10 pg/mL
[<2.2 pmol/L]), and presence of at least 2 comorbidities
potentially related to cortisol excess (e.g., T2DM, hypertension,
obesity, osteoporosis) (21).
Because changes in blood pressure and glucose tolerance
served as key efcacy endpoints, patients included in the
study were required to have uncontrolled hypertension
(hypertension cohort), and/or IGT or T2DM (hyperglycemia
cohort). Uncontrolled hypertension was dened as a mean
systolic blood pressure (SBP) of 130 mmHg and/or mean
diastolic blood pressure (DBP) of 85 mmHg using 24-h
ambulatory blood pressure monitoring (ABPM) based on the
European Society of Hypertension Position Paper (OBrien 2013)
(22). IGT was dened as a 2-h oral glucose tolerance test (oGTT)
plasma glucose value of 140-199 mg/dL (7.8-11.0 mmol/L) after
75 g of glucose, while T2DM was dened as a fasting plasma
glucose >126 mg/dL (7.0 mmol/L) or a 2-h plasma glucose 200
mg/dL (11.1 mmol/L) after a 75-g oGTT (23). Patients on
antidiabetic or antihypertensive medications were allowed in
FIGURE 1 | Study design: dose escalation in the low- and high-dose groups. ABPM, ambulatory blood pressure monitoring; oGTT, oral glucose tolerance test.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628653
the study if their doses were stable and had not been changed for
at least 1 month before the baseline assessment. Patients
receiving medications for CS underwent an appropriate
washout duration before baseline assessments were performed
(e.g., 1 month for adrenostatic medications such as metyrapone
and ketoconazole; 2 months for long-acting somatostatin analogs
and dopamine agonists; 1 month for short-acting somatostatin
analogs; and 6 weeks for mifepristone).
Patients with SBP >170 or DBP >110 mmHg, glycated
hemoglobin (HbA1c) >12%, uncontrolled hypothyroidism or
hyperthyroidism, or uncorrected hypokalemia (<3.5 mEq/L)
were excluded. Concurrent use of other medications for CS
was not allowed in the study. Patients who had undergone
radiation therapy for CS within 1 year of screening were
also excluded.
Assessments
Patients With Hypertension
Assessment of hypertension was performed using standardized,
validated 24-h ABPM to allow for an accurate measurement (22,
24). A clinically signicant response was dened as a decrease of
5 mmHg in either mean 24-h SBP or DBP from baseline
without an increase in dosage of concurrent antihypertensive
medication or initiation of additional antihypertensive
medication during the treatment period.
Patients With Hyperglycemia
To assess glucose tolerance, a 2-h oGTT was administered and
HbA1c was evaluated in all patients with hyperglycemia.
Clinically meaningful response was dened ad-hoc as one of
the following: a decrease in HbA1c of 0.5% from baseline,
normalization of the 2-h plasma glucose value on the oGTT
(<140 mg/dL [<7.8 mmol/L]) or a decrease in the 2-h plasma
glucose value on the oGTT by 50 mg/dL (2.8 mmol/L) from
baseline, or a decrease in the total daily insulin dose by 25% or a
decrease in the daily sulfonylurea dose by 50% from baseline.
Based on the criterion used in the pivotal study of mifepristone,
SEISMIC (16), the hyperglycemia response was initially dened
as a decrease of 25% from baseline in the area under the
concentration-time curve for glucose (AUC
glucose
), without an
increase in dosage of concurrent antidiabetes medication or
additional antidiabetes medication during the treatment
period. However, unlike SEISMIC, a substantial portion of the
patients enrolled in the hyperglycemia group in the current study
had IGT rather than overt diabetes, and the patients with
diabetes had much better glycemic control (mean HbA1c was
6.6%, compared with 7.4% in SEISMIC) (16). Thus, a 25%
decrease in the total AUC
glucose
from baseline would not be
suitable to detect clinically meaningful improvements in
these patients.
Secondary and Exploratory Endpoints
Considering the numerous additional comorbidities and clinical
complications associated with CS, including obesity, impaired
glucose metabolism and insulin resistance, liver steatosis,
osteoporosis, immune disorders, thrombosis, and neuropsychiatric
diseases (2,3,5,10,11,25), the effect of relacorilant on several
secondary and exploratory endpoints was also assessed. These
included changes in body weight; serum fructosamine an
indicator of glucose metabolism control (26); homeostatic model
assessment of insulin resistance (HOMA-IR); liver function tests;
serum osteocalcin, a marker of bone metabolism (25); eosinophils;
activated partial thromboplastin time (aPTT), Factor VIII, and
platelet count for assessment of coagulation; quality of life (QoL)
as assessed using the Cushing QoL Questionnaire (27); depressive
symptoms as assessed using the Beck Depression Inventory (BDI-II)
(28); and cognitive function evaluated using the Trail Making Test
(Parts A and B) (29). Hormone changes were assessed using plasma
ACTH, and cortisol (24-h UFC, LNSC, and serum cortisol).
Treatment-emergent AEs (TEAEs) were assessed at every visit for
safety. Potassium levels were monitored, particularly for the
emergence of hypokalemia.
Blood, urine, and saliva samples were collected and analyzed
centrally (by Q
2
Solutions Laboratories, Valencia, CA; Quest
Diagnostics Nichol Institute, San Juan Capistrano, CA was used
for hormonal testing only). AUC
glucose
was calculated based on
results of the 2-h oGTTs. Plasma glucose and insulin values from
the baseline oGTT assessment (pre-glucose drink) were used to
calculate HOMA-IR (30). Urinary cortisol (normal range, 4-50
µg/24h [11-138 nmol/d]) was measured by tandem mass
spectrometry (MS/MS). Serum cortisol (8-10 AM normal
range, 4.6-20.6 µg/dL [127-567 nmol/L]) levels and LNSC
levels (10-11 PM normal range, 0.09 µg/dL [2.5 nmol/L]),
were measured by liquid chromatography LC-MS/MS. Plasma
ACTH (7-10 AM normal range, 6-50 pg/mL [1.3-11.1 pmol/L])
was measured with an immunochemiluminescent assay.
Statistical Analysis
No formal sample size calculation was performed. A sample size
of 30 patients (15 per dose group) was deemed sufcient for a
preliminary evaluation of the efcacy and safety of relacorilant
and to establish estimates of efcacy for future evaluation of the
study drug. All patients who received at least one dose of study
medication were included in the safety analysis. All patients who
received at least one dose of study medication and had
postbaseline data were included in the efcacy population.
Because of the small population in the clinical responder
analyses, additional exclusions for major protocol violations
were applied to specic visits or outcomes for patients in the
hypertension and hyperglycemia groups, rather than excluding
the patient entirely.
The key efcacy endpoints (hypertension and hyperglycemia
responders) were analyzed by dose group at Week 12 or early
termination (ET) for the low-dose group and Week 16 or ET for
the high-dose group, and summarized as last observed. The
number and percentage of patients considered responders was
presented along with the 95% exact binomial two-sided
condence intervals (Clopper-Pearson). Because there was no
comparator group, statistical signicance was determined if the
lower limit of the 95% exact binomial CIs for the responder rate
was >20%. This threshold for response was selected based on the
similar threshold used in the previously reported SEISMIC study
(16). SAS statistical software version 9.4 or higher (SAS Institute,
Cary, NC) was used.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628654
Descriptive statistics were used to summarize changes from
baseline in secondary and exploratory outcomes, including
weight, fructosamine, HOMA-IR, liver function tests, serum
osteocalcin, absolute eosinophils, aPTT, Factor VIII, platelet
count, QoL, depressive symptoms, cognitive function, and
TEAEs. Means or medians and Wilcoxon signed-rank p-values
were calculated to assess change from baseline for exploratory
outcomes (fructosamine, HOMA-IR, osteocalcin, absolute
eosinophils, coagulation parameters, QoL, depressive
symptoms, cognitive function, plasma ACTH, 24-h UFC,
LNSC, and serum cortisol changes).
RESULTS
Patients
Thirty-ve patients meeting the inclusion criteria were enrolled.
All 35 received at least 1 treatment dose and were included in the
safety population. Seventeen patients were enrolled in the low-
dose group and 18 patients were enrolled in the high-dose group.
One patient did not have any postbaseline data and was excluded
from the efcacy population (n = 34) (Figure 2).
Baseline characteristics of the efcacy population (70.6%
female [12/24 premenopausal], mean ± SD age of 48.2 ± 13.33
years) are shown in Tables 1 and 2. Twenty-seven patients
(79.4%) were diagnosed with ACTH-dependent CS, and 7
(20.6%) were diagnosed with ACTH-independent CS (adrenal
adenoma). Of the patients with ACTH-dependent CS, 4 were
diagnosed with ectopic ACTH secretion. Among the total 34
patients, 47.1% (n = 16; 8 low-dose/8 high-dose group) were
diagnosed with both hyperglycemia and hypertension, 32.4%
(n = 11; 5 low-dose/6 high-dose group) were diagnosed with
hyperglycemia only, and 20.6% (n = 7; 4 low-dose/3 high-dose
group) were diagnosed with hypertension only. Among patients
in the hypertension analysis group (n = 23), mean 24-h blood
pressure (SBP/DBP) at baseline was 138.3/87.0 mmHg. Mean
HbA1c at baseline among patients in the hyperglycemia analysis
group (n = 27) was 6.6%. Among patients in the hyperglycemia
analysis group, 37.0% (10/27) had IGT and 63.0% (17/27)
had T2DM.
Key Efcacy Analyses
Among patients in the hypertension analysis population (n =
23), 5 of 12 patients (41.7%, 95% CI 15.17, 72.33) in the low-dose
FIGURE 2 | Patient disposition.
a
One patient in the high-dose group did not have any postbaseline data and was excluded from the efcacy population.
b
Two patients in
the high-dose hyperglycemia group did not have any postbaseline efcacy data while on study drug and were excluded from the hyperglycemia population.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628655
group and 7 of 11 patients (63.6%, 95% CI 30.79, 89.07) in the
high-dose group were responders at their last observed blood
pressure measurement (Table 3).
Among patients in the hyperglycemia analysis population
(n = 25), 2 of the 13 patients (15.4%, 95% CI 1.92%, 45.45) in the
low-dose group and 6 of the 12 patients (50.0%, CI 21.09%,
78.91%) in the high-dose group met the ad-hoc hyperglycemia
response at the last observed visit (Table 3). Additionally, when
both the low-dose and high-dose groups were combined, plasma
glucose levels during oGTT decreased (Figure 3), leading to a
statistically signicant decrease in mean AUC
glucose
from
baseline to the last observed visit (-2.48 hmmol/L, p< 0.01).
Secondary and Exploratory
Efcacy Analyses
Changes in Weight
In the efcacy population, the median weight at baseline was
similar among patients in the low-dose (85.5 kg) and high-dose
(91.1 kg) group. During the study, 7 of 17 patients (41.2%) in the
low-dose group and 10 of 16 patients (62.5%) in the high-dose
group lost weight. Among the patients who lost weight during
the study, the median (min, max) changes from baseline to last
observed visit were -2.0 kg (-2.5, -0.4) and -3.0 kg (-15.1, -1.1) for
the low-dose and high-dose groups, respectively. Among all
patients, the median weight change was 0.50 kg in the low-
dose group and -1.75 kg in the high-dose group.
Additional Exploratory Endpoints
Statistically signicant improvements from baseline were
observed in various exploratory endpoints related to cortisol
excess in the efcacy population (Table 4). These included
improvements in fructosamine (mean change -13.92 µmol/L,
p= 0.002) among all patients with hyperglycemia. Decreases in
the liver function parameters alanine aminotransferase (mean
change -10.62 U/L, p< 0.001) and aspartate aminotransferase
(mean change -4.94 U/L, p= 0.001), and increases in serum
osteocalcin (mean change 3.00 µg/L, p < 0.01) and eosinophil
count (mean change 0.0510
9
/L, p= 0.006) were observed in the
overall patient population. The aPTT increased signicantly
(mean change 1.45 sec, p= 0.046) and was accompanied by
signicant decreases in factor VIII (mean change -18.94%, p=
0.022) and platelet counts (mean change -68.8210
9
/L, p< 0.001).
Statistically signicant improvements from baseline to last
observed visit were observed in BDI-II Total score (mean
change -3.48, p= 0.004), Cushing QoL score (mean change
7.13, p= 0.002), and Trail Making Test Part A and Part B
(mean changes -4.13 sec, p= 0.003 and -24.69 sec, p<
0.001, respectively).
Hormone Changes
Among all patients with ACTH-dependent CS (n = 26 with
values at last visit), the median (min, max) change from baseline
to last observed visit was 3.7 (-15.6, 20.0) pmol/L [16.8 (-70.9,
90.9) pg/mL] (p= 0.003) for plasma ACTH; 4.0 (-1650.5, 1404.8)
nmol/d (1.4 [-598.0, 509.0] µg/24 h) (p= 0.873) for 24-h UFC;
-0.33 (-30.6, 47.7) nmol/L (-0.01 [-1.1, 1.7] µg/dL) (p= 0.815) for
TABLE 1 | Demographic and clinical characteristics at baseline (efcacy
population, n = 34).
Overall population
(n = 34)
Age, years (mean ± SD) 48.2 ± 13.33
Female sex, n (%) 24 (70.6)
White race, n (%) 34 (100)
Ethnicity, n (%)
Hispanic or Latino 3 (8.8)
Not Hispanic or Latino 31 (91.2)
Weight, kg (mean ± SD) 97.8 ± 27.41
BMI, kg/m
2
(mean ± SD) 35.6 ± 9.75
Etiology of CS, n (%)
ACTH-dependent: CD or ectopic, n (%) 27 (79.4)
ACTH, pmol/L (median [min, max]) 14.0 (6.0, 28)
24-h UFC, nmol/d (median [min, max]) 357.4 (83.4, 2823.8)
Adrenal adenoma (unilateral), n (%) 7 (20.6)
ACTH, pmol/L (median [min, max]) 1.0 (1.0, 1.6)
24-h UFC, nmol/d (median [min, max])
a
156.1 (56.9, 2341.0)
Osteocalcin, µg/L, (mean ± SD)
b
11.0 ± 7.55
Analysis cohorts
Hypertension, n (%) 23 (67.6)
24-h SBP, mmHg (mean ± SD) 138.3 ± 8.59
24-h DBP, mmHg (mean ± SD) 87.0 ± 5.65
Hyperglycemia, n (%) 27 (79.4)
IGT, n (%) 10 (37.0)
T2DM, n (%) 17 (63.0)
Fructosamine, µmol/L (mean ± SD) 225.3 ± 29.50
Fasting plasma glucose, mmol/L (mean ± SD) 6.6 ± 1.89
2-h oGTT plasma glucose, mmol/L (mean ± SD) 12.7 ± 4.08
AUC
glucose
,hmmol/L (mean ± SD) 26.2 ± 8.05
HbA1c, % (mean ± SD) 6.6 ± 1.26
To convert values of glucose to mg/dL, divide by 0.0555; UFC to µg/24 h, divide by 2.76.
a
Includes one patient with an adrenal adenoma who had a baseline 24-h UFC of 2,341
nmol/d (848.2 µg/d).
b
Osteocalcin n = 33.
ACTH, adrenocorticotropic hormone; AUC, area under the curve; BMI, body mass index;
CD, Cushing disease; CS, Cushing syndrome; DBP, diastolic blood pressure; HbA1c,
glycated hemoglobin; IGT, impaired glucose tolerance; oGTT, oral glucose tolerance test;
SBP, systolic blood pressure; SD, standard deviation; T2DM, type 2 diabetes mellitus;
UFC, urinary free cortisol.
TABLE 2 | Median biochemistry at baseline (efcacy population, n = 34).
Low-dose group (n = 17) High-dose group (n = 17) Overall population (n = 34)
ACTH, pmol/L (min, max) 9.8 (1.0, 25.3) 14.0 (1.0, 28) 10.8 (1.0, 28.0)
24-h UFC, nmol/d (min, max) 357.4 (122.0, 2823.8) 251.6 (56.9, 2341.0) 357.0 (56.9, 2823.8)
LNSC, nmol/L (min, max) 7.2 (1.2, 36.5) 5.4 (2.1, 32.7) 6.6 (1.2, 36.5)
Serum cortisol, nmol/L (min, max) 483.0 (141.0, 748.0) 431.0 (235.0, 1352.0) 459.5 (141.0, 1352.0)
To convert values of ACTH to pg/mL, divide by 0.22; UFC to µg/24 h, divide by 2.76; LNSC and serum cortisol to µg/dL, divide by 27.6.
ACTH, adrenocorticotropic hormone; LNSC, late-night salivary cortisol; SD, standard deviation; UFC, urinary free cortisol.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628656
LNSC; and 22.0 (-127.0, 339.0) nmol/L (0.8 [-4.6, 12.3] µg/dL)
(p= 0.239) for serum cortisol.
In the adrenal subgroup (n = 7), median (min, max) change
from baseline to last observed visit was 0.2 (0, 1.4) pmol/L (0.9 [0,
6.4] pg/mL) for plasma ACTH; 7.0 (-1662.9, 42.8) nmol/d (2.6
[-602.5, 15.5] µg/24 h) for 24-h UFC; -0.7 (-6.8, 11.8) nmol/L
(-0.03 [-0.2, 0.4] µg/dL) for LNSC; and -3.9 (-179.0, 130.0) nmol/
L (-0.1 [-6.5, 4.7] µg/dL) for serum cortisol. There were too few
patients in the adrenal subgroup to adequately perform
statistical testing.
Safety
Overall, TEAEs were reported in 94.3% of patients (33/35)
during treatment with relacorilant, including 88.2% (15/17) in
the low-dose group and 100% (18/18) in the high-dose group
(Table 5). The most commonly reported TEAEs (20%) in both
the low-dose and high-dose groups combined were back pain
(31.4% [11/35]), headache (25.7% [9/35]), peripheral edema
(25.7% [9/35]), nausea (22.9% [8/35]), pain at extremities
(22.9% [8/35]), diarrhea (20.0% [7/35]), and dizziness (20.0%
[7/35]) (Table 5). In the high-dose group, the highest incidence
of TEAEs was observed with the initial starting dose of 250 mg
(100%, 18/18), when compared with incidences of TEAEs at
higher doses later (64.3% [9/14], 76.9% [10/13], and 25.0% [2/8]
at 300, 350, and 400 mg, respectively). For musculoskeletal and
gastrointestinal TEAEs specically, the highest incidences also
occurred at the 250-mg starting dose (61.1% [11/18] and 38.9%
[7/18], respectively).
TEAEs leading to discontinuation were reported in 1 patient
(5.9%) in the low-dose group and 8 patients (44.4%) in the high-
dose group. TEAEs leading to discontinuation in more than one
patient were musculoskeletal in nature (e.g., myopathy,
back pain).
Five serious TEAEs were reported in 4 patients (pilonidal cyst,
myopathy, polyneuropathy, hypertension, and myocardial
infarction). All serious TEAEs occurred in the high-dose
group. In 3 patients, the serious TEAEs led to discontinuation
(myopathy, polyneuropathy, and myocardial infarction). For the
patient who developed a serious TEAE of myopathy, a cerebral
spinal uid analysis revealed ndings consistent with Guillain-
Barre syndrome based on the low cell count and moderately
elevated protein. The serious TEAE of polyneuropathy occurred
TABLE 3 | Summary of responder analysis in patients with hypertension (n = 23) and hyperglycemia (n = 25) at last observation.
Low-dose group High-dose group
Responder, n/N (%) 95% CI Responder, n/N (%) 95% CI
Hypertension
a
5/12 (41.7) 15.17, 72.33 7/11 (63.6) 30.79, 89.07
Hyperglycemia
b
2/13 (15.4) 1.92, 45.45 6/12 (50.0) 21.09, 78.91
CIs: 95% binomial exact two-sided condence interval (Clopper-Pearson).
Two patients in the hyperglycemia responder analysis were excluded because they had no postbaseline efcacy data collected while on study drug.
a
Response dened as a 5 mmHg decrease in mean systolic or diastolic BP from baseline without the use of additional antihypertensive medication or an increase in dosage of a
concurrent antihypertensive medication.
b
Achieving any of the ad-hoc hyperglycemia response criteria with no increase in HbA1c. Patients who achieve the response criteria but whose HbA1c increases cannot be considered
overall responders. Ad-hoc response criteria: a 0.5% decrease in HbA1c, normalization (<140 mg/dL [<7.8 mmol/L]) or 50-mg/dL (2.8 mmol/L) decrease in 2-h glucose value on oGTT,
or decrease in daily insulin (25%) or sulfonylurea dose (50%).
CI, condence interval.
FIGURE 3 | Results of oGTT tests at baseline and at last observation in the hyperglycemia group. p-value from Wilcoxon signed-rank. AUC, area under the curve;
oGTT, oral glucose tolerance test; SE, standard error.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628657
in a patient with underlying uncontrolled diabetes; nerve
conduction studies revealed parasympathetic autonomic
neuropathy and moderate sensory hypesthesia consistent with
diabetic neuropathy. The myocardial infarction (non-ST
segment elevation) event occurred in a patient with a history
of coronary artery disease, thromboembolism, pulmonary
embolism, and myocardial infarction. Cardiac catheterization
showed unchanged coronary lesions compared to previous
catheterization studies. During the study, no drug-induced
vaginal bleeding or hypokalemia was reported. No patients had
a platelet count <100,000/µL during the study, nor were there
any bleeding events related to a reduction in platelets.
No clinically signicant changes in potassium levels were
observed in the study. Mean potassium levels changed from 4.33
mmol/L at baseline to 4.21 mmol/L at week 12 in the low-dose
group, and from 4.34 mmol/L at baseline to 4.33 mmol/L at week
16 in the high-dose group (Figure 4). Mean changes in
potassium levels from baseline to last observed visit across
each dose level ranged from -0.12 to 0.02 mmol/L in the low-
dose group and from -0.31 to 0.08 mmol/L in the high-
dose group.
DISCUSSION
Relacorilant is a selective glucocorticoid receptor modulator with
no afnity for the progesterone receptor (20). In this multicenter,
two-step, dose-nding study of patients with endogenous CS and
hypertension and/or hyperglycemia, relacorilant improved both
clinical hypertension and hyperglycemia parameters in the
respective patient groups. Although not formally assessed,
differences in response rates between dose groups suggested a
dose response. In addition, a number of exploratory endpoints
related to cortisol excess were signicantly improved.
For patients in the hypertension subgroup, 41.7% (5/12) of
the low-dose group and 63.6% (7/11) of the high-dose group met
criteria for a clinically meaningful blood pressure response,
dened as patients with a decrease of 5 mmHg in either
mean 24-h SBP or DBP from baseline, and without an increase
of concurrent antihypertensive medication or additional
antihypertensive medication during the treatment period. In
the 24-week SEISMIC study, 38.1% (8/21) of patients met the
criteria for hypertension response, which for that trial was
dened as a 5 mmHg reduction from baseline in DBP
TABLE 4 | Mean change from baseline to last observation in exploratory secondary endpoints (efcacy population, n = 34).
Parameter Mean change from baseline to last observation p-value
Fructosamine (mmol/L) -13.92 0.002
HOMA-IR
a
-1.58 0.064
ALT (U/L) -10.62 < 0.001
AST (U/L) -4.94 0.001
Serum osteocalcin (mg/L) 3.00 < 0.01
Absolute eosinophils (10
9
/L) 0.05 0.006
aPTT (sec) 1.45 0.046
Factor VIII (%) -18.94 0.022
Platelet count (10
9
/L) -68.82 < 0.001
BDI-II Total score -3.48 0.004
Cushing QoL score 7.13 0.002
Trail Making Test Part A Total time to complete test (sec) -4.13 0.003
Trail Making Test Part B Total time to complete test (sec) -24.69 < 0.001
p-values for the mean change from baseline to last observation are from the Wilcoxon signed-rank test.
a
A signicant difference was observed in the high-dose treatment group (mean change -3.2, p = 0.033).
ALT, alanine aminotransferase; aPTT, activated partial thromboplastin time; AST, aspartate aminotransferase; AUC, area under the curve; BDI, Beck Depression Inventory; HOMA-IR,
homeostatic model assessment for insulin resistance; QoL, quality of life.
TABLE 5 | TEAEs by preferred term in the safety population.
TEAE, n (%) Low-dose group (n = 17) High-dose group (n = 18) Overall population (n = 35)
Patients reporting 1 TEAE 15 (88.2) 18 (100.0) 33 (94.3)
TEAE occurring in 20% of either the low-dose or high-dose group
Back pain 4 (23.5) 7 (38.9) 11 (31.4)
Headache 4 (23.5) 5 (27.8) 9 (25.7)
Peripheral edema 4 (23.5) 5 (27.8) 9 (25.7)
Nausea 3 (17.7) 5 (27.8) 8 (22.9)
Pain in extremity 4 (23.5) 4 (22.2) 8 (22.9)
Diarrhea 4 (23.5) 3 (16.7) 7 (20.0)
Dizziness 3 (17.7) 4 (22.2) 7 (20.0)
Arthralgia 2 (11.8) 4 (22.2) 6 (17.1)
Dyspepsia 1 (5.9) 4 (22.2) 5 (14.3)
Myalgia 1 (5.9) 4 (22.2) 5 (14.3)
Abdominal pain 0 4 (22.2) 4 (11.4)
TEAE, treatment-emergent adverse event.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628658
assessed as the mean of 2 sequential clinic readings. The response
achieved with relacorilant is noteworthy considering that ABPM
provides a more accurate representation of blood pressure over a
24-h time period for patients (22,24). Although the response
criteria threshold (5 mmHg) appears similar between the
studies, they are not equivalent. Average clinic assessments by
trained staff are 10/5 mmHg (SBP/DBP) higher than 24-h ABPM
(31), and treatment-associated mean 24-h AMBP reductions are
disproportionately less than ofce blood pressure reductions (22,
32). Thus, these phase 2 data suggest a potential benet for
relacorilant in improving hypertension in patients with CS.
For patients in the hyperglycemia subgroup, 15.4% (2/13) of
the low-dose group and 50.0% (6/12) the high-dose group met
the ad-hoc criteria for a clinically meaningful hyperglycemia
response, dened as either a decrease in HbA1c of 0.5% from
baseline or a normalization or improvement by 50 mg/dL (2.8
mmol/L) of the 2-h glucose value on the oGTT or a decrease in
antidiabetic medication. These criteria were selected because
each of them have been used in previous studies to
demonstrate a clinically meaningful improvement in the
hyperglycemia status of a patient (3337). Initially, responder
criteria were selected for the hyperglycemia population,
modelled after the SEISMIC trial. In the SEISMIC trial
population, the mean baseline HbA1c (7.4%), 2-h postprandial
glucose (296.2 mg/dL [16.4 mmol/L]; data on le), and fasting
glucose (149.0 mg/dL [8.3 mmol/L]) were considerably higher
than in this trial population (mean baseline HbA1c 6.6%, mean
2-h postprandial glucose 229.4 mg/dL [12.7 mmol/L], mean
fasting glucose 119.3 mg/dL [6.6 mmol/L]). In SEISMIC, to
qualify as a responder, a 25% decrease in AUC
glucose
on the
oGTT without increase or addition of new antidiabetic
medication was required. Although the AUC standard would
have set an unsafely low glycemic goal in some individual cases
in the less severely hyperglycemic population of this study, 12%
(3/25) of the pooled hyperglycemia population achieved a 25%
decrease in AUC
glucose
from baseline, and more importantly, a
signicant decrease in AUC
glucose
from baseline was observed
overall. Fructosamine is another biomarker for glucose control
and has been shown to correlate closely with HbA1c and glucose
levels in clinical trials (26). In an exploratory analysis, signicant
improvement in serum fructosamine levels was observed in the
combined hyperglycemia population, further corroborating the
clinical improvements in hyperglycemia with relacorilant noted
when applying the ad-hoc responder criteria.
Signicant improvement in several additional exploratory
endpoints were noted in the pooled patient population. Serum
osteocalcin, a marker of bone formation, has been shown to
negatively correlate with serum cortisol (25). In this study,
relacorilant was associated with a signicant increase in serum
osteocalcin levels during treatment, indicating increased bone
turnover. Nonalcoholic fatty liver disease (NAFLD) is frequently
observed in patients with endogenous hypercortisolism (5). In a
previous case report, mifepristone treatment was associated with
improvement of NAFLD based on a marked reduction in liver
enzymes in a patient with hypercortisolism due to an adrenal
adenoma (38). Although formal assessments of hepatic steatosis
were not performed in this study, relacorilant was associated with
signicant decreases in both alanine aminotransferase and aspartate
aminotransferase values. Signicant improvement was also noted in
several coagulation parameters (Factor VIII, aPTT, and platelets),
cognition, depression, and QoL. These parameters are being further
examined in larger, randomized, placebo-controlled phase 3 studies
(clinicaltrials.gov NCT03697109, NCT04308590).
The most commonly associated TEAEs with relacorilant were
primarily musculoskeletal, gastrointestinal, or nervous system-
related in nature. Relacorilant was generally better tolerated
when patients were started at 100 mg in the low-dose group.
In the high-dose group, with a starting dose of 250 mg, a higher
rate of TEAEs and premature discontinuations were observed,
with the highest incidence of TEAEs observed early on in the trial
at the starting dose of 250 mg, followed by lower incidences on
continued treatment at the higher dose levels. This was
particularly apparent for musculoskeletal and gastrointestinal
TEAEs, suggesting that the higher starting dose prompted
symptoms consistent with rapid withdrawal from excess
cortisol. Symptoms of cortisol withdrawal (e.g., nausea, fatigue,
A
B
FIGURE 4 | Potassium levels by visit in the (A) low-dose group and (B) high-dose group.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 6628659
arthralgia, headache) have also been observed in patients treated
with mifepristone (16,19) and have been shown to last for
several weeks (19).
During treatment with the GR antagonist mifepristone,
substantial increases in ACTH and cortisol were noted (16,
17). In SEISMIC, UFC levels increased 7.7-fold, and 63% of all
patients had at least a 2-fold increase in ACTH. In the present
study, the respective median changes from baseline in UFC and
ACTH were 4.0 nmol/d (1.4 µg/24 h) and 3.7 pmol/L (16.8 pg/
mL) in the ACTH-dependent CS group and 7.0 nmol/d (2.6 µg/
24 h) and 0.2 pmol/L (0.9 pg/mL) in the adrenal group. The
lower increases in ACTH and cortisol levels observed with
relacorilant from this study were consistent with ndings from
studies conducted in healthy volunteers (39). Administration of
relacorilant to healthy subjects was shown to reverse the effects of
high-dose prednisone (25 mg) (39) and was not associated with
increases in ACTH (data on le). This may reect differences in
the degree of GR antagonism in different tissues at the assessed
dosages. Although relacorilant is a potent antagonist of the GR
receptor (39), its effects in different tissues (e.g., pituitary) (40)
appear to differ from mifepristone (16). Two patients with
Cushing disease due to a macroadenoma were enrolled in the
phase 2 study and received de novo treatment with relacorilant
for 12 weeks as preoperative management (40). During
treatment, their ACTH levels remained consistent with
baseline. Two weeks after their last dose of relacorilant,
presurgical imaging revealed a reduction in the size of their
tumors (40). In vitro studies of relacorilant are underway to
further investigate these ndings. Compared with patients
treated with mifepristone, the noticeably lower increases in
UFC and ACTH levels may also explain the absence of drug-
induced hypokalemia with relacorilant, and the greater benetin
patients with hypertension (16). Elevated cortisol in CS is
associated with activation of the mineralocorticoid receptors
upon saturation of the 11b-hydroxysteroid type 2 enzyme
receptors, leading to hypokalemia and hypertension (4143).
GR antagonism with mifepristone, leading to a further increase
in cortisol levels, can exacerbate these symptoms (19). In fact,
hypokalemia occurred in 44% of patients treated with
mifepristone in the 24-week clinical trial of 50 adults with
endogenous CS (16). Also, unlike mifepristone, there was no
drug-induced vaginal bleeding in the study due to the lack of
activity at the progesterone receptor.
This study has a number of limitations. First, it is an open-
label, phase 2 dose-nding study with a relatively small sample
size (no sample size calculation) and short treatment duration,
and there was potential heterogeneity in the dose escalation
scheme based on the investigatorsclinical judgment. As part of
the study design, the low- and high-dose treatment groups were
different durations and were performed sequentially, not in
parallel, limiting the conclusions regarding a dose response.
Also, because of the small sample size, select data from
patients with major protocol deviations were included in the
analyses of clinical response. Finally, an ad-hoc analysis of
hyperglycemia response had to be conducted based on the
patient population enrolled; many of the outcome assessments
were exploratory in nature; and no adjustments for multiple
comparisons were performed. Nonetheless, these phase 2 results
provide the rst clinical evidence to suggest that relacorilant may
offer the clinical benetofpotentandhighlyspecic
glucocorticoid modulation in patients with endogenous CS
without the undesirable effects mediated by mifepristones
activity at the progesterone receptor or its frequent
mineralocorticoid activation via its marked elevation of cortisol
levels. Increases in ACTH and cortisol with relacorilant were
substantially less than with mifepristone, which may greatly
benet patients.
Two larger phase 3 studies to further investigate the clinical
efcacy and safety of relacorilant in patients with endogenous CS
and hypertension or IGT/T2DM and in patients with CS caused
by a cortisol-producing adrenal adenoma or bilateral hyperplasia
are in progress. The safety data from this phase 2 study suggest
that patients should begin relacorilant treatment at a relatively
low dose. Because the starting dose of 100 mg in the low-dose
group was well tolerated, it was chosen as the starting dose in the
phase 3 trials of relacorilant. In those trials, patients begin
treatment at 100 mg/d for 2 weeks and the dose is gradually
escalated, enhancing tolerability and treatment persistence.
Further examination of the tissue specicity of relacorilant is
also underway, including the effect of relacorilant on pituitary
size in the phase 3 study. In vitro studies of relacorilant are being
conducted to further investigate the different effects of
relacorilant and mifepristone on cortisol and somatostatin
receptor expression.
DATA AVAILABILITY STATEMENT
The datasets presented in this study can be found in online
repositories. The names of the repository/repositories and
accession number(s) can be found below: Grouped datasets
analyzed during the current study are available here. Additional
individual datasets generated during and/or analyzed are not
publicly available but are available from the corresponding
author on reasonable request.
ETHICS STATEMENT
The studies involving human participants were reviewed and
approved by the institutional review board at each study center.
The patients/participants provided their written informed
consent to participate in this study.
AUTHOR CONTRIBUTIONS
Study concept and design: AM. Study investigators who provided
study materials and/or patients: RP, IB, RF, AK, JK, MG, CM,
and MT. Statistical analysis: NE. Analyzed and interpreted
clinical data: All authors. Wrote manuscript or critically
revised it for content: All authors. All authors contributed to
the article and approved the submitted version.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 66286510
FUNDING
This study was funded by Corcept Therapeutics. Open Access
publication fees were paid by Corcept Therapeutics.
ACKNOWLEDGMENTS
The authors thank the patients who participated in the study, as
well as the investigators and clinical research staff from the study
centers. The authors also thank Dat Nguyen, PharmD, of
Corcept Therapeutics and Nicole Cooper, MS, and Sarah
Mizne, PharmD, of MedVal Scientic Information Services,
LLC for medical writing and editorial assistance, which was
funded by Corcept Therapeutics. This manuscript was prepared
according to the International Society for Medical Publication
Professionals’“Good Publication Practice for Communicating
Company-Sponsored Medical Research: GPP3.Data from this
paper were presented at the American Association of Clinical
Endocrinologists Annual Congress, May 16-20, 2018, Boston,
MA. Abstract 1219.
REFERENCES
1. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM,
et al. The Diagnosis of Cushings Syndrome: An Endocrine Society Clinical
Practice Guideline. J Clin Endocrinol Metab (2008) 93:152640. doi: 10.1210/
jc.2008-0125
2. Ferrau F, Korbonits M. Metabolic Syndrome in Cushings Syndrome Patients.
Front Horm Res (2018) 49:85103. doi: 10.1159/000486002
3. van der Pas R, Leebeek FW, Hoand LJ, de Herder WW, Feelders RA.
Hypercoagulability in Cushings Syndrome: Prevalence, Pathogenesis and
Treatment. Clin Endocrinol (Oxf) (2013) 78:4818. doi: 10.1111/cen.12094
4. Minetto M, Reimondo G, Osella G, Ventura M, Angeli A, Terzolo M. Bone
Loss Is More Severe in Primary Adrenal Than in Pituitary-Dependent
Cushings Syndrome. Osteoporos Int (2004) 15:85561. doi: 10.1007/
s00198-004-1616-3
5. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson JP, et al.
Hepatic Steatosis in Cushings Syndrome: A Radiological Assessment Using
Computed Tomography. Eur J Endocrinol (2003) 149:5438. doi: 10.1530/
eje.0.1490543
6. Debono M, Bradburn M, Bull M, Harrison B, Ross RJ, Newell-Price J. Cortisol
as a Marker for Increased Mortality in Patients With Incidental
Adrenocortical Adenomas. J Clin Endocrinol Metab (2014) 99:446270.
doi: 10.1210/jc.2014-3007
7. Di Dalmazi G, Vicennati V, Garelli S, Casadio E, Rinaldi E, Giampalma E,
et al. Cardiovascular Events and Mortality in Patients With Adrenal
Incidentalomas That Are Either non-Secreting or Associated With
Intermediate Phenotype or Subclinical Cushings Syndrome: A 15-Year
Retrospective Study. Lancet Diabetes Endocrinol (2014) 2:396405.
doi: 10.1016/s2213-8587(13)70211-0
8. Dekkers OM, Horváth-Puhó E, Jorgensen JO, Cannegieter SC, Ehrenstein V,
Vandenbroucke JP, et al. Multisystem Morbidity and Mortality in Cushings
Syndrome: A Cohort Study. J Clin Endocrinol Metab (2013) 98:227784.
doi: 10.1210/jc.2012-3582
9. Geer EB, Shaq I, Gordon MB, Bonert V, Ayala A, Swerdloff RS, et al.
Biochemical Control During Long-Term Follow-Up of 230 Adult Patients
With Cushing Disease: A Multicenter Retrospective Study. Endocr Pract
(2017) 23:96270. doi: 10.4158/ep171787.Or
10. Lacroix A, Feelders RA, Stratakis CA, Nieman LK. Cushings Syndrome.
Lancet (2015) 386:91327. doi: 10.1016/s0140-6736(14)61375-1
11. Pivonello R, Isidori AM, De Martino MC, Newell-Price J, Biller BM, Colao A.
Complications of Cushings Syndrome: State of the Art. Lancet Diabetes
Endocrinol (2016) 4:61129. doi: 10.1016/s2213-8587(16)00086-3
12. Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price J, Savage MO,
et al. Treatment of Cushings Syndrome: An Endocrine Society Clinical
Practice Guideline. J Clin Endocrinol Metab (2015) 100:280731.
doi: 10.1210/jc.2015-1818
13. Pivonello R, De Leo M, Cozzolino A, Colao A. The Treatment of Cushings
Disease. Endocr Rev (2015) 36:385486. doi: 10.1210/er.2013-1048
14. Feelders RA, Newell-Price J, Pivonello R, Nieman LK, Hoand LJ, Lacroix A.
Advances in the Medical Treatment of Cushings Syndrome. Lancet Diabetes
Endocrinol (2019) 7:30012. doi: 10.1016/s2213-8587(18)30155-4
15. Pivonello R, Ferrigno R, De Martino MC, Simeoli C, Di Paola N, Pivonello C,
et al. Medical Treatment of Cushings Disease: An Overview of the Current
and Recent Clinical Trials. Front Endocrinol (Lausanne) (2020) 11:648.
doi: 10.3389/fendo.2020.00648
16. Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart DE, Gross C.
Mifepristone, a Glucocorticoid Receptor Antagonist, Produces Clinical and
Metabolic Benets in Patients With Cushings Syndrome. J Clin Endocrinol
Metab (2012) 97:203949. doi: 10.1210/jc.2011-3350
17. Fleseriu M, Findling JW, Koch CA, Schlaffer S-M, Buchfelder M, Gross C.
Changes in Plasma ACTH Levels and Corticotroph Tumor Size in Patients
With Cushings Disease During Long-Term Treatment With the
Glucocorticoid Receptor Antagonist Mifepristone. J Clin Endocrinol Metab
(2014) 99:371827. doi: 10.1210/jc.2014-1843
18. Brown DR, East HE, Eilerman BS, Gordon MB, King EE, Knecht LA, et al.
Clinical Management of Patients With Cushing Syndrome Treated With
Mifepristone: Consensus Recommendations. Clin Diabetes Endocrinol (2020)
6:18. doi: 10.1186/s40842-020-00105-4
19. Yuen KC, Williams G, Kushner H, Nguyen D. Association Between
Mifepristone Dose, Efcacy, and Tolerability in Patients With Cushing
Syndrome. Endocr Pract (2015) 21:108792. doi: 10.4158/ep15760.Or
20. Hunt HJ, Belanoff JK, Walters I, Gourdet B, Thomas J, Barton N, et al.
Identication of the Clinical Candidate (R)-(1-(4-Fluorophenyl)-6-((1-
Methyl-1H-Pyrazol-4-Yl)Sulfonyl)-4,4a,5,6,7,8-Hexahydro-1H-Pyrazolo[3,4-
G]Isoquinolin-4a-Yl)(4-(Triuoromethyl)Pyridin-2-Yl)Methanone
(CORT125134): A Selective Glucocorticoid Receptor (GR) Antagonist. J Med
Chem (2017) 60:340521. doi: 10.1021/acs.jmedchem.7b00162
21. Fassnacht M, Arlt W, Bancos I, Dralle H, Newell-Price J, Sahdev A, et al.
Management of Adrenal Incidentalomas: European Society of Endocrinology
Clinical Practice Guideline in Collaboration With the European Network for
the Study of Adrenal Tumors. Eur J Endocrinol (2016) 175:G134.
doi: 10.1530/eje-16-0467
22. OBrien E, Parati G, Stergiou G, Asmar R, Beilin L, Bilo G, et al. European
Society of Hypertension Position Paper on Ambulatory Blood Pressure
Monitoring. J Hypertens (2013) 31:173168. doi: 10.1097/HJH.0b013e
328363e964
23. Grant RW, Donner TW, Fradkin JE, Hayes C, Herman WH, Hsu WC, et al.
Standards of Medical Care in Diabetes2015. Diabetes Care (2015) 38:S193.
doi: 10.2337/dc15-S003
24. UngerT,BorghiC,CharcharF,KhanNA,PoulterNR,PrabhakaranD,etal.2020
International Society of Hypertension Global Hypertension Practice Guidelines.
Hypertension (2020) 75:133457. doi: 10.1161/hypertensionaha.120.15026
25. Tóth M, Grossman A. Glucocorticoid-Induced Osteoporosis: Lessons From
Cushings Syndrome. Clin Endocrinol (Oxf) (2013) 79:111. doi: 10.1111/
cen.12189
26. Malmström H, Walldius G, Grill V, Jungner I, Gudbjörnsdottir S, Hammar N.
Fructosamine Is a Useful Indicator of Hyperglycaemia and Glucose Control in
Clinical and Epidemiological StudiesCross-Sectional and Longitudinal
Experience From the AMORIS Cohort. PLoS One (2014) 9:e111463.
doi: 10.1371/journal.pone.0111463
27. Webb SM, Badia X, Barahona MJ, Colao A, Strasburger CJ, Tabarin A, et al.
Evaluation of Health-Related Quality of Life in Patients With Cushings
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 66286511
Syndrome With a New Questionnaire. Eur J Endocrinol (2008) 158:62330.
doi: 10.1530/eje-07-0762
28. Beck AT, Steer RA, Brown GK. Manual for the Beck Depression Inventory-Ii.
San Antonio, TX: Psychological Corporation (1996).
29. Tombaugh TN. Trail Making Test A and B: Normative Data Stratied by Age
and Education. Arch Clin Neuropsychol (2004) 19:20314. doi: 10.1016/s0887-
6177(03)00039-8
30. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC.
Homeostasis Model Assessment: Insulin Resistance and Beta-Cell Function
From Fasting Plasma Glucose and Insulin Concentrations in Man.
Diabetologia (1985) 28:4129. doi: 10.1007/bf00280883
31. Head GA, Mihailidou AS, Duggan KA, Beilin LJ, Berry N, Brown MA, et al.
Denition of Ambulatory Blood Pressure Targets for Diagnosis and
Treatment of Hypertension in Relation to Clinic Blood Pressure:
Prospective Cohort Study. BMJ (2010) 340:c1104. doi: 10.1136/bmj.c1104
32. Schmieder RE, Schmidt ST, Riemer T, Dechend R, Hagedorn I, Senges J, et al.
Disproportional Decrease in Ofce Blood Pressure Compared With 24-Hour
Ambulatory Blood Pressure With Antihypertensive Treatment: Dependency
on Pretreatment Blood Pressure Levels. Hypertension (2014) 64:106772.
doi: 10.1161/hypertensionaha.113.03140
33. Gerich J. Pathogenesis and Management of Postprandial Hyperglycemia: Role
of Incretin-Based Therapies. Int J Gen Med (2013) 6:87795. doi: 10.2147/
ijgm.S51665
34. Esposito K, Giugliano D, Nappo F, Marfella R. Campanian Postprandial
Hyperglycemia Study Group. Regression of Carotid Atherosclerosis by
Control of Postprandial Hyperglycemia in Type 2 Diabetes Mellitus.
Circulation (2004) 110:2149. doi: 10.1161/01.Cir.0000134501.57864.66
35. Castinetti F, Guignat L, Giraud P, Muller M, Kamenicky P, Drui D, et al.
Ketoconazole in Cushings Disease: Is it Worth a Try? J Clin Endocrinol Metab
(2014) 99:162330. doi: 10.1210/jc.2013-3628
36. GoldbergRB, Einhorn D, Lucas CP, Rendell MS, Damsbo P, Huang WC, et al. A
Randomized Placebo-Controlled Trial of Repaglinide in the Treatment of Type
2Diabetes.Diabetes Care (1998) 21:1897903. doi: 10.2337/diacare.21.11.1897
37. Marre M, VanGaal L, Usadel KH, BallM, Whatmough I, Guitard C. Nateglinide
Improves Glycaemic Control When Added to Metformin Monotherapy: Results
of a Randomized Trial With Type 2 Diabetes Patients. Diabetes Obes Metab
(2002) 4:17786. doi: 10.1046/j.1463-1326.2002.00196.x
38. Ragucci E, Nguyen D, Lamerson M, Moraitis AG. Effects of Mifepristone on
Nonalcoholic Fatty Liver Disease in a Patient With a Cortisol-Secreting
Adrenal Adenoma. Case Rep Endocrinol (2017) 2017:6161348. doi: 10.1155/
2017/6161348
39. Hunt H, Donaldson K, Strem M, Zann V, Leung P, Sweet S, et al. Assessment
of Safety, Tolerability, Pharmacokinetics, and Pharmacological Effect of Orally
Administered CORT125134: An Adaptive, Double-Blind, Randomized,
Placebo-Controlled Phase 1 Clinical Study. Clin Pharmacol Drug Dev
(2018) 7:40821. doi: 10.1002/cpdd.389
40. Terzolo M, Iacuaniello D, Pia A, Adriano P, Moraitis A, Pivonello R. Tumor
Shrinkage With Preoperative Relacorilant Therapy in Two Patients With
Cushing Disease Due to Pituitary Macroadenomas [Abstract]. J Endocr Soc
(2019) 3(suppl 1):SUN463. doi: 10.1210/js.2019-SUN-463
41. Torpy DJ, Mullen N, Ilias I, Nieman LK. Association of Hypertension and
Hypokalemia With Cushings Syndrome Caused by Ectopic ACTH Secretion:
A Series of 58 Cases. Ann N Y Acad Sci (2002) 970:13444. doi: 10.1111/
j.1749-6632.2002.tb04419.x
42. Howlett TA, Drury PL, Perry L, Doniach I, Rees LH, Besser GM. Diagnosis
and Management of ACTH-Dependent Cushings Syndrome: Comparison of
the Features in Ectopic and Pituitary ACTH Production. Clin Endocrinol
(Oxf) (1986) 24:699713. doi: 10.1111/j.1365-2265.1986.tb01667.x
43. Isidori AM, Graziadio C, Paragliola RM, Cozzolino A, Ambrogio AG, Colao
A, et al. The Hypertension of Cushings Syndrome: Controversies in the
Pathophysiology and Focus on Cardiovascular Complications. J Hypertens
(2015) 33:4460. doi: 10.1097/hjh.0000000000000415
Conict of Interest: The authors declare that this study received funding from
Corcept Therapeutics (Menlo Park, CA, USA). The funder had a role in study
design, data collection and analysis, and AM, as an author of the manuscript and
employee of Corcept Therapeutics, had a role in the study design, the decision to
publish, the interpretation of clinical data, the revision of the manuscript, and
approval of the nal manuscript to submit. Open Access publication fees were
paid by Corcept Therapeutics. RP: Consultant: Ferring, Ipsen, Novartis, Pzer,
ViroPharma-Shire; Speaker: Novartis, ViroPharma-Shire; Research support:
Corcept Therapeutics, Novartis, ViroPharma-Shire; Grant support: IBSA,
Novartis, Pzer, ViroPharma-Shire. IB: Consultant: HRA Pharma, Sparrow
Pharmaceutics, Strongbridge; Data and Safety Monitoring Panel, Adrenas. RF:
Consultant: Corcept Therapeutics; Speaker: HRA Pharma. AK: Consultant:
Strongbridge; Research support: Corcept Therapeutics. MG: Research support:
Corcept Therapeutics, Crinetics, Ionis, Ipsen, Novartis, Novo Nordisk, Opko,
Strongbridge, Teva. CM: Consultant: Horizon Therapeutics; Research support:
Corcept Therapeutics, Eli Lilly, Medtronic. MT: Consultant: HRA Pharma;
Research support: Corcept Therapeutics. NE: Consultant: Corcept Therapeutics,
Pentara, Trialwise. AM: Employee: Corcept Therapeutics.
The remaining author (JK) declares that the research was conducted in the absence
of any commercial or nancial relationships that could be construed as a potential
conict of interest.
The reviewer AALC declared a shared afliation with one of the authors, RP, to the
handling editor at time of review.
Author NE was employed by company Trialwise.
Copyright © 2021 Pivonello, Bancos, Feelders, Kargi, Kerr, Gordon, Mariash, Terzolo,
Ellison and Moraitis. This is an open-access article distributed under the terms of the
Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) and the
copyright owner(s) are credited and that the original publication in this journal is
cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Pivonello et al. Relacorilant in Endogenous Hypercortisolism
Frontiers in Endocrinology | www.frontiersin.org July 2021 | Volume 12 | Article 66286512
... Relacorilant was employed in seven patients with ACTH-independent Cushing's syndrome, including patients with MACS, and in the low-dose group (100-200 mg/d; n = 17), 5/12 patients (41.7%) with hypertension achieved a good response. In the high-dose group (250-400 mg/d; n = 18), 7/11 patients (63.6%) with hypertension achieved a good response [42]. ...
... Baxdrostat, an aldosterone synthase inhibitor, is also a promising therapeutic option for patients with resistant hypertension and/or aldosterone oversecretion [45] (Figure 1). [42]. Due to the high cardiovascular risk linked to hypercortisolism, the goals of hypertension treatment should be to maintain BP levels below 130/80 mmHg. ...
... In terms of the main findings, in relation to diabetes control, they reported that in the low-dose group (100-200 mg/d; n = 17), 2/13 patients (15.4%) with hyperglycemia had a good response. In the high-dose group (250-400 mg/d; n = 18), 6/12 patients (50%) with hyperglycemia had a good response [42]. ...
Article
Full-text available
Mild autonomous cortisol secretion (MACS) is associated with a higher cardiometabolic risk than that observed in patients with nonfunctioning adrenal adenomas and in the general population. In patients with MACS, the excess of glucocorticoids affects various metabolic pathways, leading to different manifestations of metabolic syndrome and other comorbidities. Hypertension and diabetes mellitus are two of the most common cardiometabolic comorbidities associated with MACS, reaching a prevalence of up to 80% and up to 40%, respectively. In addition, they are the comorbidities that experienced a greater improvement after adrenalectomy in patients with MACS. Hypertension pathogenesis is multifactorial, including the coexistence of comorbidities such as obesity or diabetes and the role of the different polymorphisms of the glucocorticoid receptor gene, among others. Glucocorticoid-induced diabetes mellitus is mainly related to the detrimental effects of glucocorticoids on insulin-dependent glucose uptake in peripheral tissues, gluconeogenesis and insulin secretion. There are no specific recommendations for hypertension and diabetes treatment in patients with MACS. Thus, considering the similar underlying pathogenesis of hypertension and diabetes mellitus in overt and mild hypercortisolism, our recommendation is to follow this general stepwise approach: surgically remove the adrenal culprit lesion to induce remission from hypercortisolism; control hypercortisolism with steroidogenesis inhibitors; and treat elevated blood pressure or high glucose levels using carefully selected anti-hypertensives and glucose-lowering medications if blood pressure and glucose levels remain uncontrolled, respectively. In this review, we summarize the epidemiology, physiopathology and management of diabetes mellitus and hypertension in patients with MACS.
... Relacorilant is a highly selective glucocorticoid receptor antagonist that does not affect progesterone receptors. In a phase II trial in CS, it reduced hyperglycemia, hypertension, while increase in cortisol and ACTH were lower vs those with mifepristone [129]. ...
Article
Full-text available
Context Functioning pituitary adenomas (FPAs) include most frequently prolactinomas, somatotroph or corticotroph adenomas, while thyrotroph and gonadotroph adenomas are very rare. Despite their benign histological nature (aggressive tumors are rare and malignant ones exceptional), FPAs could cause significant morbidity and increased mortality due to complications associated with hormonal excess syndromes and/or mass effect leading to compression of adjacent structures. This mini review will focus on the increasing role of medical therapy in the multimodal treatment, which also includes transsphenoidal surgery (TSS) and radiotherapy. Evidence synthesis Most patients with prolactinomas are treated only with medications, but surgery could be considered for some patients in a specialized pituitary center, if higher chances of cure. Dopamine agonists, especially cabergoline, are efficient in reducing tumor size and normalizing prolactin. TSS is the first-line treatment for all other FPAs, but most patients require complex adjuvant treatment, including a combination of therapeutic approaches. Medical therapy is the cornerstone of treatment in all patients after unsuccessful surgery or when surgery cannot be offered and includes somatostatin receptor ligands and dopamine agonists (almost all FPAs), growth hormone receptor antagonists (acromegaly), adrenal steroidogenesis inhibitors and glucocorticoid receptor blockers (Cushing’s disease). Novel medical treatments, especially for acromegaly and Cushing’s disease are under research. Conclusions An enlarged panel of effective drugs available with increased knowledge of predictive factors for response and/or adverse effects will enhance the possibility to offer a more individualized treatment. This would not only improve disease control and prognosis, but also quality of life.
... However, GR antagonism did not alter adipose tissue weight and lipid accumulation in DHT-treated mice. In other models for metabolic disease, benefits on metabolic health were observed on treatment with GR antagonists [54][55][56][57][58]. It thus seems that many activities of GR antagonists are lost in PCOS, possibly due to elevated androgen exposure that potentially takes over (part of) glucocorticoid effects. ...
Article
Full-text available
Context Polycystic ovary syndrome (PCOS) is a complex metabolic disorder associated with obesity, insulin resistance, and dyslipidemia. Hyperandrogenism is a major characteristic of PCOS. Increased androgen exposure is believed to deregulate metabolic processes in various tissues as part of the PCOS pathogenesis, predominantly through the androgen receptor (AR). Notably, various metabolic features in PCOS are similar to those observed after excess glucocorticoid exposure. Objective We hypothesized that glucocorticoid receptor (GR) signaling is involved in the metabolic symptoms of PCOS. Methods In a PCOS model of chronic dihydrotestosterone (DHT) exposure in female mice, we investigated whether GR signaling machinery was (de)regulated, and if treatment with a selective GR antagonist alleviated the metabolic symptoms. Results We observed an upregulation of GR mRNA expression in the liver after DHT exposure. In white adipose tissues and liver we found that DHT upregulated Hsd11b1, which encodes for the enzyme that converts inactive into active glucocorticoids. We found that preventive but not therapeutic administration of a GR antagonist alleviated DHT-induced hyperglycemia and restored glucose tolerance. We did not observe strong effects of GR antagonism in DHT-exposed mice on other features like total fat mass and lipid accumulation in various tissues. Conclusion We conclude that GR activation may play a role in glucose metabolism in DHT-exposed mice.
Article
Full-text available
The primary goal of acromegaly treatment is to normalize biochemical parameters as it significantly reduces the risks of complications and comorbidities associated with the disease. First-line medical treatment is commonly represented by injectable somatostatin analogues (SRLs) after surgery. In June 2020, with the integration of Transient Permeation Enhancer® technology, oral octreotide capsules (OOCs) received regulatory approval from the US Food and Drug Administration for long-term maintenance treatment in patients with acromegaly who have responded to and tolerated treatment with octreotide or lanreotide. We reviewed the clinical pharmacological data on the development and clinical use of OOCs. The pharmacokinetic and pharmacodynamic data on OOCs showed a dose–dependent increase in octreotide levels and remarkable suppression of growth hormone secretion. The efficacy and safety of OOCs were investigated in four clinical trials conducted on patients with complete or partially controlled acromegaly. The trials resulted in the maintenance of biochemical control after switching from injectable SRLs to OOCs, with a comparable side-effect profile. Moreover, the acromegaly symptoms improved in patients on OOC. The data showed a patient preference to continue in the OOC arm for the extension phase of the trials. From the clinical pharmacological perspective, oral formulation of octreotide has the advantage of efficacy and safety with respect to injectable octreotide.
Article
Full-text available
Glucocorticoids exert pleiotropic effects on all tissues to regulate cellular and metabolic homeostasis. Synthetic forms are used therapeutically in a wide range of conditions for their anti-inflammatory benefits, at the cost of dose and duration-dependent side effects. Significant variability occurs between tissues, disease states, and individuals with regard to both the beneficial and deleterious effects. The glucocorticoid receptor (GR) is the site of action for these hormones and a vast body of work has been conducted understanding its function. Traditionally, it was thought that the anti-inflammatory benefits of glucocorticoids were mediated by transrepression of pro-inflammatory transcription factors, while the adverse metabolic effects resulted from direct transactivation. This canonical understanding of the GR function has been brought into question over the past 2 decades with advances in the resolution of scientific techniques, and the discovery of multiple isoforms of the receptor present in most tissues. Here we review the structure and function of the GR, the nature of the receptor isoforms, and the contribution of the receptor to glucocorticoid sensitivity, or resistance in health and disease.
Article
Purpose The majority of patients with metastatic prostate cancer who receive androgen-deprivation therapy and androgen receptor (AR) signaling inhibitors (ARSI) progress. Activation of the glucocorticoid receptor (GR) is associated with ARSI resistance. This single-arm phase I trial assessed safety and pharmacokinetic (PK) feasibility of a combined AR antagonist (enzalutamide) and selective GR modulator (relacorilant) in patients with metastatic castration-resistant prostate cancer (mCRPC). Patients and Methods This was a phase I trial (NCT03674814) of relacorilant and enzalutamide in patients with refractory mCRPC enrolled using a 6+3 design. The enzalutamide dose was kept constant at 120 mg/d with escalating doses of relacorilant based on safety and PK measures in cohorts of ≥6 patients. The primary objective was safety and establishment of pharmacologically active doses. Secondary objectives were related to antitumor activity. Results Thirty-five patients with mCRPC were enrolled. Twenty-three were accrued across three dose cohorts in the dose-escalation phase, and 12 enrolled at the recommended phase II dose. The combination was generally well tolerated, safe, and achieved desirable enzalutamide PK. RP2D of 120 + 150 mg/d, respectively, was established. Median time on study was 2.2 months with four patients remaining on study for longer than 11 months. Four of 12 evaluable patients had a prostate-specific antigen (PSA) partial response. Conclusions This is the first prospective trial combining an AR antagonist and a nonsteroidal selective GR modulator. The combination was safe and well tolerated with PSA response and prolonged disease control observed in a limited subset of patients. Further prospective trials are justified to evaluate efficacy and identify predictive biomarkers of response.
Article
Cushing’s Disease (CD), hypercortisolism due to pituitary ACTH secreting neuroendocrine neoplasm, is associated with increased morbidity and, if untreated, mortality in about half of the affected individuals. Consequently, the timely initiation of effective treatment is mandatory. Neurosurgery is the first line and the only potentially curative treatment; however, 30% of patients will have persistent disease post-surgery. Furthermore, a small percentage of those initially controlled will develop hypercortisolism during long-term follow- up. Therefore, patients with persistent or recurrent disease, as well as those considered non-eligible for surgery, will need a second-line therapeutic approach, i.e., pharmacotherapy. Radiation therapy is reserved as a third-line therapeutic option due to its slower onset of action and its unfavorable profile regarding complications. During the past few years, the understanding of molecular mechanisms implicated in the physiology of the hypothalamus-pituitary-adrenal axis has evolved, and new therapeutic targets for CD have emerged. In the present review, currently available treatments, compounds currently tested in ongoing clinical trials, and interesting, potentially new targets emerging from unraveling molecular mechanisms involved in the pathophysiology of Cushing’s disease are discussed.
Article
Objetivo: Avaliar a eficácia e a segurança das novas abordagens terapêuticas no tratamento da síndrome de Cushing. Métodos: Através da plataforma de dados PubMed, foi realizada uma Revisão Integrativa a fim de encontrar fontes relevantes para este intuito. A pesquisa foi realizada através da estratégia de pesquisa: ("Cushing Syndrome"[MeSH]) AND ((Treatment) OR (Chronotherapy) OR (Steroid Synthesis Inhibitors) OR (Antiglucocorticoid)). Foram 600 artigos inicialmente e através da aplicação dos critérios de inclusão e exclusão, 17 artigos foram selecionados para análise neste estudo. Resultados: Estudos apontam que Osilodrostat é seguro e efetivo controlando o hipercortisolismo, porém têm variados efeitos adversos. Cetoconazol é eficaz e seguro para doença ativa após cirurgia transesfenoidal. Pasireotide apresenta melhoria da qualidade de vida, sinais clínicos e possui diversos efeitos colaterais. O ácido 9-cis retinoico pode ser benéfico com efeito adrenal adicional e o S-707106 melhora sensibilidade à insulina e reduz obesidade. Levocetoconazol mostrou efetividade no quadro clínico, tolerabilidade e segurança. Relacorilant apresentou benefícios clínicos e Metirapona mostrou boa resolubilidade e segurança. Nas recidivas, alguns pontos devem ser considerados quando utilizarem radioterapia externa como terapia auxiliar. Considerações finais: As novas abordagens terapêuticas estudadas podem ser consideradas seguras e eficazes para a síndrome de Cushing.
Article
Full-text available
Cushing's disease (CD) is a serious endocrine disorder characterized by chronic hypercortisolism, or Cushing's syndrome (CS), caused by a corticotroph pituitary tumor, which induces an excessive adrenocorticotropic hormone (ACTH) and consequently cortisol secretion. CD presents a severe clinical burden, with impairment of the quality of life and increase in mortality. Pituitary surgery represents the first-line therapy, but it is non-curative in one third of patients, requiring additional treatments. Among second-line treatments, medical therapy is gradually gaining importance, although the current medical treatments are unable to reach optimal efficacy and safety profile. Therefore, new drugs and new formulations of presently available drugs are currently under clinical investigation in international clinical trials, in order to assess their efficacy and safety in CD, or in the general population of CS. Among pituitary-directed agents, pasireotide, in the twice-daily subcutaneous formulation, has been demonstrated to be an effective treatment both in clinical trials and in real-world studies, and extension studies of the phase II and III clinical trials reported evidence of long-term efficacy with general good safety profile, although associated with frequent hyperglycemia, which requires monitoring of glucose metabolism. Moreover, the most recent once-monthly intramuscular formulation, pasireotide long-acting release (LAR), showed similar efficacy and safety, but associated with potential better compliance profile in CD. Roscovitine is an experimental drug currently under investigation. Among adrenal-directed agents, metyrapone is the only historical agent currently under investigation in a prospective, multicenter, international clinical trial, that would likely clarify its efficacy and safety in a large population of patients with CS. Osilodrostat, a novel agent with a mechanism of action similar to metyrapone, seems to offer a rapid, sustained, and effective disease control of CD, according to recently completed clinical trials, whereas levoketoconazole, a different chemical formulation of the historical agent ketoconazole, is still under investigation in clinical trials, with preliminary evidences showing an effective and safe control of CS. ATR-101 is an experimental drug currently under investigation. Among glucocorticoid receptor-directed drugs, mifepristone has been demonstrated to improve clinical syndrome and comorbidities, especially hypertension and impairment of glucose metabolism, but the occurrence of hypokalemia and in women uterine disorders, due to the concomitant action on progestin receptor, requires caution, whereas the preliminary evidence on relacorilant, characterized by high selectivity for glucocorticoid receptor, suggested good efficacy in the control of hypertension and impairment of glucose metabolism, as well as a good safety profile, in CS. Finally, a limited experience has demonstrated that combination therapy might be an interesting approach in the management of CD. The current review provides a summary of the available evidences from current and recent clinical trials on CD, with a specific focus on preliminary data.
Article
Full-text available
Background While surgery is the first-line treatment for patients with endogenous hypercortisolism (Cushing syndrome [CS]), mifepristone has been shown to be a beneficial medical treatment option, as demonstrated in the SEISMIC (Study of the Efficacy and Safety of Mifepristone in the Treatment of Endogenous Cushing Syndrome) trial. Mifepristone is a competitive glucocorticoid receptor antagonist and progesterone receptor antagonist that is associated with several treatment effects and adverse events that clinicians need to be aware of when considering its use. The objective of this review was to provide updated clinical management recommendations for patients with CS treated with mifepristone. Methods A panel of endocrinologists from the US with extensive experience in treating patients with CS, including with mifepristone, convened as part of a clinical advisory board to develop a consensus on the practical, real-world clinical management of patients on mifepristone. Results Comprehensive considerations and recommendations are provided for managing mifepristone-associated effects, including symptoms of cortisol withdrawal, hypokalemia, and change in thyroid function; effects related to its antiprogesterone activity; and rash. Additional management strategies to address concomitant medications and special clinical situations, such as surgery and use in specific populations, are also provided. Conclusion Safe and effective use of mifepristone requires clinical judgment and close patient monitoring to ensure optimal clinical outcomes. These consensus recommendations provide useful, practical guidance to clinicians using mifepristone.
Article
Full-text available
Cushing syndrome (CS), a complex, multisystemic condition resulting from prolonged exposure to cortisol, is frequently associated with nonalcoholic fatty liver disease (NAFLD). In patients with adrenal adenoma(s) and NAFLD, it is essential to rule out coexisting endocrine disorders like CS, so that the underlying condition can be properly addressed. We report a case of a 49-year-old woman with a history of hypertension, prediabetes, dyslipidemia, biopsy-confirmed steatohepatitis, and benign adrenal adenoma, who was referred for endocrine work-up for persistent weight gain. Overt Cushing features were absent. Biochemical evaluation revealed nonsuppressed cortisol on multiple 1-mg dexamethasone suppression tests, suppressed adrenocorticotropic hormone, and low dehydroepiandrosterone sulfate. The patient initially declined surgery and was treated with mifepristone, a competitive glucocorticoid receptor antagonist. In addition to improvements in weight and hypertension, substantial reductions in her liver enzymes were noted, with complete normalization by 20 weeks of therapy. This case suggests that autonomous cortisol secretion from adrenal adenoma(s) could contribute to the metabolic and liver abnormalities in patients with NAFLD. In conclusion, successful management of CS with mifepristone led to marked improvement in the liver enzymes of a patient with long-standing NAFLD.
Article
Full-text available
CORT125134 is an orally active, high-affinity, selective antagonist of the glucocorticoid receptor that is being developed for indications that may benefit from the modulation of cortisol activity. This first-in-human study was conducted to evaluate the dose-related safety, tolerability, pharmacokinetics and pharmacological effects of CORT125134 and its active metabolite CORT125201. Eighty-one healthy male or female subjects received a single dose of 5 to 500 mg CORT125134 or matching placebo across 9 cohorts; 1 cohort received 150 mg CORT125134 after a high-fat breakfast; and 46 subjects received 50 to 500 mg CORT125134 or matching placebo once daily for up to 14 days across 4 cohorts. CORT125134 was well tolerated at doses up to 250 mg per day for 14 days. CORT125134 was absorbed rapidly and eliminated with a mean half-life ranging from 11 to 19 hours. Steady state was achieved by day 7. Exposure increased in a greater than proportional manner, particularly at lower doses. Exposure to CORT125201 at steady state was less than 5% that of parent CORT125134. Evidence for the desired pharmacological effect (glucocorticoid receptor antagonism) was demonstrated by the ability of CORT125134 to prevent several effects of the glucocorticoid receptor agonist prednisone.
Article
Background: Relacorilant (RELA, Corcept Therapeutics) is a highly selective glucocorticoid receptor antagonist (GRA) that modulates the effects of excess cortisol, while showing no significant antagonism at the progesterone, mineralocorticoid, androgen, or estrogen receptors. It is currently being investigated for the treatment of patients with endogenous Cushing syndrome (CS) of any etiology. Results from the open-label Phase 2 study (NCT02804750) indicated that treatment with RELA was generally well tolerated and significantly improved glycemic and hypertension control in patients with endogenous hypercortisolism. In this Phase 2 study, two patients with Cushing disease (CD) showed evidence of tumor shrinkage on post-treatment MRIs performed after study completion. Cases: Two patients with de novo CD due to a macroadenoma were treated for 3 months with RELA prior to previously scheduled transsphenoidal pituitary surgeries. Pituitary imaging performed after treatment with RELA revealed significant shrinkage of the tumors. Patient 1 is a 50y/o woman with a pituitary macroadenoma measuring 10 x 6.3 mm on MRI. She presented with cushingoid features (moon face, dorsal & supraclavicular fat, plethora, central obesity, easy bruising, striae) and had confirmatory biochemical tests (ACTH 12.2 pmol/L (NR 1.3-11.1 pmol/L); UFC 177.7 nmol/d (NR 11.1-138 nmol/d); DST 156 ng/mL (15.6 mcg/dL). Patient 2 is a 43y/o man with a pituitary macroadenoma measuring 22 x 25 x 26 mm with suprasellar extension, right displacement of the pituitary stalk, and invasion of the left cavernous sinus on MRI. He also had cushingoid features and confirmatory biochemical tests (ACTH 20.9 pmol/L (NR 1.3-11.1 pmol/L); UFC 356.5 nmol/d (NR 11.1-138 nmol/d); DST 247 ng/mL (24.7 mcg/dL). After completing the 3-month study, pre-operative MRIs revealed reduction in the size of their tumors. The pituitary tumor for patient 1 decreased from 10 x 6.3 mm prior to treatment to 8.0 x 5.7 mm after treatment with RELA. The pituitary tumor for patient 2, decreased from 22 x 25 x 26 mm prior to treatment to 21 x 22 x 19 mm after treatment with RELA. Conclusion: Pituitary imaging performed as part of the pre-operative standard of care revealed tumor shrinkage in two patients with macroadenomas treated for 3 months with RELA. This unexpected finding will be formally investigated in an ongoing Phase 3 study with RELA (GRACE Study, NCT03697109). A potential explanation for this finding could be tumor sensitization to endogenous (hypothalamic) somatostatin via upregulation of somatostatin receptors type 2 (SSTR2) which are downregulated in patients with hypercortisolism. If confirmed, this finding may potentially support the role of RELA as pre-operative medical treatment in patients with CD with invasive macroadenomas.
Article
Cushing's syndrome is associated with multisystem morbidity and, when suboptimally treated, increased mortality. Medical therapy is an option for patients if surgery is not successful and can be classified into pituitary-directed drugs, steroid synthesis inhibitors, and glucocorticoid receptor antagonists. In the last decade there have been new developments in each drug category. Targeting dopamine and somatostatin receptors on corticotroph adenomas with cabergoline or pasireotide, or both, controls cortisol production in up to 40% of patients. Potential new targets in corticotroph adenomas include the epidermal growth factor receptor, cyclin-dependent kinases, and heat shock protein 90. Osilodrostat and levoketoconazole are new inhibitors of steroidogenesis and are currently being evaluated in multicentre trials. CORT125134 is a new selective glucocorticoid receptor antagonist under investigation. We summarise the drug therapies for various forms of Cushing's syndrome and focus on emerging drugs and drug targets that have the potential for new and effective tailor-made pharmacotherapy for patients with Cushing's syndrome.
Chapter
Cushing's syndrome (CS), including visceral obesity, dyslipidemia, hypertension and diabetes among its many manifestations, is "a model" of metabolic syndrome. Glucocorticoid (GC) excess, through a combination of effects on liver, muscle, adipose tissue and pancreas, increases gluconeogenesis and impairs insulin sensitivity, leading to carbohydrate abnormalities. Dyslipidemia is a common finding in CS as a consequence of GC-related increased lipolysis, lipogenesis and adipogenesis. CS patients experience typical changes in body composition, with fat redistribution resulting in accumulation of visceral adipose tissue. Hypertension, myocardial and vascular abnormalities along with the metabolic changes and the characteristic coagulopathy increase cardiovascular morbidity and mortality. Metabolic syndrome features can persist long after normalisation of cortisol levels.
Article
Objectives: Cushing's disease (CD) results from excessive exposure to glucocorticoids caused by an adrenocorticotropic hormone-secreting pituitary tumor. Inadequately-treated CD is associated with significant morbidity and elevated mortality. Multi-center data on CD patients treated in routine clinical practice are needed to assess treatment outcomes in this rare disorder. The study purpose was to describe the burden of illness and treatment outcomes for CD patients. Methods: Eight pituitary centers in 4 US regions participated in this multi-center retrospective chart review study. Subjects were CD patients diagnosed at ≥18 years of age within the past 20 years. Descriptive statistical analyses were conducted to examine presenting signs, symptoms, comorbidities, and treatment outcomes. Results: Of 230 patients, 79% were female (median age at diagnosis: 39 years, range 18-78). Length of follow-up was 0-27.5 years (median: 1.9). Pituitary adenomas were 0-51mm. The most common presenting comorbidities included hypertension (67.3%), polycystic ovary syndrome (43.5%), and hyperlipidemia (41.5%). Biochemical control was achieved with initial pituitary surgery in 41.4% (91/220), not achieved in 50.0% (110/220), and undetermined in 8.6% (19/220). At the end of follow-up, control had been achieved with a variety of treatment methods in 49.1% (110/224), not achieved in 29.9% (67/224) and undetermined in 21.0% (47/224). Conclusion: Despite multiple treatments, at the end of follow-up, biochemical control was still not achieved in up to 30% of patients. These multi-center data demonstrate that in routine clinical practice, initial and long-term control is not achieved in a substantial number of patients with CD.
Article
The non-selective glucocorticoid receptor (GR) antagonist, mifepristone, has been approved in the USA for the treatment of selected patients with Cushing's syndrome. Whilst this drug is highly effective, lack of selectivity for GR leads to unwanted side effects in some patients. Optimization of the previously described fused azadecalin series of selective GR antagonists led to the identification of CORT125134, which is currently being evaluated in a Phase 2 clinical study in patients with Cushing's syndrome.