ArticlePDF Available

Androgen Receptor Modulation Optimized for Response (ARMOR) Phase I and II Studies: Galeterone for the Treatment of Castration-Resistant Prostate Cancer

Authors:

Abstract and Figures

Purpose: Galeterone is a selective, multitargeted agent that inhibits CYP17, antagonizes the androgen receptor (AR), and reduces AR expression in prostate cancer cells by causing an increase in AR protein degradation. These open-label phase I and II studies (Androgen Receptor Modulation Optimized for Response-1 [ARMOR1] and ARMOR2 part 1) evaluated the efficacy and safety of galeterone in patients with treatment-naive nonmetastatic or metastatic castration-resistant prostate cancer (CRPC) and established a dose for further study. Experimental design: In ARMOR1, 49 patients received increasing doses (650-2,600 mg) of galeterone in capsule formulation; 28 patients in ARMOR2 part 1 received increasing doses (1,700-3,400 mg) of galeterone in tablet formulation for 12 weeks. Patients were evaluated biweekly for safety and efficacy, and pharmacokinetic parameters were assessed. Results: In ARMOR1, across all doses, 49.0% (24/49) achieved a ≥30% decline in prostate-specific antigen (PSA; PSA30) and 22.4% (11/49) demonstrated a ≥50% PSA decline (PSA50). In ARMOR2 part 1, across all doses, PSA30 was 64.0% (16/25) and PSA50 was 48.0% (12/25). In the 2,550-mg dose cohort, PSA30 was 72.7% (8/11) and PSA50 was 54.5% (6/11). Galeterone was well tolerated; the most common adverse events were fatigue, increased liver enzymes, gastrointestinal events, and pruritus. Most were mild or moderate in severity and required no action. Conclusion: The efficacy and safety from ARMOR1 and ARMOR2 part 1 and the pharmacokinetic results support the galeterone tablet dose of 2,550 mg/d for further study. Galeterone was well tolerated and demonstrated pharmacodynamic changes consistent with its selective, multifunctional AR signaling inhibition.
Content may be subject to copyright.
Cancer Therapy: Clinical
Androgen Receptor Modulation Optimized for
Response (ARMOR) Phase I and II Studies:
Galeterone for the Treatment of Castration-
Resistant Prostate Cancer
Bruce Montgomery
1
, Mario A. Eisenberger
2
, Matthew B. Rettig
3
, Franklin Chu
4
,
Roberto Pili
5
, Joseph J. Stephenson
6
, Nicholas J. Vogelzang
7
, Alan J. Koletsky
8
,
Luke T. Nordquist
9
, William J. Edeneld
10
, Khalid Mamlouk
11
, Karen J. Ferrante
11
, and
Mary-Ellen Taplin
12
Abstract
Purpose: Galeterone is a selective, multitargeted agent that
inhibits CYP17, antagonizes the androgen receptor (AR), and
reduces AR expression in prostate cancer cells by causing an
increase in AR protein degradation. These open-label phase I
and II studies [Androgen Receptor Modulation Optimized
for Response-1 (ARMOR1) and ARMOR2 part 1] evaluated
the efcacy and safety of galeterone in patients with treat-
ment-naive nonmetastatic or metastatic castration-resistant
prostate cancer (CRPC) and established a dose for further
study.
Experimental Design: In ARMOR1, 49 patients received
increasing doses (6502,600 mg) of galeterone in capsule
formulation; 28 patients in ARMOR2 part 1 received increasing
doses (1,7003,400 mg) of galeterone in tablet formulation for
12 weeks. Patients were evaluated biweekly for safety and
efcacy, and pharmacokinetic parameters were assessed.
Results: In ARMOR1,across all doses, 49.0% (24/49) achieveda
30% decline in prostate-specic antigen (PSA; PSA30) and
22.4% (11/49) demonstrated a 50% PSA decline (PSA50). In
ARMOR2 part 1, across all doses, PSA30 was 64.0% (16/25) and
PSA50 was 48.0% (12/25). In the 2,550-mg dose cohort, PSA30
was 72.7% (8/11) and PSA50 was 54.5% (6/11). Galeterone was
well tolerated; the most common adverse events were fatigue,
increased liver enzymes, gastrointestinalevents, and pruritus. Most
were mild or moderate in severity and required no action and there
were no apparent mineralocorticoid excess (AME) events.
Conclusions: The efcacy and safety from ARMOR1 and
ARMOR2 part 1 and the pharmacokinetic results support the
galeterone tablet dose of 2,550 mg/d for further study. Galeterone
was well tolerated and demonstrated pharmacodynamic changes
consistent with its selective, multifunctional AR signaling inhibi-
tion. Clin Cancer Res; 22(6); 135663. 2015 AACR.
Introduction
Despite recent advances in the treatment of castration-resistant
prostate cancer (CRPC), prostate cancer remains the second most
common cancer-related mortality in men in the United States (1).
The development of a new generation of therapies targeting the
androgen axis has been based on an expanded understanding of
the molecular mechanisms of CRPC. It is now understood that in
the clinical setting of castrate levels of serum testosterone, prostate
tumors adapt by upregulating tissue androgens and androgen
receptors (AR) to maintain proliferation. Tumor androgen levels
remain sufciently elevated to stimulate ARs as a result of tumor
conversion of circulating adrenal androgens and de novo androgen
synthesis (25). In addition, prostate cancer adapts to androgen-
deprivation therapy by AR gene amplication, upregulation of AR
transcripts, or protein expression (6, 7). Thus, inhibition of the
synthesis of nongonadal androgens and blockade of AR remain
key targets in CRPC therapy.
Abiraterone and enzalutamide have improved outcomes for
patients with metastatic CRPC (mCRPC). Although abiraterone
and enzalutamide have been shown to improve overall survival
(OS), these agents are not curative and not without safety and
tolerability issues (811). In addition, a signicant proportion of
patients do not respond; and in those who do respond, therapy
will eventually fail because of the development of resistance
(9, 10, 1214). A major component of resistance to second-
generation AR-targeting agents may be mediated by AR splice
1
University of Washington,Seattle, Washington.
2
Sidney Kimmel Com-
prehensive Cancer Center at Johns Hopkins University, James Bucha-
nan Brady Urological Institute, Baltimore, Maryland.
3
UCLA Jonsson
Comprehensive Cancer Center, Los Angeles, California.
4
San Berna-
dino Urological Associates, San Bernadino, California.
5
Indiana
University School of Medicine, Indianapolis, Indiana.
6
Institute for
Translational Oncology Research, Greenville, South Carolina.
7
Com-
prehensive Cancer Centers of Nevada and U.S. Oncology Research,
Las Vegas, Nevada.
8
Lynn Cancer Institute, Boca Raton, Florida.
9
Urol-
ogy Cancer Center and GU Research Network, Omaha, Nebraska.
10
Greenville Hospital System and University Medical Center, Green-
ville, South Carolina.
11
Tokai Pharmaceuticals, Cambridge, Massachu-
setts.
12
Dana-Farber Cancer Institute, Boston, Massachusetts.
Note: Supplementary data for this article are available at Clinical Cancer
Research Online (http://clincancerres.aacrjournals.org/).
Corresponding Author: Mary-Ellen Taplin, Dana-Farber Cancer Institute, 450
Brookline Avenue, Boston, MA 02215. Phone: 617-632-3237; Fax: 617-632-2165;
E-mail: Mary_Taplin@dfci.harvard.edu
doi: 10.1158/1078-0432.CCR-15-1432
2015 American Association for Cancer Research.
Clinical
Cancer
Research
Clin Cancer Res; 22(6) March 15, 2016
1356
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
variants, such as AR-V7, which are produced in tumor cells as a
result of aberrant RNA splicing of the wild-type AR transcript. The
resultant truncated AR protein lacks the C-terminal domain to
which androgen binds and is the primary site of action of
nonsteroidal antiandrogens such as enzalutamide. Furthermore,
splice variants have been shown to be constitutively active tran-
scription factors, leading to the activation of androgen-responsive
genes even at castrate levels of androgens (15, 16). Mutations in
the AR may also contribute to resistance in CRPC, and AR point
mutations allow activation of the receptor by nonphysiologic
ligands (e.g., cortisol, progesterone, utamide, bicalutamide;
refs. 17, 18, 19). As a result, androgen-independent, but
AR-dependent, tumor growth occurs, and tumors become resis-
tant to therapeutic agents that alter androgen production (e.g.,
abiraterone) or antagonize binding to the AR (e.g., bicalutamide,
enzalutamide). Recent data demonstrated that patients with
detectable circulating tumor cells harboring AR-V7 had inferior
responses to abiraterone or enzalutamide, including inferior
prostate-specic antigen (PSA) response, clinical and radiograph-
ic progression-free survival (PFS), and poor OS (12, 13).
Galeterone is a selective, multitargeted agent that disrupts
androgen signaling at multiple points in the pathway. Preclinical
data have shown that galeterone is a selective potent CYP17
inhibitor and a potent AR antagonist, but unlike other available
agents that target androgen signaling, galeterone reduces AR
expression in prostate cancer cells by causing an increase in AR
protein degradation (2026). Preclinical in vitro and in vivo data
have shown that galeterone treatment in prostate cancer models
resulted in a signicant reduction in both full-length AR and AR-
V7 splice variant levels. In addition, galeterone has been shown to
have activity against AR point mutations T878A (20-25) and, in
preliminary ndings, to have activity in cells expressing the AR
point mutation F876L (27).
This article reports the safety and efcacy of galeterone in a
phase I study, Androgen Receptor Modulation Optimized for
Response (ARMOR1), and the dose-escalation component of the
phase II ARMOR2 study (ARMOR2 part 1). The dose-escalation
component of ARMOR2 was conducted to determine the phase II
and phase III dose of a galeterone spray dry dispersion (SDD)
tablet. This formulation was developed after a healthy volunteer
study conrmed a signicant food effect with the capsule formu-
lation that was used in ARMOR1 (Supplementary Data). The SDD
tablet formulation was shown in a healthy volunteer study to not
be affected by food, providing similar exposure (area under the
concentration-time curve, AUC) in fed and fasted states (28).
Results of this study also demonstrated equivalent serum con-
centrations using either 1,700 mg of the SDD tablet or 2,600 mg of
the capsule, which was the highest dose studied in ARMOR1.
Thus, the dose-escalation portion of ARMOR2 was conducted to
evaluate the safety and tolerability of escalating doses of the SDD
formulation and to determine the recommended dose for
ARMOR2 part 2 and ARMOR3.
Patients and Methods
Patients
Eligible men had histologically conrmed nonmetastatic (M0)
or metastatic (M1) adenocarcinoma of the prostate, a life expec-
tancy of >12 weeks, and progressive disease despite ongoing
androgen-deprivation therapy. Patients were required to have
progressive disease according to Prostate Cancer Clinical Trials
Working Group 1 [PCWG1] criteria (29) in ARMOR1, or PCWG2
criteria (30) in ARMOR2 part 1, ongoing treatment with gonad-
otropin-releasing hormone analogs or orchiectomy (serum tes-
tosterone <50 mg/dL), and an Eastern Cooperative Oncology
Group (ECOG) performance status of 1. ARMOR1 excluded
patients who had previously received chemotherapy, ketocona-
zole, abiraterone, or enzalutamide. ARMOR2 part 1 permitted the
enrollment of abiraterone-refractory patients, provided it had
been discontinued 4 weeks before enrollment and that the
duration of therapy was 6 months before PSA progression or
>6 weeks with documentation of an initial response followed by
PSA progression. Previous ketoconazole treatment was permitted
upon agreement between the investigator and the study sponsor.
Patients with nonhepatic visceral metastases and/or tumor-asso-
ciated bone pain that required active pain management were
excluded from ARMOR1. Patients with indeterminate lung
nodules were eligible. Other exclusion criteria included any
previous radium-223, strontium, or samarium therapy within 8
weeks of enrollment; radiotherapy 4 weeks before enrollment or
completed radiotherapy in ARMOR1; or radiotherapy 3 weeks
(2 weeks for single-fraction radiotherapy) in ARMOR2 part 1.
Patients were excluded if they had previous treatment with inves-
tigational drugs or agents that could have interfered with the
efcacy and safety assessments. Patients with abnormal labora-
tory test results, including serum creatinine level >1.5 times the
upper limit of normal (ULN), liver function test results >1.5
ULN, hemoglobin level 9.0 g/dL, platelet count 100 10
9
/L,
absolute neutrophil count 1.5 10
9
/L, and serum potassium
level <3.5 mmol/L, were ineligible, as were those with serious
concurrent illnesses or conditions, including heart failure, uncon-
trolled hypertension, angina, active autoimmune disease, or
gastrointestinal disorders or gastric bypass surgery that could have
interfered with study medication absorption. Written informed
consent was obtained from participants before enrollment.
Study design
ARMOR1 (NCT00959959) was a phase I, multicenter, open-
label, dose-escalation study conducted in collaboration with the
Department of Defense Prostate Cancer Clinical Trials Consor-
tium, designed to assess the tolerability, safety, and efcacy of oral
Translational Relevance
Despite the recent advances in the understanding and
treatment of metastatic castration-resistant prostate cancer
(mCRPC), it remains a lethal disease. Androgen receptor (AR)
signaling remains a primary target of therapy, as the under-
standing of both the disease and mechanisms of resistance
expand. Galeterone, a selective, multitargeted agent, is distinct
from other mCRPC therapies in that it combines the mechan-
isms of current agentsCYP17 inhibition and AR antago-
nismwith the novel mechanism of increasing AR protein
degradation. These rst assessments of galeterone in mCRPC
identied a well-tolerated dose that resulted in clinically
signicant reductions in prostate-specic antigen, and dem-
onstrate the potential of this agent. In vitro data and results of
these studies have informed future investigation of galeterone,
which will include AR-related biomarker analyses.
Galeterone for Metastatic Castration-Resistant Prostate Cancer
www.aacrjournals.org Clin Cancer Res; 22(6) March 15, 2016 1357
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
galeterone for chemotherapy-naive patients with CRPC. The
primary goals were to nd the optimal dose of galeterone with
an acceptable safety prole, dened as an observed dose-lim-
iting toxicity (DLT) rate of 35%, and to identify a dose for
further phase II study. The dose equivalence component of
ARMOR2 (i.e., part 1; NCT01709734) evaluated the pharma-
cokinetics (PK), safety, and efcacy of a new formulation of
galeterone with improved bioavailability. A micronized pow-
der formulation (capsule) was used in ARMOR1 and an SDD
formulation was used in ARMOR2 part 1. These studies were
designed and monitored in accordance with Sponsor proce-
dures, which comply with the ethical principles of Good
Clinical Practice, as required by the major regulatory author-
ities, and in accordance with the Declaration of Helsinki and
the FDA regulations. The protocols were approved by the
institutional review board of each study site.
In ARMOR1, galeterone capsules (micronized powder, 325
mg) were administered orally as (i) 650 mg in the evening, (ii)
975 mg in the evening, (iii) 975 mg in the morning, (iv) 1,300 mg
in the evening, (v) 1,950 mg in the evening, (vi) 1,950 mg divided
into morning and evening doses, (vii) 2,600 mg in the evening, or
(viii) 2,600 mg divided into morning and evening doses, accord-
ing to the cohort they entered. All doses were administered with a
patient-selected meal, except for the 975 mg morning dose cohort,
which received a high-fat, high-calorie nutritional supplement
(Novasource Renal, Nestle HealthCare Nutrition, Florham Park,
NJ) in place of the meal. Enrollment target was 6 patients per dose
cohort. If an acceptable safety prole was determined by the
internal monitoring committee (IMC; DLT rate 35% or 2of
6 patients in cohorts of 6 patients), subsequent dose levels and
schedules were opened for enrollment. If 3 of 6 patients expe-
rienced DLTs, dose de-escalation was required. DLTs were dened
as any study drugrelated grade 3 or higher adverse event [AE;
National Cancer Institute Common Terminology Criteria for
Adverse Events (CTCAE) version 4.0] considered to be possibly,
probably, or denitely related to the study drug.
In ARMOR2 part 1, galeterone SDD tablets (425 mg) were
administered at doses of 1,700, 2,550, and 3,400 mg once daily
with the morning meal. Enrollment target was 6 patients per dose
level. Dose escalation occurred when no clinically signicant
grade 2 or greater sustained AEs or serious, unexpected grade 3
or higher AEs occurred in a dose group 2 weeks after the last
patient in that cohort received his rst dose.
The planned treatment duration of both studies was 12 weeks,
with optional extension dosing for eligible patients based on
safety and tolerability during the 12-week phase. Extension dos-
ing was continued until the patient withdrew, experienced unac-
ceptable toxicity, the disease progressed, or the patient died.
Assessments
Safety assessments, conducted at baseline and every 2 weeks
during the 12-week study and every 4 weeks during the optional
extension phase, included physical examination, vital signs, elec-
trocardiogram (ECG), serum chemistry, hematology, urinalysis,
and performance status. AEs that occurred during the study and up
to 30 days after the last dose of study drug were collected, coded
according to Medical Dictionary of Regulatory Activities, version
12.1, and graded using CTCAE version 4.0. PSA was determined at
each study visit.
In the rst 4 dosing cohorts of ARMOR1, blood samples for PK
analysis were obtained predose and at 4 hours on day 1. In the
remaining cohorts, blood samples were obtained before (hour
0) and 1, 2, 4, and 6 hours after the rst dose on day 1. At all
remaining visits, if the regimen for the cohort included a
morning dose, blood samples were obtained at 6 hours after
their dose; for all other cohorts, blood samples were obtained
at any time during the visit. In ARMOR2 part 1, blood samples
forPKanalyseswereobtainedbefore(hour0)and2,3,4,5,
and 6 hours after the day 1 dose, and predose on days 7, 14, 21,
28, and 84. Additional samples were obtained in consenting
patients on day 1 at 8, 12, 16, and 24 hours postdose and on
day 84 at 2, 3, 4, 5, 6, 8, 12, 16, and/or 24 hours postdose.
Blood samples were also obtained at each study visit of
ARMOR2 part 1 for determination of pregnenolone, 17-hydro-
xyprogesterone, deoxycorticosterone, 11-deoxycortisol, cortico-
sterone, cortisol, dehydroepiandrosterone sulfate (DHEAS),
androstenedione, and testosterone concentrations.
Data Analysis
Efcacy endpoints included the proportion of responders [PSA
decrease 50% [PSA50] and 30% (PSA30)], maximal decrease
in PSA from baseline to 12 weeks or PSA nadir, changes from
baseline in tumor response as assessed by bone scan and CT or
MRI using PCWG2 and RECIST v1.1. PSA efcacy was based on
the intent-to-treat population (ITT), dened as enrolled patients
who received at least 1 dose of study drug. Response was based on
measurable disease in both studies. Time to progression, PFS
dened as the time from rst dose of study drug until objective
CRPC progression or death, whichever occurred rst, and OS were
the endpoints assessed in the ARMOR1 extension phase. Descrip-
tive statistics were used for most variables (n, mean, SD, median,
minimum, and maximum for continuous variables and frequency
and percentage for categorical variables).
Results
Patients
Baseline patient and disease characteristics are presented
in Table 1. In ARMOR1, 49 patients were enrolled in 8 cohorts,
with 6 patients in each, except cohort 4, which enrolled 7 patients.
Twelve patients discontinued the study before completion of 12
weeks because of treatment-emergent AEs [TEAEs; n¼5; nausea,
chronic obstructive pulmonary disease exacerbation (event onset
before dosing), elevated aspartate aminotransferase/alanine ami-
notransferase levels (AST/ALT; n¼2), acute renal failure
[reversible after resolution of rhabdomyolysis, which occurred
while the patient was receiving simvastatin therapy and became
evident after the patient fell], disease progression (n¼5), or
withdrawal of consent/personal choice [n¼2; Table 2)]. Twenty-
two of the 37 patients who completed the study were eligible for
the optional extension phase, and 21 patients were dosed. Over-
all, all patients received 650 to 2,600 mg galeterone daily for <1to
20 months. In ARMOR2 part 1, 28 patients were enrolled in 3
dosing cohorts, with 6 patients in the 1,700-mg cohort, 14 in the
2,550-mg cohort (abiraterone-resistant, n¼3), and 8 in the
3,400-mg cohort. Six patients discontinued the study before 12
weeks because of TEAEs [n¼4; angioedema (in an African-
American who was receiving the angiotensin-converting enzyme
inhibitor, lisinopril), rash, weakness, and tremulousness] or dis-
ease progression (n¼2). All 3 patients with abiraterone-resistant
disease completed the 12-week phase of the study. Nineteen of 22
patients who completed the study participated in the optional
Montgomery et al.
Clin Cancer Res; 22(6) March 15, 2016 Clinical Cancer Research1358
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
extension phase; 2 of the patients with abiraterone-resistant
disease were not eligible for the extension because of disease
progression (Table 2). Overall duration of therapy ranged from <1
month to 14 months.
Safety and tolerability
ARMOR1. Safety reviews were completed after all patients were
dosed in each cohort and the IMC recommended continued
escalation following review of all doses. There were 2 deaths, 1
from disease progression and 1 from acute septic shock followed
by acute metabolic acidosis and renal failure, which was not
related to galeterone. All patients experienced at least 1 TEAE
during the 12-week phase, with most being mild or moderate in
severity (91.5%) and comparable among cohorts. The majority
(73%) of the AEs required no action. The most common
TEAEs were fatigue [17 patients (34.7%)], increased AST level
[16 patients (32.7%)], increased ALT level [15 patients (30.6%)],
nausea [12 patients (24.5%)], diarrhea [11 patients (22.4%)], and
pruritus [11 patients (22.4%); Table 3]. The most common
treatment-related TEAEs were increased AST level [7 patients
(14.3%)], nausea [5 patients (10.2%)], increased bilirubin level
[4 patients (8.2%)], fatigue [4 patients (8.2%)], and diarrhea
[3 patients (6.1%)]. The majority of patients (85.7%) in the
extension phase experienced mild or moderate TEAEs that were
consistent with those reported during the treatment phase.
ARMOR2 Part 1. Galeterone tablets were well tolerated at all
doses, as assessed by the IMC. Safety reviews were completed after
all patients were dosed in each cohort, and the IMC recommended
continued escalation. Most patients (93%) experienced at least 1
TEAE, with the majority (91%) being grade 1 or 2 in severity and
comparable among cohorts. Most (72%) AEs required no inter-
vention. There were no DLTs at any dose level. The most common
TEAEs were nausea [13 patients (46.4%)], fatigue [9 patients
(32.1%)], pruritus [9 patients (32.1%)], vomiting [8 patients
(28.6%)], and decreased appetite [6 patients (21.4%); Table 3].
The most common treatment-related TEAEs were nausea
[10 patients (35.7%)]; pruritus [9 patients (32.1%)]; fatigue,
vomiting, and decreased appetite [6 patients (21.4%] for each);
and constipation, diarrhea, increased ALT level, and dizziness
[3 patients (10.7%)] for each). Although edema and hypokalemia
were observed, they were independent events in different patients
and no combined apparent mineralocorticoid excess events were
seen (Table 4).
Pharmacokinetics
The PK analysis plan of ARMOR1 was not designed to fully
characterize the PK of galeterone. There was no consistency or
dose dependence with respect to plasma concentrations and
regimen. There was little or no difference in mean concentrations
in the single daily doses, with only the 650-mg dose demonstrat-
ing lower mean concentrations, and the PK of the 975-mg dose
was no different after the supplement, compared with a patient-
selected meal. Dividing the dose did not have a signicant effect
on exposure (AUC).
The PK analysis plan of ARMOR2 was not designed to fully
characterize the PK of galeterone. The ARMOR2 part 1 PK
parameters after single doses of 1,700, 2,550, and 3,400 mg
of the SDD tablet formulation were similar among doses.
Exposure, expressed as AUC from predose to 6 hours postdose
(AUC
06
), was 2,646 1,748 h ng/mL, 2,684 2,043 h ng/mL,
Table 2. Treatment cohorts and patient disposition
ARMOR1Galeterone capsules ARMOR2 Part 1Galeterone SDD tablets
(N¼49) (N¼28)
Dosing cohort
Enrolled,
n
Completed
12-week
study, n
Entered
extension
phase, nCohort
Enrolled,
n
Completed
12-week
study, n
Entered
extension
phase, n
650 mg with meal 6 3 3 1,700 mg 6 6 6
975 mg with meal 6 5 2 2,550 mg 14 11 9
1,300 mg with meal 6 5 3 3,400 mg 8 5 4
1,950 mg with meal 7 5 2
975 mg with supplement
a
64 4
1,950 mg divided doses with meal 6 5 2
b
2,600 mg with meal 6 5 2
2,600 mg divided doses with meal 6 5 3
a
Novasource Renal, Nestle HealthCare Nutrition, Florham Park, New Jersey.
b
Three patients were eligible for the extension phase; however, only 2 patients were dosed with galeterone.
Table 1. Baseline characteristics
Characteristic
ARMOR1 ARMOR2 Part 1
(N¼49) (N¼28)
Age, median (range), y 68 (4789) 70 (4890)
Ethnicity, n(%)
White 43 (87.8) 24 (85.7)
African-American or black 3 (6.1) 2 (7.1)
Asian 1 (2.0) 1 (3.6)
Other 2 (4.1) 1 (3.6)
Metastatic disease (M1), n(%) 25 (51.0) 24 (85.7)
Bone, n25 24
Nodal, n15 10
Bone and nodal, n98
Visceral (liver and/or lung), n71
Visceral and bone, n61
Visceral and nodal, n30
Soft tissue (not nodal, liver, or lung), n17 11
Previous therapies, n(%)
Medical and/or surgical castration 49 (100) 28 (100)
Immunotherapy 1 (2) 2 (7.1)
Radiotherapy 27 (55) 16 (57.1)
Surgery 24 (49) 12 (42.9)
Abiraterone NA 3 (10.7)
Enzalutamide NA 0
ECOG, n(%)
0 45 (91.8) 22 (78.6)
1 4 (8.2) 5 (17.9)
Missing 0 1 (3.6)
Gleason score, median (range)
a
7(610) 8 (610)
PSA, median (range), ng/dL 24 (6200.6) 17.6 (3.36,760)
Abbreviations: NA, not applicable.
a
Data were missing in 2 patients in ARMOR1 and 1 patient in ARMOR 2 Part 1.
Galeterone for Metastatic Castration-Resistant Prostate Cancer
www.aacrjournals.org Clin Cancer Res; 22(6) March 15, 2016 1359
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
and 2,528 1,529 h ng/mL for the 1,700, 2,550, and 3,400 mg
doses, respectively.
Efcacy endpoints
ARMOR1. The ITT population for PSA efcacy included 49
patients. Across all doses tested, 24 of 49 (49.0%) achieved a
PSA30 and 11 of 49 patients (22.4%) demonstrated PSA50
(Fig. 1A). During the study, one patient in the 650 mg/d group
discontinued his gonadotropin-releasing hormone analog and
one patient in the 975 mg/d group underwent transurethral
resection of the prostate. Excluding these patients, across groups
the PSA30 was 51.1% (24/47) and the PSA50 was 23.4% (11/47).
An increase in response rate was observed with higher doses. At
the 2,600 mg dose, 9 of 12 patients (75.0%) demonstrated a
PSA30 and 5 of 12 patients (41.7%) demonstrated a PSA50. There
was no difference in PSA response between groups that had
divided dosing and groups that had once-daily dosing. Of the
evaluable patients [those with measurable target lesions at screen-
ing or baseline who had a follow-up scan at the 14-week (nal)
study visit; n¼17], 2 patients had a partial response (PR) and 10
patients had stable disease (SD), according to RECIST. In the
extension phase, disease progression ultimately occurred in 20 of
the 21 patients. No consistent trends were observed in time to
progression (range, 14592 days), PFS, or OS [shortest: 189 days,
cohort 3 (1,300 mg/d)] between treatment cohorts. Best overall
response assessed by RECIST was SD in 13 of 17 patients (76.5%)
in the extension phase; the remaining 4 patients had disease
progression.
ARMOR2 Part 1. The ITT population for PSA efcacy in treatment-
na
ve patients included 25 patients. Three patients had received
prior abiraterone treatment. Across the 3 doses in treatment-naive
patients, the decline in PSA from baseline in the ITT population
was 30% in 16 of 25 patients (64.0%) and 50% in 12 of 25
patients (48.0%). In the 2,550-mg dose cohort, 8 of 11 treatment-
naive patients (72.7%) had a 30% decline in PSA from baseline
and 6 of 11 patients (54.5%) had a 50% decline in PSA from
baseline. In the 1,700-mg dose cohort 50% (3/6 patients)
achieved a PSA30 and PSA50. In the 3,400 mg dose cohort,
62.5% (5/8 patients) achieved a PSA30 and 37.5% (3/8 patients)
achieved a PSA50 (Fig. 1B). One patient in the 2,550-mg/d group
had only 1 post-baseline PSA measurement (performed at
2 weeks) and 1 patient in the 3,400 mg/d group had no post-
baseline measurement of PSA. Excluding these patients, the
PSA30 and PSA50 were 80% and 60% in the 2,550 mg/d group,
and 71.4% and 42.9% in the 3,400 mg/d group. Of the 3 patients
treated with 2,550 mg/d who had prior treatment with abirater-
one, 1 patient (33%) achieved PSA30, 1 patient had a maximal
percent change of 2%, and 1 patient had an increase from
baseline. Of the 26 evaluable patients with measurable disease
at baseline, 20 (76.9%) patients had SD and 1 patient had PR at 12
weeks.
Steroidogenic pathway markers
Galeterone resulted in overall reductions in median serum
testosterone, DHEAS, and androstenedione concentrations.
Median corticosterone level was increased from a median baseline
of 204 ng/dL to 1,377.5 ng/dL at week 12, and cortisol and
deoxycorticosterone levels were generally unchanged (Table 5).
Discussion
Results of ARMOR1 and ARMOR2 part 1 demonstrated that
galeterone, an agent that previous studies have shown inhibits
androgen production, blocks the ligand-binding domain of AR,
and suppresses AR levels in vitro, is safe and shows promising PSA
Table 3. Treatment-emergent AEs occurring in >10% of patients in ARMOR1 or ARMOR2 Part 1
ARMOR1 ARMOR2 Part 1
(N¼49) (N¼28)
AE
Grade 1 or
2, n(%)
Grade 3 or
higher, n(%)
Grade 1 or
2, n(%)
Grade 3 or
higher, n(%)
Abdominal pain 5 (10.2) 0 1 (3.6) 0
Increased alkaline phosphatase level 7 (14.3) 0 0 0
Increased ALT level 7 (14.3) 8 (16.3) 1 (3.6) 3 (10.7)
Decreased appetite 6 (12.2) 0 6 (21.4) 0
Arthralgia 6 (12.2) 0 1 (3.6) 0
Increased AST level 13 (26.5) 3 (6.1) 1 (3.6) 1 (3.6)
Back pain 1 (2.0) 0 3 (10.7) 0
Increased bilirubin level 6 (12.2) 1 (2.0) 0 0
Constipation 5 (10.2) 0 3 (10.7) 1 (3.6)
Cough 7 (14.3) 0 3 (10.7) 0
Diarrhea 11 (22.4) 0 4 (14.3) 1 (3.6)
Dizziness 3 (6.1) 0 3 (10.7) 0
Fall 0 0 3 (10.7) 0
Fatigue 16 (32.7) 1 (2.0) 9 (32.1) 0
Nausea 12 (24.5) 0 13 (46.4) 0
Pruritus 11 (22.4) 0 9 (32.1) 0
Rash 5 (10.2) 0 0 1 (3.6)
Urinary tract infection 4 (8.2) 0 4 (14.3) 0
Vomiting 6 (12.2) 0 8 (28.6) 0
Decreased weight 5 (10.2) 0 4 (14.3) 0
Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase.
Table 4. Summary of Potential AME AEs in ARMOR1 or ARMOR2 Part 1
Number of
incidences AE
Attribution:
related/unrelated
a
1 Grade 2 hypokalemia 1/0
3 Grade 3 hypokalemia 1/2
1 Grade 1 peripheral edema 0/1
3 Grade 2 peripheral edema 2/1
a
All events were individual occurrences and not considered AME symptoms.
Montgomery et al.
Clin Cancer Res; 22(6) March 15, 2016 Clinical Cancer Research1360
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
responses in patients with mCRPC. Results from phase I healthy
volunteer PK studies and the PK results of ARMOR2 part 1 support
a 2,550 mg/d dose of galeterone SDD tablet for use in future trials.
All doses tested had similar safety and tolerability proles.
Results of these studies demonstrate that galeterone is well tol-
erated in men with CRPC, with infrequent grade 3 and 4 toxicities.
The most common treatment-related AEs were nausea, vomiting,
fatigue, pruritus, and decreased appetite. Of these events, the vast
majority (90%) were grade 1 or 2 and did not require any
intervention. Of note, there were no apparent mineralocorticoid
excess AEs, supporting results of preclinical studies demonstrating
the specicity of galeterone for CYP17 lyase compared with
hydroxylase (19). This hypothesis is further supported by the
steroidogenic marker results showing no change in deoxycorti-
costerone or cortisol and a small increase in corticosterone,
relative to a large increase observed with abiraterone even in the
absence of coadministration of steroids with galeterone (31). The
reductions in testosterone are slightly less than those seen at full
dose abiraterone, but similar to that found in the dose escalation
study (31).
Signicant PSA declines were observed with all dose levels.
Patients in ARMOR1 had an overall PSA30 and PSA50 of 49% and
22%, respectively, with the highest dose (2,600 mg) showing
PSA30 and PSA50 of 75% and 42%, respectively. In ARMOR2 part
1, 2,550 mg of the SDD tablet formulation, the dose found to
-100
-75
-50
-25
0
25
50
75
100
Maximal PSA change from baseline (%)
1,950 mg/d
(n = 13)
2,600 mg/d
(n = 12)
1,300 mg/d
(n = 6)
975 mg/d
(n = 12)
650 mg/d
(n = 6)
*
-100
-75
-50
-25
0
25
50
75
100
Maximal PSA change from baseline (%)
**
1,700 mg/d
(n = 6)
2,550 mg/d
(n = 11)
3,400 mg/d
(n = 7)
Figure 1.
A, the maximal percentage of change
in PSA from baseline at 12 weeks by
total daily dose in treatment-naive
patients in ARMOR1 (n¼49).
Patterned data points reect 1 patient
who discontinued his gonadotropin-
releasing hormone analog (650 mg/d
group) and 1 patient who underwent
transurethral resection of the prostate
(975-mg/d group). B, the maximal
percentage of change in PSA from
baseline by total daily dose in
evaluable treatment-naive patients in
ARMOR2 Part 1 (n¼25). Patterned
data point reects a patient who only
had 1 post-baseline PSA measurement
(at 2 weeks). One patient in the
3,400 mg/d group (n¼8) is not
included in the graph because no post-
baseline PSA measurements were
completed. Abiraterone-refractory
patients (N¼3) were not included in
this analysis. Reference lines: green,
50%; orange, 30%. , maximal PSA
values >100% increase from baseline.
Table 5. Median (range) concentrations of the steroidogenic pathway markers
in ARMOR2 Part 1
Median (range)
Steroid Baseline Week 12
Testosterone, ng/dL 7.5 (322) 2 (<114)
Androstenedione, ng/dL 32 (781) 14 (<534)
DHEAS, mg/dL 37.5 (<15220) 18 (<15105)
Corticosterone, ng/dL 204 (<20874) 1,377.5 (974,375)
Deoxycorticosterone, ng/dL <16 (<1618) <16 (<1689)
Cortisol, mg/dL 14.7 (1.828.7) 18.1 (4.135)
Galeterone for Metastatic Castration-Resistant Prostate Cancer
www.aacrjournals.org Clin Cancer Res; 22(6) March 15, 2016 1361
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
provide exposure similar to that of 2,600 mg of the capsule,
resulted in greater PSA30 and PSA50 of 80% and 60%, respec-
tively. These results are comparable with those observed in phase I
and II trials of abiraterone and enzalutamide (8, 11, 31). Of
note, these results were marginally better than the 3,400 mg
(PSA30 ¼71%, PSA50 ¼43%) and 1,700 mg (PSA30 ¼50%,
PSA50 ¼50%) doses.
Although ARMOR1 showed that increasing the dose resulted in
a better PSA response, a phase I healthy volunteer PK study
showed that the capsule formulation was confounded by a food
effect and resulted in exposure that plateaued above 1,950 mg
(Appendix; ref. 28). The lack of a clear food effect in ARMOR1
could be attributed to the study design, in that the blood sampling
strategy was not optimal for assessment of PK parameters, and
patient-selected meals precluded assessment of the effect of fat
and calories.
ARMOR2 part 1 served as a bridging study between the original
capsule formulation and the SDD tablet formulation, which was
developed to have improved relative bioavailability over the
capsule. In PK studies in healthy volunteers, the SDD tablet was
shown to result in dose-related increases in exposure that were
similar in fed and fasted states that plateaued at doses above
2,550 mg (32). In addition, it was found that the exposure after
1,700 mg of the SDD tablet was similar to that with 2,600 mg of
the original capsule formulationthe dose in ARMOR1 that
resulted in the best efcacy numbers (28). ARMOR2 part 1
evaluated increasing doses of the SDD tablet formulation starting
at the 1,700 mg dose. The PK results of this study showed that
there was no increase in exposure with higher doses. Although the
lack of increase in exposure between the 1,700 and the 2,550 mg
dose was not consistent with earlier PK evaluations of the SDD
tablet, it could again be attributed to study design, in that the
sampling strategy was not optimal for a full PK assessment. The
results from the PK, safety and PSA decline data support the choice
of the 2,550 mg dose for use in phase II and III clinical studies. The
phase II studies have been completed and are in follow-up, and
the phase III study is planned (ARMOR3-SV). The ability of
galeterone to target splice variant AR through enhanced degrada-
tion suggests that it may have potential activity in tumors expres-
sing these resistant variants. The phase III, ARMOR3-SV study will
target splice variant (AR-V7) positive tumors and is based on PSA
responses seen patients with C-terminal loss in the treatment
na
ve cohort of ARMOR2 (33).
Conclusion
The efcacy and safety results from ARMOR1 and ARMOR2
part 1, and the PK results from phase I healthy volunteer studies
and ARMOR2 part 1 support the recommended dose of galeter-
one 2,550 mg daily taken with food for ARMOR2 part 2 and the
phase III study (ARMOR3-SV) using the SDD tablet formulation
with improved bioavailability. Galeterone is well tolerated in
CRPC patients and demonstrates pharmacodynamic changes
consistent with its selective multifunctional AR signaling inhibi-
tion. The analysis of galeterone is ongoing in expanded patient
cohorts in ARMOR2 part 2 and is ongoing for a phase III trial
(ARMOR3-SV) comparing galeterone with enzalutamide in treat-
ment-naive patients with mCRPC whose prostate tumors express
the AR-V7 splice variant.
Disclosure of Potential Conicts of Interest
L.T. Nordquist is a consultant/advisory board member for Bayer Pharma-
ceuticals. W.J. Edeneld reports receiving speakers bureau honoraria from
Astellas and Novartis. K.J. Ferrante holds ownership interest (including
patents) in, and is a consultant/advisory board member for Tokai Pharmaceu-
ticals. M.-E. Taplin reports receiving commercial research grants, other com-
mercial research support, speakers bureau honoraria from, and is a consultant/
advisory board member for Tokai Pharmaceuticals. No potential conicts of
interest were disclosed by the other authors.
Authors' Contributions
Conception and design: B. Montgomery, M.A. Eisenberger, M.B. Rettig,
N.J. Vogelzang, M.-E. Taplin
Development of methodology: B. Montgomery, K. Mamlouk, M.-E. Taplin
Acquisition of data (provided animals, acquired and managed patients,
provided facilities, etc.): B. Montgomery, M.B. Rettig, F. Chu, R. Pili,
J.J. Stephenson, N.J. Vogelzang, A.J. Koletsky, W.J. Edeneld, K. Mamlouk,
M.-E. Taplin
Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
computational analysis): B. Montgomery, M.A. Eisenberger, J.J. Stephenson,
N.J. Vogelzang, K. Mamlouk, M.-E. Taplin
Writing, review, and/or revision of the manuscript: B. Montgomery,
M.A. Eisenberger, M.B. Rettig, F. Chu, R. Pili, J.J. Stephenson, N.J. Vogelzang,
A.J. Koletsky, L.T. Nordquist, W.J. Edeneld, K. Mamlouk, K.J. Ferrante,
M.-E. Taplin
Administrative, technical, or material support (i.e., reporting or organizing
data, constructing databases): B. Montgomery, K. Mamlouk, M.-E. Taplin
Study supervision: B. Montgomery, J.J. Stephenson, A.J. Koletsky, W.J. Eden-
eld, K. Mamlouk
Other (study data review and clinical interpretation in preparation for
writing of the article): K.J. Ferrante
Grant Support
Data analysis support was provided by Sarah Hunter of Cd3. Technical
editorial and medical writing support was provided by Beth Kamp, PharmD.
Funding for this support was provided by Tokai Pharmaceuticals, Cambridge,
MA.
The costs of publication of this article were defrayed in part by the payment of
page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Received June 16, 2015; revised September 29, 2015; accepted October 11,
2015; published OnlineFirst November 2, 2015.
References
1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin
2014;64:929.
2. Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano
CS, et al. Maintenance of intratumoral androgens in metastatic prostate
cancer: a mechanism for castration-resistant tumor growth. Cancer Res
2008;68:444754.
3. Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, Wood CA, et al.
Androgen levels increase by intratumoral de novo steroidogenesis during
progression of castration-resistant prostate cancer. Cancer Res 2008;68:
640715.
4. Dillard PR, Lin MF, Khan SA. Androgen-independent prostate cancer cells
acquire the complete steroidogenic potential of synthesizing testosterone
from cholesterol. Mol Cell Endocrinol 2008;295:11520.
5. Chang KH, Li R, Papari-Zareei M, Watumull L, Zhao YD, Auchus RJ, et al.
Dihydrotestosterone synthesis bypasses testosterone to drive castration-
resistant prostate cancer. Proc Natl Acad Sci U S A 2011;108:1372833.
Montgomery et al.
Clin Cancer Res; 22(6) March 15, 2016 Clinical Cancer Research1362
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
6. Edwards J, Krishna NS, Grigor KM, Bartlett JM. Androgen receptor gene
amplication and protein expression in hormone refractory prostate
cancer. Br J Cancer 2003;89:5526.
7. Linja MJ, Porkka KP, Kang Z, Savinainen KJ, Janne OA, Tammela TL, et al.
Expression of androgen receptor coregulators in prostate cancer. Clin
Cancer Res 2004;10:103240.
8. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al.
Abiraterone in metastatic prostate cancer without previous chemotherapy.
N Engl J Med 2013;368:13848.
9. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al.
Abiraterone and increased survival in metastatic prostate cancer. N Engl J
Med 2011;364:19952005.
10. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al.
Increased survival with enzalutamide in prostate cancer after chemother-
apy. N Engl J Med 2012;367:118797.
11. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS,
et al. Enzalutamide in metastatic prostate cancer before chemotherapy.
N Engl J Med 2014;371:42433.
12. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al.
AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer.
N Engl J Med 2014;371:102838.
13. Efstathiou E, Titus M, Wen S, Hoang A, Karlou M, Ashe R, et al. Molecular
characterization of enzalutamide-treated bone metastatic castration-resis-
tant prostate cancer. Eur Urol 2015;67:5360.
14. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, et al. A clinically
relevantandrogen receptormutation confers resistance to second-generation
antiandrogens enzalutamide and ARN-509. Cancer Discov 2013;3:10209.
15. Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, et al. A novel androgen
receptor splice variant is up-regulated during prostate cancer progression
and promotes androgen depletion-resistant growth. Cancer Res 2009;69:
230513.
16. Mostaghel EA, Plymate SR, Montgomery B. Molecular pathways: targeting
resistance in the androgen receptor for therapeutic benet. Clin Cancer Res
2014;20:7918.
17. Brooke GN, Bevan CL. The role of androgen receptor mutations in prostate
cancer progression. Curr Genomics 2009;10:1825.
18. Taplin ME. Drug insight: role of the androgen receptor in the development
and progression of prostate cancer. Nat Clin Pract Oncol 2007;4:23644.
19. Carreira S, Romanel A, Goodall J, Grist E, Ferraldeschi R, et al. Tumor clone
dynamics in lethal prostate cancer. Science Translational Medicine 2014;
254:ra125
20. Jacoby D, Williams M. Differenential effects of galeterone, abiraterone,
orteronel, and ketoconazole on CYP17 and steroidogenesis. J Clin Oncol
2013. Abstr 184.
21. Handratta VD, Vasaitis TS, Njar VC, Gediya LK, Kataria R, Chopra P, et al.
Novel C-17-heteroaryl steroidal CYP17 inhibitors/antiandrogens: synthe-
sis, in vitro biological activity, pharmacokinetics, and antitumor activity in
the LAPC4 human prostate cancer xenograft model. J Med Chem 2005;48:
297284.
22. Vasaitis T, Belosay A, Schayowitz A, Khandelwal A, Chopra P, Gediya LK,
et al. Androgen receptor inactivation contributes to antitumor efcacy of
17a-hydroxylase/17,20-lyase inhibitor 3beta-hydroxy-17-(1H-benzimid-
azole-1-yl)androsta-5,16-diene in prostate cancer. Mol Cancer Ther
2008;7:234857.
23. Schayowitz A, Sabnis G, Njar VC, Brodie AM. Synergistic effect of a novel
antiandrogen, VN/124-1, and signal transduction inhibitors in prostate
cancer progression to hormone independence in vitro. Mol Cancer Ther
2008;7:12132.
24. Purushottamachar P, Godbole AM, Gediya LK, Martin MS, Vasaitis TS,
Kwegyir-Afful AK, et al. Systematic structure modications of multitarget
prostate cancer drug candidate galeterone to produce novel androgen
receptor down-regulating agents as an approach to treatment of advanced
prostate cancer. J Med Chem 2013;56:488098.
25. BrunoRD, Vasaitis TS, Gediya LK, Purush ottamachar P, Godbole AM, Ates-
Alagoz Z, et al. Synthesis and biological evaluations of putative metabol-
ically stable analogs of VN/124-1 (TOK-001): head to head anti-tumor
efcacy evaluation of VN/124-1 (TOK-001) and abiraterone in LAPC-4
human prostate cancer xenograft model. Steroids 2011;76:126879.
26. Kwegyir-Afful AK, Senthilmurugan R, Purushottamachar P, Ramamurthy
VP, Njar VCO. Galeterone and VNPT55 induce proteasomal degradation of
AR/AR-V7, induce signicant apoptosis via cytochrome c release and
suppress growth of castration-resistant prostate cancer xenografts in vivo.
Oncogarget 2015;6:2744027460.
27. AlNakouzi N, Wang C, Jacoby D, Gleave ME, Zoubeidi A. Galeterone
suppresses castration-resistant and enzalutamide-resistant prostate cancer
growth in vitro [abstract]. In: Proceedings of the AACR-NCI-EORTC Inter-
national Conference on Molecular Targets and Cancer Therapeutics; 2013
Oct 1923; Boston, Massachusetts: AACR; 2013. Abstract nr C89.
28. Kramer WG, Vince B, McGarry C. Comparison of the pharmacokinetics
(PK) of galeterone novel oral formulation. J Clin Oncol 2013;Supplement:
e16075.
29. Bubley GJ, Carducci M, Dahut W, Dawson N, Daliani D, Eisenberger M,
et al. Eligibility and response guidelines for phase II clinical trials in
androgen-independent prostate cancer: recommendations from the Pros-
tate-Specic Antigen Working Group. J Clin Oncol 1999;17:34617.
30. Scher HI, Morris MJ, Basch E, Heller G. End points and outcomes in
castration-resistant prostate cancer: from clinical trials to clinical practice.
J Clin Oncol 2011;29:3695704.
31. RyanCJ, Smith MR, Fong L, Rosenberg JE, Kantoff P, Raynaud F, et al. Phase
I clinical trial of the CYP17 inhibitor abiraterone acetate demonstrating
clinical activity in patients with castration-resistant prostate cancer who
received prior ketoconazole therapy. J Clin Oncol 2010;28:14818.
32. TOK-200-09 Clinical Study Report. Tokai Pharmaceuticals; 2013.
33. Taplin ME, Chi KN, Chu F, Cochran J, Edeneld WJ, Eisenberger M, et al.
Galeterone in 4 patient populations of men with CRPC: results from
ARMOR2 [abstract]. In: Proceedings of the European Society of Medical
Oncology Annual Meeting; 2014 Sep 2630, 2014; Madrid, Spain: ESMO;
2014. Abstract nr 757O.
www.aacrjournals.org Clin Cancer Res; 22(6) March 15, 2016 1363
Galeterone for Metastatic Castration-Resistant Prostate Cancer
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/22/6/1356/2034468/1356.pdf by guest on 09 June 2023
... We have recently developed galeterone (Gal), a potent drug that concurrently targets AR and Mnk1/2 [27]. Though Gal successfully advanced through phase 2-clinical trials in treating mCRPC, it faced setbacks in the pivotal phase 3 clinical trials due to the limited number of patients studied, and relatively higher clinical doses administered [28]. Consequently, we have synthesized a series of galeterone derivatives, the next generation galeterone analogs (NGGA), and evaluated their potential development to treat all stages of PCa focusing on enhanced efficacy, safety, and minimum effective dose (MED) [29]. ...
Article
Full-text available
Prostate cancer (PCa) relies in part on AR-signaling for disease development and progression. Earlier, we developed drug candidate galeterone, which advanced through phase 2-clinical trials in treating castration-resistant PCa (CRPC). Subsequently, we designed, synthesized, and evaluated next-generation galeterone-analogs including VNPP433-3β which is potently efficacious against pre-clinical models of PCa. This study describes the mechanism of action of VNPP433-3β that promotes degradation of full-length AR (fAR) and its splice variant AR-V7 besides depleting MNK1/2 in in vitro and in vivo CRPC models that stably overexpresses fAR. VNPP433-3β directly engages AR within the cell and promotes proteasomal degradation of fAR and its splice variant AR-V7 by enhancing the interaction of AR with E3 ligases MDM2/CHIP but disrupting AR-HSP90 binding. Next, VNPP433-3β decreases phosphorylation of 4EBP1 and abates binding of eIF4E and eIF4G to 5′ cap of mRNA by depleting MNK1/2 with consequent depletion of phosphorylated eIF4E. Finally, RNA-seq demonstrates modulation of multiple pathways that synergistically contribute to PCa inhibition. Therefore, VNPP433-3β exerts its antitumor effect by imposing 1) transcriptional regulation of AR and AR-responsive oncogenes 2) translational regulation by disrupting mRNA-5′cap-dependent translation initiation, 3) reducing AR half-life through enhanced proteasomal degradation in vitro and AR-overexpressing tumor xenografts in vivo.
... 204 A phase 1 study in patients with CRPC observed that~50% of men had a PSA decline after 12 weeks of treatment with galeterone, and no adrenal mineralocorticoid excess was noted. 205 Therefore, orteronel and galeterone are potentially attractive drugs for longer duration therapy and overcoming drug resistance, although a clinical study showed that orteronel did not meet the primary endpoint of overall survival. 206 Clinical studies of galeterone compared to enzalutamide in mCRPC expressing AR-V7 have been conducted, but the result do not meet the primary endpoint. ...
Article
Full-text available
Prostate cancer (PCa) affects millions of men globally. Due to advances in understanding genomic landscapes and biological functions, the treatment of PCa continues to improve. Recently, various new classes of agents, which include next-generation androgen receptor (AR) signaling inhibitors (abiraterone, enzalutamide, apalutamide, and darolutamide), bone-targeting agents (radium-223 chloride, zoledronic acid), and poly(ADP-ribose) polymerase (PARP) inhibitors (olaparib, rucaparib, and talazoparib) have been developed to treat PCa. Agents targeting other signaling pathways, including cyclin-dependent kinase (CDK)4/6, Ak strain transforming (AKT), wingless-type protein (WNT), and epigenetic marks, have successively entered clinical trials. Furthermore, prostate-specific membrane antigen (PSMA) targeting agents such as 177Lu-PSMA-617 are promising theranostics that could improve both diagnostic accuracy and therapeutic efficacy. Advanced clinical studies with immune checkpoint inhibitors (ICIs) have shown limited benefits in PCa, whereas subgroups of PCa with mismatch repair (MMR) or CDK12 inactivation may benefit from ICIs treatment. In this review, we summarized the targeted agents of PCa in clinical trials and their underlying mechanisms, and further discussed their limitations and future directions.
... AR-Vs are expected to be used as novel therapeutic targets for PC. Galeterone inhibits the AR signaling pathway through a variety of mechanisms, including inhibition of CYP17, competitive inhibition of AR, and degradation of full-length AR and AR-V7 proteins [46]. In this multicenter phase 3 clinical trial, 953 patients with mCRPC were recruited to screen for AR-V7 status using the AdnaTest for CTC mRNA. ...
Article
Full-text available
Purpose Liquid biopsy refers to the detection and analysis of the components from biological fluids non-invasively, including circulating tumor cells, nucleic acids, and extracellular vesicles (EVs). It is necessary to review the clinical value of liquid biopsy assays in PC and explore its potential application. Materials and methods We systematically reviewed of PubMed was performed to identify relevant literature on potential clinical applications of circulating tumor cells, circulating nucleic acids, and EVs in prostate cancer (PC). Results Liquid biopsy has emerged as a powerful tool to elucidate dynamic genomic, transcriptomic, and epigenomic tumor profiling in real-time. Here, the potential clinical applications of liquid biopsy include early detection, prognosis of survival, assessment of treatment response, and mechanisms of drug resistance in PC. Conclusions Liquid biopsy provides great value in diagnosis, prognosis, and treatment response in PC. Characterization of liquid biopsy components provides benefits both to unravel underlying resistance mechanisms and to exploit novel clinically actionable targets in PC. In addition, we suggest that analysis of multiparametric liquid biopsies should be analyzed comprehensively, assisting in monitoring tumor characteristics in real-time, guiding therapeutic selection, and early therapeutic switching during disease progression.
... [16][17][18] The positivity of these markers indicates poor prognosis in AR-targeting treatment. However, due to controversy over the testing techniques and the lack of validation with a large cohort, 19 the clinical availability of these markers is still limited. IDC-P as a pathological entity is easy to be detected by routine pathological testing. ...
Article
Full-text available
Background: To explore whether metastatic castration-resistant prostate cancer (mCRPC) patients with distinct intraductal carcinoma of the prostate (IDC-P) subtypes respond differently to abiraterone and docetaxel treatment. Methods: We retrospectively analyzed data of 170 mCRPC patients receiving abiraterone or docetaxel as first-line therapy. PSA response, PSA progression-free survival (PSA-PFS), radiographic progression-free survival (rPFS), and overall survival (OS) were analyzed based on the presence of IDC-P and its subpatterns. Results: IDC-P was confirmed in 91/170 (53.5%) patients. Among them 36/91 (39.6%) and 55/91 (60.4%) harbored IDC-P patterns 1 and 2, respectively. Patients with IDC-P pattern 1 shared similar clinical outcomes to those without IDC-P in both abiraterone and docetaxel treatment. However, against cases without IDC-P or with IDC-P pattern 1, patients with IDC-P pattern 2 had markedly poorer prognosis in either abiraterone (mPSA-PFS: 11.9 vs. 11.1 vs. 6.1 months, p < 0.001; mrPFS: 18.9 vs. 19.4 vs. 9.6 months, p < 0.001) or docetaxel (mPSA-PFS: 6.2 vs. 6.6 vs. 3.0 months, p < 0.001; mrPFS: 15.1 vs. 12.6 vs. 5.5 months, p < 0.001) treatment. For patients without IDC-P, docetaxel had comparable therapeutic efficacy with abiraterone. However, the efficacy of docetaxel was significantly inferior to abiraterone in patients with either IDC-P pattern 1 (mPSA-PFS: 6.6 vs. 11.1 months, p = 0.021; mrPFS: 12.6 vs. 19.4 months, p = 0.027) or pattern 2 (mPSA-PFS: 3.0 vs. 6.1 months, p = 0.003; mrPFS: 5.5 vs. 9.6 months, p = 0.007). Conclusion: Compared to docetaxel, abiraterone exhibited better efficacy in patients with IDC-P of either pattern. However, IDC-P pattern 2 responded unsatisfactorily to either abiraterone or docetaxel therapy. Novel therapeutic strategies for IDC-P pattern 2 need further investigations.
Article
Full-text available
Breast cancer is a major cause of death worldwide. The complexity of endocrine regulation in breast cancer may allow the cancer cells to escape from a particular treatment and result in resistant and aggressive disease. These breast cancers usually have fewer treatment options. Targeted therapies for cancer patients may offer fewer adverse side effects because of specificity compared to conventional chemotherapy. Signaling pathways of nuclear receptors, such as the estrogen receptor (ER), have been intensively studied and used as therapeutic targets. Recently, the role of the androgen receptor (AR) in breast cancer is gaining greater attention as a therapeutic target and as a prognostic biomarker. The expression of constitutively active truncated AR splice variants in breast cancer is a possible mechanism contributing to treatment resistance. Therefore, targeting both the full-length AR and AR variants, either through the activation or suppression of AR function, depending on the status of the ER, progesterone receptor, or human epidermal growth factor receptor 2, may provide additional treatment options. Studies targeting AR in combination with other treatment strategies are ongoing in clinical trials. The determination of the status of nuclear receptors to classify and identify patient subgroups will facilitate optimized and targeted combination therapies.
Article
Galeterone, 3β-(hydroxy)-17-(1H-benzimidazole-1-yl)androsta-5,16-diene (Gal, 1) and VNPP433-3β, 3β-(1H-imidazole-1-yl-17-(1H-benzimidazole-1-yl)androsta-5,16-diene (2) are potent molecular glue degrader modulators of AR/AR-V7 and Mnk1/2-eIF4E signaling pathways, and are promising Phase 3 and Phase 1 drug candidates, respectively. Because appropriate salts can be utilized to create new chemical entities with enhanced aqueous solubility, in vivo pharmacokinetics, and enhanced in vitro and in vivo efficacies, the monohydrochloride salt of Gal (3) and the mono- and di-hydrochlorides salts of compound 2, compounds 4 and 5, respectively, were synthesized. The salts were characterized using 1H NMR, 13C NMR and HRMS analyses. Compound 3 displayed enhanced in vitro antiproliferative activity (7.4-fold) against three prostate cancer cell lines but surprisingly decreased plasma exposure in the pharmacokinetics study. The antiproliferative activities of the compound 2 salts (4 and 5) were equivalent to that of compound 2, but their oral pharmacokinetic profiles were significantly enhanced. Finally, and most importantly, oral administration of the parent compounds (1 and 2) and their corresponding salts (3, 4 and 5) caused dose-dependent potent inhibition/regression of aggressive and difficult-to-treat CWR22Rv1 tumor xenografts growth, with no apparent host toxicities and were highly more efficacious than the blockbuster FDA-approved prostate cancer drugs, Enzalutamide (Xtandi) and Docetaxel (Taxotere). Thus, the HCl salts of Gal (3) and VNPP433-3β (4 and 5) are excellent orally bioavailable candidates for clinical development.
Article
Galeterone, a novel prostate cancer candidate treatment, was discontinued after a Phase III clinical trial due to lack of efficacy. Galeterone is weakly basic and exhibits low solubility in biorelevant media (i.e., ~ 2 µg/mL in fasted simulated intestinal fluid). It was formulated as a 50–50 (w/w) galeterone-hypromellose acetate succinate spray-dried dispersion to increase its bioavailability. Despite this increase, the bioavailability of this formulation may have been insufficient and contributed to its clinical failure. We hypothesized that reformulating galeterone as an amorphous solid dispersion by KinetiSol® compounding could increase its bioavailability. In this study, we examined the effects of composition and manufacturing technology (Kinetisol and spray drying) on the performance of galeterone amorphous solid dispersions. KinetiSol compounding was utilized to create galeterone amorphous solid dispersions containing the complexing agent hydroxypropyl-β-cyclodextrin or hypromellose acetate succinate with lower drug loads that both achieved a ~ 6 × increase in dissolution performance versus the 50–50 spray-dried dispersion. When compared to a spray-dried dispersion with an equivalent drug load, the KinetiSol amorphous solid dispersions formulations exhibited ~ 2 × exposure in an in vivo rat study. Acid–base surface energy analysis showed that the equivalent composition of the KinetiSol amorphous solid dispersion formulation better protected the weakly basic galeterone from premature dissolution in acidic media and thereby reduced precipitation, inhibited recrystallization, and extended the extent of supersaturation during transit into neutral intestinal media.Graphical Abstract
Article
VNPP433-3β (compound 2, (3β-(1H-imidazole-1-yl)-17-(1H-benzimidazole-1-yl)-androsta-5,16-diene), a multitarget anticancer agent has emerged as our lead next generation galeterone analogs (NGGA). Compound 2 is currently in development as potential new therapeutic for prostate and pancreatic cancers. The preliminary toxicity study reveals that the compound 2 was better tolerated by the normal male CD-1 mice than the male Nude mice. The maximum tolerated dose (MTD) in the Nude mice was estimated to be between 25 < 50 mg/kg. After oral dosing of compound 2 to male and female rats, the plasma concentration versus time curves were very consistent between animals and the AUClast increased with dose. Many plasmas concentration versus time curves profiles were nearly flat over 24 hr., suggesting extended absorption from the GI tract. Consequently, reliable values for half-life and AUCinf were not determined. Calculated oral bioavailability (using oral AUClast and excluding the outlier IV animal) ranged from 32-47%. This should be considered a minimum value since the contribution to true AUC beyond 24 hr. is clearly not zero. Clearly, these toxicology and pharmacokinetics parameters pave the way for understanding the anticancer pharmacological actions and provide a meaningful basis for further preclinical development and eventual clinical development.
Article
We report herein, the discovery of BMS-737 (compound 33) as a potent, non-steroidal, reversible small molecule inhibitor demonstrating 11-fold selectivity for CYP17 lyase over CYP17 hydroxylase, as well as a clean xenobiotic CYP profile for the treatment of castration-resistant prostate cancer (CRPC). Extensive SAR studies on the initial lead 1 at three different regions of the molecule resulted in the identification of BMS-737, which demonstrated a robust 83% lowering of testosterone without any significant perturbation of the mineralocorticoid and glucocorticoid levels in cynomologous monkeys in a 1-day PK/PD study.
Chapter
First-line therapy for metastatic prostate cancer (PCa) is targeted at the androgen receptor (AR), which governs gene expression programmes that impact pro-tumorigenic phenotypes, including proliferation, survival, and DNA repair. As is the case for many cancers, ageing is one of the most significant risk factors for PCa. PCa develops mainly in older men, with a significantly higher frequency in African-American men. Diagnosis rarely occurs in men younger than 40 years old, with the majority of new cases diagnosed between the ages of 65 and 79. Current data suggest that only up to 10% of PCa cases are hereditary, especially in early onset disease, while other risk factors proposed in some studies, such as smoking and excess body weight, have not been universally accepted. This chapter will outline the mechanistic underpinnings of response and resistance to AR-directed therapy, including androgen deprivation therapy (ADT) and AR antagonists.
Article
Full-text available
Aim: Galeterone is an oral small molecule that disrupts androgen receptor (AR) signaling via multi-targeted mechanisms of action (MOA): (1) selective inhibition of CYP17 lyase; (2) competitive antagonism of androgen binding to AR; and (3) degradation of AR protein. Galeterone is a single molecule with potential advantage beyond approved CYP17 and AR inhibitors in that it combines AR degradation with AR antagonism and CYP17 suppression and has no steroid requirement. Methods: ARMOR2 (NCT#01709734) is a two part, Ph 2 study designed to confirm dose of reformulated galeterone (1700 mg-3400 mg QD for 12 wks) (Part 1) and assess safety and efficacy of 2550 mg QD for 12 wks (Part 2). As of April 18, 2014, 4 cohorts of CRPC patients (pts) have been treated at 2550 mg: non-metastatic treatment (tx) naive (TN) (M0 TN, n = 18), metastatic tx naive (M1 TN, n = 36), abiraterone refractory (Abi-R n = 21) and enzalutamide refractory (Enza-R, n = 2). Additional arms have been added. Results: PSA response was seen at 2550 mg in all tx groups. In 36 M1 TN pts, PSA declines of 30% and 50% (PSA30 and PSA50) were acheived in 89% and 81% of patients respectively. The M1 TN pts had best overall response per RECIST with 14/15 with stable disease (SD) and 1/15 with partial response (PR) in evaluable pts at 3 mos. Safety: galeterone was well tolerated at all doses (parts 1 and 2) with gr 1 or 2 treatment related AEs occurring in 57% of pts; the most frequent were nausea (34%), fatigue (23%) and pruritis (22%). There was a total of 19% gr 3 and 2% gr 4 related AEs. There was no mineralcorticoid excess (ME) or seizures. Maximum Response w/in 12 Weeks 2550 mg Dose N* PSA30 N (%) PSA50 N (%) Best Overall Response Per RECIST PR/SD (N**) M0 TN 14 10 (71) 9 (64) 0/2 (5) M1 TN 36 32 (89) 29 (81) 1/14 (15) Abi-R 12 2 (17) 0 (0) 0/5 (7) Enza-R 2 0 (0) 0 (0) NA Maximum Response w/in 12 Weeks 2550 mg Dose N* PSA30 N (%) PSA50 N (%) Best Overall Response Per RECIST PR/SD (N**) M0 TN 14 10 (71) 9 (64) 0/2 (5) M1 TN 36 32 (89) 29 (81) 1/14 (15) Abi-R 12 2 (17) 0 (0) 0/5 (7) Enza-R 2 0 (0) 0 (0) NA Conclusions: Galeterone has shown signifiant biochemical and clinical activity and is well tolerated with no ME or seizures. The study will continue to explore safety and efficacy of galeterone in pts with progression following abiraterone and/or enzalutamide, chemotherapy and in additional arms. Also, data on CTCs will be presented and correlated with outcomes. Disclosure: M. Taplin: Advisory Boards: Medivation, Janssen, Dendron, Bayer, Tokai (individually < $10,000 each) Research Funding: Genentech, Medivation, Bayer, Janssen; K.N. Chi. J. Cochran, W.J. Edenfield, U. Emmenegger, E.I. Heath, A. Koletsky, D. Lipsitz, R. Pili, O. Sartor and N.D. Shore: Investigator for the Tokai Pharmaceuticals clinical study TOK-200-10 (ARMOR2); F. Chu, M. Eisenberger, A. Hussain,L. Nordquist and M. Rettig: Grant support from TOKAI for participation in the ARMOR 2 study; R. Dhillon and J. Roberts: Employee of study sponsor - Tokai Pharmaceuticals; B. Montgomery: Grant support from TOKAI for participation in the ARMOR 2 study.
Article
Full-text available
Biosynthesis of extragonadal androgen may contribute to the progression of castration-resistant prostate cancer. We evaluated whether abiraterone acetate, an inhibitor of androgen biosynthesis, prolongs overall survival among patients with metastatic castration-resistant prostate cancer who have received chemotherapy. We randomly assigned, in a 2:1 ratio, 1195 patients who had previously received docetaxel to receive 5 mg of prednisone twice daily with either 1000 mg of abiraterone acetate (797 patients) or placebo (398 patients). The primary end point was overall survival. The secondary end points included time to prostate-specific antigen (PSA) progression (elevation in the PSA level according to prespecified criteria), progression-free survival according to radiologic findings based on prespecified criteria, and the PSA response rate. After a median follow-up of 12.8 months, overall survival was longer in the abiraterone acetate-prednisone group than in the placebo-prednisone group (14.8 months vs. 10.9 months; hazard ratio, 0.65; 95% confidence interval, 0.54 to 0.77; P<0.001). Data were unblinded at the interim analysis, since these results exceeded the preplanned criteria for study termination. All secondary end points, including time to PSA progression (10.2 vs. 6.6 months; P<0.001), progression-free survival (5.6 months vs. 3.6 months; P<0.001), and PSA response rate (29% vs. 6%, P<0.001), favored the treatment group. Mineralocorticoid-related adverse events, including fluid retention, hypertension, and hypokalemia, were more frequently reported in the abiraterone acetate-prednisone group than in the placebo-prednisone group. The inhibition of androgen biosynthesis by abiraterone acetate prolonged overall survival among patients with metastatic castration-resistant prostate cancer who previously received chemotherapy. (Funded by Cougar Biotechnology; COU-AA-301 ClinicalTrials.gov number, NCT00638690.).
Article
Full-text available
Galeterone (Gal) is a first-in-class multi-target oral small molecule that will soon enter pivotal phase III clinical trials in castration resistant prostate cancer (CRPC) patients. Gal disrupts androgen receptor (AR) signaling via inhibition of CYP17, AR antagonism and AR degradation. Resistance to current therapy is attributed to up-regulation of full-length AR (fAR), splice variants AR (AR-Vs) and AR mutations. The effects of gal and VNPT55 were analyzed on f-AR and AR-Vs (AR-V7/ARv567es) in LNCaP, CWR22Rv1 and DU145 (transfected with AR-Vs) human PC cells in vitro and CRPC tumor xenografts. Galeterone/VNPT55 decreased fAR/AR-V7 mRNA levels and implicates Mdm2/CHIP enhanced ubiquitination of posttranslational modified receptors, targeting them for proteasomal degradation. Gal and VNPT55 also induced significant apoptosis in PC cells via increased Bax/Bcl2 ratio, cytochrome-c release with concomitant cleavage of caspase 3 and PARP. More importantly, gal and VNPT55 exhibited strong in vivo anti-CRPC activities, with no apparent host toxicities. This study demonstrate that gal and VNPT55 utilize cell-based mechanisms to deplete both fAR and AR-Vs. Importantly, the preclinical activity profiles, including profound apoptotic induction and inhibition of CRPC xenografts suggest that these agents offer considerable promise as new therapeutics for patients with CRPC and those resistant to current therapy.
Article
Full-text available
It is unclear whether a single clone metastasizes and remains dominant over the course of lethal prostate cancer. We describe the clonal architectural heterogeneity at different stages of disease progression by sequencing serial plasma and tumor samples from 16 ERG-positive patients. By characterizing the clonality of commonly occurring deletions at 21q22, 8p21, and 10q23, we identified multiple independent clones in metastatic disease that are differentially represented in tissue and circulation. To exemplify the clinical utility of our studies, we then showed a temporal association between clinical progression and emergence of androgen receptor (AR) mutations activated by glucocorticoids in about 20% of patients progressing on abiraterone and prednisolone or dexamethasone. Resistant clones showed a complex dynamic with temporal and spatial heterogeneity, suggesting distinct mechanisms of resistance at different sites that emerged and regressed depending on treatment selection pressure. This introduces a management paradigm requiring sequential monitoring of advanced prostate cancer patients with plasma and tumor biopsies to ensure early discontinuation of agents when they become potential disease drivers.
Article
Full-text available
Background: The androgen-receptor isoform encoded by splice variant 7 lacks the ligand-binding domain, which is the target of enzalutamide and abiraterone, but remains constitutively active as a transcription factor. We hypothesized that detection of androgen-receptor splice variant 7 messenger RNA (AR-V7) in circulating tumor cells from men with advanced prostate cancer would be associated with resistance to enzalutamide and abiraterone. Methods: We used a quantitative reverse-transcriptase-polymerase-chain-reaction assay to evaluate AR-V7 in circulating tumor cells from prospectively enrolled patients with metastatic castration-resistant prostate cancer who were initiating treatment with either enzalutamide or abiraterone. We examined associations between AR-V7 status (positive vs. negative) and prostate-specific antigen (PSA) response rates (the primary end point), freedom from PSA progression (PSA progression-free survival), clinical or radiographic progression-free survival, and overall survival. Results: A total of 31 enzalutamide-treated patients and 31 abiraterone-treated patients were enrolled, of whom 39% and 19%, respectively, had detectable AR-V7 in circulating tumor cells. Among men receiving enzalutamide, AR-V7-positive patients had lower PSA response rates than AR-V7-negative patients (0% vs. 53%, P=0.004) and shorter PSA progression-free survival (median, 1.4 months vs. 6.0 months; P<0.001), clinical or radiographic progression-free survival (median, 2.1 months vs. 6.1 months; P<0.001), and overall survival (median, 5.5 months vs. not reached; P=0.002). Similarly, among men receiving abiraterone, AR-V7-positive patients had lower PSA response rates than AR-V7-negative patients (0% vs. 68%, P=0.004) and shorter PSA progression-free survival (median, 1.3 months vs. not reached; P<0.001), clinical or radiographic progression-free survival (median, 2.3 months vs. not reached; P<0.001), and overall survival (median, 10.6 months vs. not reached, P=0.006). The association between AR-V7 detection and therapeutic resistance was maintained after adjustment for expression of full-length androgen receptor messenger RNA. Conclusions: Detection of AR-V7 in circulating tumor cells from patients with castration-resistant prostate cancer may be associated with resistance to enzalutamide and abiraterone. These findings require large-scale prospective validation. (Funded by the Prostate Cancer Foundation and others.).
Article
e16075 Background: Galeterone (gal) is an orally available steroid analog for the treatment of castration resistant prostate cancer (CRPC). A significant food effect, similar to abiraterone, was found when gal, as powder in capsule (PIC), was dosed in a phase I CRPC trial and confirmed in a PK study in human volunteers.The objective of this study was to assess single-dose PK of gal after reformulation and to evaluate effect of food. Methods: In this single-center open-label study, 24 male subjects were assigned to receive once daily doses of between 100 and 3400 mg in either a 3-treatment, 3-period or 4-treatment, 4-period dose scheme. Treatment periods were separated by 7-day washout intervals. Plasma concentrations were measured over 72 hours to assess PK. Two novel tablet formulations were administered in 100 mg doses under fasted and/or fed condition compared to 2600 mg-PIC administered under fed conditions. Escalating doses of PIC (975 to 2600 mg) and tablet (850 to 3400 mg) formulations were evaluated to assess linearity. Results: Gal was well tolerated. Food effect has been negated with the tablet. Exposure from 1700 mg galeterone tablet (fed or fasted) is comparable to a 2,600 mg dose of PIC (fed) (Table). For tablet, AUC(inf) increased in a dose-related and dose-proportional manner from 6,689 ± 2,602 hxng/mL (850 mg) to 20131 ± 9,930 hxng/mL (2,550 mg). Conclusions: The tablet formulation negates the food effect previously observed. Gal may be dosed either fed or fasted with no difference in absorption, which reduces intra-patient variability in exposure. Tablet has more consistent bioavailability and a linear increase in AUC between escalating doses. [Table: see text]
Article
184 Background: Because CYP17 has a central role in converting progestogens to androgens, inhibitors have been developed to treat CRPC. CYP17 has both hydroxylase and lyase catalytic functions. However, selective hydroxylase inhibition causes build-up of progestogens and mineralocorticoids, resulting in secondary mineralocorticoid excess (ME), edema, hypokalemia and hypertension (treated clinically with prednisone). We analyzed four inhibitors for their effects on CYP17 hydroxylase and lyase activities, and more globally on the steroidogenic pathway. Methods: Human CYP17 expressed in yeast microsomes was incubated with pregnenolone or 17α-hydroxypregnenolone and products quantitated using LC/MS/MS. Galeterone, abiraterone, orteronel or ketoconazole were added, and hydroxylase or lyase IC 50 values calculated. In a separate experiment, H295R adenocarcinoma cells were incubated with a range of drug concentrations between 0.316 nM and 10 μM for 24 hours and media was analyzed for steroid production by LC/MS/MS. Results: All drugs inhibited CYP17. However, potency and selectivity for hydroxylase and lyase varied significantly. Galeterone was the most potent and selective CYP17 lyase inhibitor. Abiraterone most selectively inhibited hydroxylase, while orteronel and ketoconazole were less potent lyase inhibitors than galeterone. In the cell-based assay, all drugs inhibited testosterone synthesis ≥94% at 1 μM. However, at this concentration, evidence of hydroxylase inhibition was supported by significant elevation of progesterone (abiraterone increased 293-fold), mineralocorticoids (orteronel increased DOC 74-fold), or reductions in cortisol (abiraterone, orteronel, ketoconazole reduced by 91%, 70% and 94%, respectively). In contrast, galeterone produced minimal changes in cortisol (decreased 14%) and other intermediate precursors. Conclusions: Galeterone is a selective and potent CYP17 lyase inhibitor that also antagonizes and degrades the AR, and exhibits minimal evidence of deleterious steroid changes associated with ME. These experimental data recapitulate phase I clinical experience where no ME was observed or prednisone required in CRPC patients.
Article
Background: Biosynthesis of extragonadal androgen may contribute to the progression of castration-resistant prostate cancer. We evaluated whether abiraterone acetate, an inhibitor of androgen biosynthesis, prolongs overall survival among patients with metastatic castration-resistant prostate cancer who have received chemotherapy. Methods: We randomly assigned, in a 2:1 ratio, 1195 patients who had previously received docetaxel to receive 5 mg of prednisone twice daily with either 1000 mg of abiraterone acetate (797 patients) or placebo (398 patients). The primary end point was overall survival. The secondary end points included time to prostate-specific antigen (PSA) progression (elevation in the PSA level according to prespecified criteria), progression-free survival according to radiologic findings based on prespecified criteria, and the PSA response rate. Results: After a median follow-up of 12.8 months, overall survival was longer in the abiraterone acetate?prednisone group than in the placebo?prednisone group (14.8 months vs. 10.9 months; hazard ratio, 0.65; 95% confidence interval, 0.54 to 0.77; P<0.001). Data were unblinded at the interim analysis, since these results exceeded the preplanned criteria for study termination. All secondary end points, including time to PSA progression (10.2 vs. 6.6 months; P<0.001), progression-free survival (5.6 months vs. 3.6 months; P<0.001), and PSA response rate (29% vs. 6%, P<0.001), favored the treatment group. Mineralocorticoid-related adverse events, including fluid retention, hypertension, and hypokalemia, were more frequently reported in the abiraterone acetate?prednisone group than in the placebo?prednisone group. Conclusions: The inhibition of androgen biosynthesis by abiraterone acetate prolonged overall survival among patients with metastatic castration-resistant prostate cancer who previously received chemotherapy.
Article
Background: Despite the progress and innovation in chemo- and immunotherapies, androgen deprivation therapy remains the standard treatment of metastatic prostate cancer. However, progression to castrate resistance disease (CRPC) occurs in the majority of patients and intriguingly, over 80% of CRPC specimens continue to express androgen receptor (AR) and androgen-responsive genes, indicating that the AR axis remains paradoxically activated despite castration. Hence, several new classes of AR-targeting agents have been recently introduced into clinic, including more potent AR antagonists (enzalutamide). Despite the substantial clinical efficacy reported with Enzalutamide, resistance to this therapy has already been observed and reactivation of AR signaling following treatment occurs. Thus, the next major clinical challenge that we will face is identifying new therapies that prevent or treat anti-androgen resistance. Galeterone is a novel drug that exhibits three mechanisms of action to inhibit AR activity, via inhibition of de novo androgen synthesis, blocking the ligand binding domain to prevent androgen binding, and inducing AR degradation. Thus, in this study we evaluated the efficacy of this multi-potent drug to inhibit AR activity in enzalutamide resistant cells. Results: As a pre-clinical model of enzalutamide resistance, drug resistant and CRPC cell lines were derived from three generations of serially passaged enzalutamide resistant, or vehicle control treated, LNCaP xenografts. Resistant cells were maintained in vitro under constant exposure to 10 μM of enzalutamide and were used to study the anti-cancer and AR targeting effects of galeterone in the enzalutamide resistant setting. Using crystal violet and MTT assays, we found that galeterone had anti-proliferative effects in LNCaP cells, in CRPC cells, and most importantly, in those resistant to enzalutamide. Compared to enzalutamide treatment, galeterone induced a drastic decrease in Probasin luciferase reporter (AR) activity, a greater reduction in AR and prostate-specific antigen (PSA) protein expression, and the inhibition of AR nuclear translocation in all cell lines. Strikingly, these effects were still observed in resistant cells, which show no decrease in AR expression or nuclear translocation upon enzalutmide treatment. Together, our data show that galeterone strongly inhibits AR activity and suppresses castration-resistant LNCaP growth as well as enzalutamide -resistant cell growth in vitro. Conclusion: In this study, we provide a preclinical proof-of-principle that Galeterone is a potent inhibitor of the AR pathway and may represent the next generation of hormone therapy for patients with not only CRPC but also Enzalutamide resistant disease. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C89. Citation Format: Nader Al Nakouzi, Chris Wang, Douglas Jacoby, Martin E. Gleave, Amina Zoubeidi. Galeterone suppresses castration-resistant and enzalutamide-resistant prostate cancer growth in vitro. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C89.
Article
Background: Enzalutamide is a novel antiandrogen with proven efficacy in metastatic castration-resistant prostate cancer (mCRPC). Objective: To evaluate enzalutamide's effects on cancer and on androgens in blood and bone marrow, and associate these with clinical observations. Design, setting, and participants: In this prospective phase 2 study, 60 patients with bone mCRPC received enzalutamide 160mg orally daily and had transilial bone marrow biopsies before treatment and at 8 wk of treatment. Outcome measurements and statistical analysis: Androgen signaling components (androgen receptor [AR], AR splice variant 7 (ARV7), v-ets avian erythroblastosis virus E26 oncogene homolog [ERG], cytochrome P450, family 17, subfamily A, polypeptide 1 [CYP17]) and molecules implicated in mCRPC progression (phospho-Met, phospho-Src, glucocorticoid receptor, Ki67) were assessed by immunohistochemistry; testosterone, cortisol, and androstenedione concentrations were assessed by liquid chromatography-tandem mass spectrometry; AR copy number was assessed by real-time polymerase chain reaction. Descriptive statistics were applied. Results and limitations: Median time to treatment discontinuation was 22 wk (95% confidence interval, 19.9-29.6). Twenty-two (37%) patients exhibited primary resistance to enzalutamide, discontinuing treatment within 4 mo. Maximal prostate-specific antigen (PSA) decline ≥ 50% and ≥ 90% occurred in 27 (45%) and 13 (22%) patients, respectively. Following 8 wk of treatment, bone marrow and circulating testosterone levels increased. Pretreatment tumor nuclear AR overexpression (> 75%) and CYP17 (> 10%) expression were associated with benefit (p = 0.018). AR subcellular localization shift from the nucleus was confirmed in eight paired samples (with PSA decline) of 23 evaluable paired samples. Presence of an ARV7 variant was associated with primary resistance to enzalutamide (p = 0.018). Limited patient numbers warrant further validation. Conclusions: The observed subcellular shift of AR from the nucleus and increased testosterone concentration provide the first evidence in humans that enzalutamide suppresses AR signaling while inducing an adaptive feedback. Persistent androgen signaling in mCRPC was predictive of benefit and ARV7 was associated with primary resistance. Patient summary: We report a first bone biopsy study in metastatic prostate cancer in humans that searched for predictors of outcome of enzalutamide therapy. Benefit is linked to a pretreatment androgen-signaling signature. Trial registration: ClinicalTrials.gov identifier NCT01091103.