throbber
WCK1082
`Wockhardt Bio AG v. Janssen Oncology, Inc.
`IPR2016-01582
`
`1
`
`

`

`Published OnlineFirst November 2, 2035; DOI: 10.1158/1078-0432.CCR—15-i432
`
`Galeterone for Metastatic Castration-Resistant Prostate Cancer
`
`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
`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 agents—GYM? inhibition and AR antago—
`nism—with the novel mechanism of increasing AR protein
`degradation. These first assessments of gaieterone in mCRPC
`identified a well-tolerated dose that resulted in clinically
`significant reductions in prostate-specific antigen, and dem-
`onstrate the potential of this agent. In Vim) data and results of
`these studies have informed future investigation ofgaleterone,
`which will include Ali-related biomarker analyses.
`
`
`
`variants, such as AR-V7, which are produced in tumor cells as a
`result ofaberrant RNA splicing ofthe wildvtype 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 ofandrogen-responsive
`genes even at castrate levels of androgens [15. 1G}. 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, flutamide, bicalutamide;
`refs 17,
`18, 19). As a resuit, androgenrindependent, but
`Ail-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,
`enzalutarnide). Recent data demonstrated that patients with
`detectable circulating tumor cells harboring Alt—V? had inferior
`responses to abiraterone or enzalutamide,
`including inferior
`prostatespecific antigen (PSA) response, clinical and radiograph-
`ic progression-free survival (PPS), 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 (20—26). Preclinical in trim;- and in vii/'0 data
`have shown that galeterone treatment in prostate cancer models
`resulted in a significant reduction in both full-length AR and AR-
`V? splice variant levels. In addition, galeterone has been shown to
`have activity against AR point mutations T87BA [20-25] and, in
`preliminary findings, to have activity in cells expressing the AR
`point mutation F876L (27).
`This article reports the safety and efficacy of galeterone in a
`phase 1 study, Androgen Receptor Modulation Optimized for
`Response (ARMORI), and the dose-escalation component of the
`phase 11 ARMOR2 study (ARMOR2 part 1). The dosewescalation
`component ofARMORZ 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 confirmed a significant food effect with the capsule formu-
`lation that was used in ARMORI (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 ofthe SDD tablet or2,600 mg of
`the capsule, which was the highest dose studied in ARMORI.
`Thus, the dose-escalation portion of ARMOR2 was conducted to
`evaluate the safety and tolerability of escalating doses ofthe SDD
`formulation and to determine the recommended dose for
`ARMORZ part 2 and ARMORB.
`
`Patients and Methods
`Patients
`
`Eligible men had histologically confirmed 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 ]PCWGI] criteria (29) in ARMORI, or PCWCZ
`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 (ECOC) performance status of S1. ARMORI excluded
`patients who had previously received chemotherapy, ketocona-
`zole, abiraterone, or enzalutamide. ARMOR2 part 1 permitted the
`enrollment of abirateronerrefractory patients, provided it had
`been discontinued 24 weeks before enrollment and that the
`duration of therapy was 26 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 ARMORI. Patients with indeterminate lung
`nodules were eligible. Other exclusion criteria included any
`previous radium-223, strontium, or samarium therapy within 8
`weeks ofenrollment; radiotherapy 34 weeks before enrollment or
`completed radiotherapy in ARMOR]; or radiotherapy S3 weeks
`(:2 weeks for single-fraction radiotherapy) in ARMORZ pan 1.
`Patients were excluded if they had previous treatment with inves-
`tigational drugs or agents that could have interfered with the
`efficacy 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 x
`ULN, hemoglobin level 59.0 gde, platelet count 3100 x 109,11,
`absolute neutrophil count 51.5 x 109/1, and serum potassium
`level <35 mmolfL, 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
`ARMORi (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 effi cacy oforal
`
`www.aacrjournals.org
`
`Clln Cancer Res: 22(6) March 15, 2016
`
`1357
`
`Downloaded from clincancerres.aacrjournals.org on March 31, 2017. © 2016 American Association for Cancer Research.
`
`2
`
`2
`
`

`

`Published OnlineFirsi November 2, 2015; DOI: 10.1158/1078~D432.CCR-15-1432
`
`Montgomery et al.
`
`galeterone for chemotherapy-naive patients with CRPC. The
`primary goals were to find the optimal dose of galeterone with
`an acceptable safety profile, defined as an observed dose-lim-
`iting toxicity (DLT) rate of 535%, and to identify a dose for
`further phase II study. The dose equivalence component of
`ARMOR2 (i.e,, part I; NCT01709734) evaluated the pharma-
`cokinetics (PK), safety, and efficacy of a new formulation of
`galeterone with improved bioavailability. A micronized pow—
`der formulation (capsule) was used in ARMORI and an SDD
`formulation was used in ARMORZ 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 Ilelsinki and
`the FDA regulations. The protocols were approved by the
`institutional review board of each study site.
`In ARMORI, galeterone capsules (micronized powder, 325
`mg) were administered orally as (i) 650 mg in the evening, (ii)
`975 mgin 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
`patientaselected meal, except for the 975 mg morning dose cohort,
`which received a high-fat, highecalorie nutritional supplement
`(Novasource Renal, Nestle llealthCare Nutrition, Florham Park,
`NJ) in place ofthe meal. Enrollment target was 6 patients per dose
`cohort. If an acceptable safety profile was determined by the
`internal monitoring committee (lMC; DLT rate 535% or 52 of
`5 patients in cohorts of 6 patients), subsequent dose levels and
`schedules were opened for enrollment. If 23 of 6 patients expe
`rienced DLTs, dose de-escalation was required. DLTs were defined
`as any study drug—related grade 3 or higher adverse event [AEU'
`National Cancer Institute Common Terminology Criteria for
`Adverse Events (CTCAE) version 4.0] considered to be possibly,
`probably, or definitely 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 significant
`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 first dose.
`The planned treatment duration ofboth studies was 12 weeks,
`with optional extension closing for eligible patients based on
`safety and tolerability during the lzeweek 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 first 4 closing cohorts ofARMORI, 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 first 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 ARMORZ part 1, blood samples
`for PK analyses were obtained before (hour 0) and 2, 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 S, 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 ofpregnenolone, 17-hydro-
`xyprogesterone, deoxycorticosterone, 11-deoxycortisol, cortico-
`sterone, cortisol, dehydroepiandrosterone sulfate (DIIEAS),
`androstenedione, and testosterone concentrations.
`
`Data Analysis
`Efficacy endpoints included the proportion of responders [PSA
`decrease 250% [PSASO] and 230% (1351960)), 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
`MR1 using PCWCZ and RBCIST v1.1. PSA efficacy was based on
`the intentvto-treat population (UT), defined as enrolled patients
`who received at least 1 dose ofstudy drug. Response was based on
`measurable disease in both studies. Time to progression, PFS
`defined as the time from first dose of study drug until objective
`CRPC progression or death, whichever occurred first, and 08 were
`the endpoints assessed in the ARMORI extension phase, Descrip-
`tive statistics were used for most variables (it, mean, SD, median,
`minimum, and maximum forcontinuous variables and frequency
`and percentage for categorical variables).
`
`Results
`Patients
`Baseline patient and disease characteristics are presented
`in Table 1, In ARMORl, 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; n = 5; nausea,
`chronic obstructive pulmonary disease exacerbation (event onset
`before (losing), elevated aspartate aminotransferasefalanine ami-
`notransferase levels
`(AST/ALT; n = 2), 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 (n = 5), or
`withdrawal of consent/personal choice [it = 2; 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<1 to
`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, n = 3), and S in the
`3,400-mg cohort. Six patients discontinued the study before 12
`weeks because of TEAEs [n = 4; angioedema (in an African-
`American who was receiving the angiotensinuconverting enzyme
`inhibitor, lisinopril), rash, weakness, and tremulousness] or dis-
`ease progression (n : 2). All 3 patients with abiraterone-resistant
`disease completed the 12-week phase ofthe study. Nineteen of22
`patients who completed the study participated in the optional
`
`1358 Clln Cancer Res; 22(6) March 15, 2016
`
`Clinical Cancer Research
`
`3
`Downloaded from clincancerresaacrjournalsorg on March 31. 2017. © 2016 American Association for Cancer Research.
`
`3
`
`

`

`Published OnlineFirst November 2, 2015; DOI: 10.1158/1078-0432.CCR-15-1432
`
`Tablet. Baseline characteristics
`
`ARMOR1
`(N = 49)
`68 (47a89)
`
`ARMORZ Part1
`(N = 28)
`70 (48-90)
`
`43 (are)
`3 (6.1)
`1 (2.0)
`2 (4‘1)
`25 (51.0)
`25
`15
`9
`7
`6
`3
`17
`
`49 (100)
`l (2)
`27 (55)
`24 (49)
`NA
`NA
`
`24 (85.7)
`2 (7.1)
`1 (3.6)
`1 (3.6)
`24 (85.7)
`24
`10
`8
`1
`1
`0
`11
`
`28 (100)
`2 (7.1)
`16 (57.1)
`12012.9)
`3 (i0.7)
`0
`
`characteristic
`Age. median (range). y
`Ethnicity, n (%)
`White
`African-American or black
`Asian
`Other
`Metastatic disease (M1). in 0%.)
`Bone. n
`Nodal. n
`Bone and nodal. n
`Visceral (liver and/or lung). n
`Visceral and bone. 17
`Visceral and nodal. n
`Soft tissue (not nodal. liver. or lung). 17
`Previous therapies. n (5%)
`Medical and/or surgical castration
`Immunotherapy
`Radiotherapy
`Surgery
`Abiraterone
`Enzalutamide
`ECOG, n (‘36)
`O
`1
`Missing
`Gleason score. median (range)6
`PSA. median (range). ng/dL
`Abbreviations: NA. not applicable.
`6Data were missing in 2 patients in ARMOR] and l patient in ARMOR 2 Part 1.
`
`45 (91.8)
`4 (8.2)
`0
`7 (6-10)
`24 (6-2005)
`
`22 (78.6)
`5 (17.9)
`l (3.6)
`3 (5—10)
`17.5 (SJ-6.760)
`
`extension phase; 2 of the patients with abiraterone-resistant
`disease were not eligible for the extension because of disease
`progression (Table 2). Overall duration oftherapy ranged from <1
`month to 14 months.
`
`Safety and tolerability
`ARMORI. Safety reviews were completed after all patients were
`closed in each cohort and the mac 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 A125
`required no action. The most common
`TEAEs were fatigue [17 patients (34.7%)], increased AST level
`
`Gaieterone for Metastatic Castration-Resistant Prostate Cancer
`
`[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
`treatmenterelated 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.
`
`ARMORZ Part 1. Galeterone tablets were well tolerated at all
`doses, as assesSed by the IMC. Safety reviews were completed after
`all patients were closed in each cohort. and the mac 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 ARMORI 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 ESQ—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 significant effect
`on exposure (AUC).
`The PK analysis plan of ARMOR2 was not designed to fully
`characterize the PK of galeterone. The ARMORZ 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
`(AUC0_6), was 2,646 -_i: 1.748 h - ngme. 2,684 :l: 2,043 h- ng/‘mL,
`
`Table 2. Treatment cohorts and patient disposition
`ARMORl—Galelerone capsules
`SN: 49)
`
`—
`Entered
`extension
`phase, 11
`3
`2
`3
`
`ARMORZ Part l—Galeterone 500 tablets
`(N = 281
`Completed
`12-week
`study, 11
`6
`11
`5
`
`Enrolled,
`n
`6
`14
`8
`
`Entered
`Extension
`phase, :1
`6
`9
`4
`
`Cohort
`1.700 mg
`2.550 mg
`3,400 mg
`
`Completed
`12-week
`Enrolled,
`study, it
`n
`Dosing cohort
`3
`6
`650 mg with meal
`5
`5
`975 mg with meal
`5
`6
`1.300 mg with meal
`5
`7
`1.950 mg with meal
`4
`6
`975 mg with supplementa
`5
`6
`1.950 mg divided doses with meal
`5
`6
`2,600 mg with meal
`5
`6
`2,500 mg divided doses with meal
`Novasource Renal. Nestle HealthCare Nutrition. Florham Park. New Jersey,
`bThree patients were eligible for the extension phase; however. only 2 patients were closed with gaieterone.
`
`24 2
`
`b
`
`23 “
`
`www.aacrjournals.org
`
`Clin Cancer Res; 22(6) March 15. 2016
`
`1359
`
`4
`Downloaded from clincancerres.aacnournalsorg on March 31. 2017. © 2016 American Association for Cancer Research.
`
`4
`
`

`

`Published OnlineFirst November 2, 2015; DOI: 10.1158l1078—0432.CCR-154432
`
`Montgomery et al.
`
`Table 3. Treatment-emergent AEs occurring in >10% of patients in ARMOR] or ARMORZ Part1
`ARMORl
`(N = 49)
`
`Grade 1 or
`2, n (9%)
`A5
`5 (10.2)
`Abdominal pain
`7 (14.3)
`increased alkaline phosphatase level
`7 (14.3)
`increased ALT level
`6 (12.2)
`Decreased appetite
`6 (12.2)
`Arthralgia
`13 (26.5)
`increased AST level
`i (2.0)
`Back pain
`6 (12.2)
`increased bllirubin level
`5 (10.2)
`Constipation
`7 (14.3)
`Cough
`11 (22,4)
`Diarrhea
`3 (6.1)
`Dizziness
`0
`Fall
`16 (32.7)
`Fatigue
`12 (24.5)
`Nausea
`11 (22.4)
`Pruritus
`5 (10.2)
`Rash
`4 (8.2)
`Urinary tract infection
`6 02.2)
`Vomiting
`5 (10.2)
`Decreased weight
`Abbreviations: ALT, alanine aminotransterase; AST, aspartate aminotransterase.
`
`Grade 3 or
`higher, a ($6)
`0
`0
`8 (16.3)
`0
`0
`3 (6.1)
`0
`1 (2.0)
`O
`0
`0
`0
`0
`1 (2.0)
`0
`O
`0
`0
`O
`0
`
`ARMORZ Part 1
`(N : 28)
`
`Grade 3 or
`higher, n (as)
`0
`0
`3 (10.7)
`0
`0
`1 (3.6)
`0
`0
`1 (3.6)
`0
`1 (3.6)
`0
`0
`0
`0
`0
`1 (3.6)
`0
`0
`0
`
`Gradei or
`2,1106)
`1 (3.6)
`0
`1 (3.6)
`6 (21.4)
`1 (3.6)
`1 (3.6)
`3 (10,7)
`O
`3 (10.7)
`3 (10.?)
`4 (14.3)
`3 (10.7)
`3 (10.7)
`9 (32.1)
`13 (46.4)
`9 (32.1)
`0
`4 (14,3)
`B (28.6)
`4 (14.3)
`
`and 2,528 :i: 1,529 b - ng/mL for the 1,700, 2,550, and 3,400 mg
`doses, respectively.
`
`Efficacy endpoints
`ARMORI. The TIT population for PSA efficacy included 49
`patients. Across all doses tested, 24 of 49 (49.0%} achieved a
`PSASO 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 PSASO 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
`PSA3O and 5 of12 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 (final)
`study visit; 71 :- 17], 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, 14—592 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 of17 patients (76.5%)
`in the extension phase;
`the remaining 4 patients had disease
`progression.
`
`Table 4. Summary of Potential AME AEs in ARMOR1 or ARMORZ Part 1
`Number of
`Attribution:
`AE
`incidences
`related/unrelateda
`Grade 2 hypokalemia
`1
`1/0
`Grade 3 hypokalemia
`3
`1/2
`Grade 1 peripheral edema
`1
`0/1
`Grade 2 peripheral edema
`3
`2/1
`aAll events were individual occurrences and not considered AME symptoms.
`
`ARMOR2 Part 1. The HT population for PSA efficacy in treatment?
`na'ive 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 230% in 16 of 25 patients (64.0%) and 250% in 12 of 25
`patients (48.0%). In the 2,550-mg dose cohort, 8 of 11 treatment—
`naive patients (72.7%) had a 230% decline in PSA from baseline
`and 6 of 11 patients (54.5%) had a 250% decline in PSA from
`baseline.
`in the 1,700—mg dose cohort 50% (3/6 patients)
`achieved a PSASO 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 PSASU (Fig. 13). 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 PSASO were 80% and 60% in the 2,550 rug/cl group,
`and 71.4% and 42.9% in the 3,400 mg/d group. Ofthe 3 patients
`treated with 2,550 rng/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
`atbaseline, 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, Dl-IEAS, and androstenedione concentrations.
`Median corticosterone level was increased from a median baseline
`of 204 ng/dL to 1,377.5 ng/di. at week 12, and cortisol and
`deoxycorticosterone levels were generally unchanged (Table 5).
`
`Discussion
`
`Results of ARMORI 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 virro, is safe and shows promising PSA
`
`1360 Clin Cancer Res; 22(6) March 15, 2016
`
`Clinical Cancer Research
`
`Downloaded from clincancerreseacdournalsorg on March 3?, 2017. © 2016 American Association for Cancer Research.
`
`5
`
`

`

`Published OnlineFirst November 2, 2015; DOI: 10.1158/1OTB-0432.0CR-15-1432
`
`Galeterone for Metastatic Castration-Resistant Prostate Cancer
`
`975 mm
`In=12l
`
`1.300 mgld
`tn=6i
`
`1,950 mgr‘d
`in=133
`
`2.600 mm
`(n=12)
`
`l
`550 mgrd a
`01:6)
`I
`f
`s
`’
`3
`a
`i
`5
`I
`
`"mm"- m-“m-"n
`----------" -‘----------------------
`
`--------‘-----
`-----------
`——-------“-- -------""""-“‘-----
`
`------
`---------""-"
`
`"-
`
`100
`
`0
`
`75
`
`r:
`a
`‘r
`E
`s 5°
`s
`E 25
`o
`c
`a:
`g’
`g
`0 25
`q _
`m
`o.
`'5 —50
`x
`ll
`E '75
`
`-
`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(n : 49).
`Patterned data points reflect 1 patient
`.
`who discontinued his gonadotropinw
`releasrng 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 PEA from
`baseline by total daily dose in
`evaluable treatment-naive patients in
`
`ARMOR2 Part 1 (n z 25} Patterned
`data point reflects a patient who only
`had i post-baseline PSA measurement
`(at 2 weeks). One patient in the
`3.400 mg/d group (n = 8) is not
`included in the graph because no post-
`baseline PSA measurements were
`completed. Abiraterone-refractory
`patients (N = 3) were not inciuded in
`this analysis. Reference lines: green,
`—50%,‘ orange, —30%. '. maximal PSA
`values >100% increase from baseline.
`
`400
`
`100
`'HI"I
`
`InD
`
`M"I
`
`1,700 my”
`m = 5)
`
`2550 my“
`m :1”
`
`“on maid
`(n = 7}
`
`,
`
`mm
`
`I
`
`NUI
`
`is.Do
`
`I
`
`'HlL'I'I
`
`I
`
`.4 OD
`
`
`
`MaximalPSAchangefrombaseline(“15.)
`
`
`
`
`
`responses in patients with rnCRPC. Results from phase 1 healthy
`volunteer PK studies and the PK results ofARMORZ part 1 support
`a 2,550 rug/d dOse of galeterone SDD tablet for use in future trials.
`All doses tested had similar safety and tolerability profiles.
`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,
`
`Table 5. Median (range) concentrations of the steroidogenic pathway markers
`in ARMORZ Part i
`
`Steroid
`Testosterone. ng/dL
`Androstenedione, ng/dL
`DHEAS, ug/dL
`Corticosterone, ng/dL
`Deoxycorticosterone. ng/dL
`Cortisol. pig/6L
`
`Baseline
`7.5 (3—22)
`32 (7-81)
`37.5(<15-220)
`204 (do-874)
`<16 (<lG—18)
`14,7 (LB-28.7)
`
`Median (range)
`Week 12
`2 (<i-14)
`14 (<S~34)
`18 (<15—i05)
`1.3775 (ST-4,375)
`<16 (<i6~89)
`18.i (4.i-35)
`
`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 specificity 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 ofcoadministration 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).
`Significant PSA declines were observed with all dose levels,
`Patients in ARMORI had an overall PSASO and PSASO of49% and
`22%, respectively, with the highest close (2,600 mg) showing
`PSASO and PSASO of75% and 42%, respectively. In ARMORZ part
`1, 2,550 mg of the SDD tablet formulation, the dose found to
`
`wwwaacriournalsorg
`
`Clin Cancer Res; 22(6) March 15. 2016
`
`3361
`
`Downloaded from clincancerres.aacrjournalsorg on March 3615, 2017. © 2016 American Association for Cancer Research.
`
`6
`
`

`

`Published OnlineFirst November 2, 2015; DOI: 10.1158l1078-0432.CCR-15-1432
`
`Montgomery et al.
`
`provide exposure similar to that of 2,600 mg of the capsule,
`resulted in greater PSASO and PSASO of 80% and 60%, respec—
`tively. These results are comparable with those observed in phase 1
`and 11 trials of abiraterone and enzalutamide (8. 11, 31). Of
`note,
`these results were marginally better than the 3,400 mg
`(PSA30 = 71%, PSASO = 43%) and 1,700 mg (PSA30 = 50%,
`PSASO = 50%) doses.
`Although ARMORI showed that increasing the dose resulted in
`a better PSA response, a phase 1 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 ARMOR]
`could be attributed to the study design, in thatthe blood sampling
`strategy was not optimal for assessment of PK parameters, and
`patienteselected meals precluded assessment of the effect of fat
`and calories.
`
`ARMORZ part 1 served as a bridging study between the original
`capsule formulation and the SDD tablet formulation, which was
`developed to have improved relative bioavailabiiity 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 ofthe SDD tablet was similar to that with 2,600 mg of
`the original capsule formulation—the dose in ARMORi
`that
`resulted in the best efficacy numbers (28). ARMORz 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 ofincrease 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
`ofthe 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 (ARMOiB-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-terrninal loss in the treatment
`naive cohort ofARMOR2 (33).
`
`Conclusion
`
`The efficacy and safety results from ARMOR] and ARMORZ
`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 ARMORZ part 2 and the
`
`phase III study (ARMORS-SV) using the SDD tablet formulation
`with improved bioavailability. Caleterone is well tolerated in
`CREE 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 ARMORZ 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 Conflicts of Interest
`L.T. Nordquist is a consultant/advisory board member for Bayer Pharma-
`ceuticals. WJ. Edenfield reports receiving speakers bureau honoraria from
`Astellas and Novartis. K.]. 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

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket