throbber
V O L U M E 2 2 䡠 N U M B E R 6 䡠 M A R C H 1 5 2 0 0 4
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`Antiandrogen Withdrawal Alone or in Combination
`With Ketoconazole in Androgen-Independent Prostate
`Cancer Patients: A Phase III Trial (CALGB 9583)
`Eric J. Small, Susan Halabi, Nancy A. Dawson, Walter M. Stadler, Brian I. Rini, Joel Picus,
`Preston Gable, Frank M. Torti, Ellen Kaplan, and Nicholas J. Vogelzang
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Antiandrogen withdrawal (AAWD) results in a prostate-specific antigen (PSA) response (decline in PSA
`level of ⱖ 50%) in 15% to 30% of androgen-independent prostate cancer (AiPCa) patients. Thereafter,
`adrenal androgen ablation with agents such as ketoconazole (K) is commonly utilized. The therapeutic
`effect of AAWD alone was compared with simultaneous AAWD and K therapy.
`
`Patients and Methods
`AiPCa patients were randomized to undergo AAWD alone (n ⫽ 132), or together with K (400 mg orally
`[po] tid) and hydrocortisone (30 mg po each morning, 10 mg po each evening; n ⫽ 128). Patients who
`developed progressive disease after AAWD alone were eligible for deferred treatment with K.
`
`Results
`Eleven percent of patients undergoing AAWD alone had a PSA response, compared to 27% of patients
`who underwent AAWD and simultaneous K (P ⫽ .0002). Objective responses were observed in 2% of
`patients treated with AAWD alone compared to 20% in patients treated with AAWD/K (P ⫽ .02). There
`was no difference in survival. PSA and objective responses were observed in 32% and 7%, respectively,
`of patients receiving deferred K, and were more common in patients with prior AAWD response.
`Treatment with K was well tolerated, and resulted in a decline in adrenal androgen levels, which rose at
`the time of disease progression.
`
`Conclusion
`K has modest activity in AiPCa patients, while AAWD alone has minimal activity. Adrenal androgen levels
`fall with treatment with K and then climb at the time of progression, suggesting that progressive disease
`while on K may be due to tachyphylaxis to the adrenolytic properties of K.
`
`J Clin Oncol 22:1025-1033.
`
`INTRODUCTION
`
`Prostate cancer is the most common cancer in
`men and will account for more than 30,000
`deaths in 2003 [1]. The vast majority of deaths
`are due to the development of metastatic dis-
`ease unresponsive to androgen deprivation.
`Androgen deprivation, with gonadal andro-
`gen suppression with or without antiandro-
`gen, has been the standard of care in pa-
`tients with metastatic disease since the
`1940s [2] and is being increasingly utilized
`in patients with less advanced disease [3].
`The benefits of adding an antiandrogen
`such as flutamide or bicalutamide to go-
`
`nadal androgen suppression at the time of
`initiating androgen deprivation (early use)
`are modest at best [4]. The late addition of
`an antiandrogen after initial failure of an-
`drogen deprivation (late use) seems to result
`in prostate-specific antigen (PSA) declines,
`and in some cases, objective responses [5].
`Thus, at one point or another, most patients
`with advanced prostate cancer will be
`treated with an antiandrogen. Despite an-
`drogen deprivation, including the use of an
`antiandrogen, most patients will experience
`disease progression. For patients with pro-
`gressive disease, despite androgen depriva-
`tion, withdrawal of antiandrogen has been
`
`From the University of California San
`Francisco, San Francisco, and US Naval
`Medical Center, University of California
`San Diego, San Diego, CA; Cancer and
`Leukemia Group B Statistical Center,
`Duke University Medical Center,
`Durham, and Wake Forest Comprehen-
`sive Cancer Center, Winston-Salem,
`NC; Greenebaum Cancer Center,
`University of Maryland, Baltimore, MD;
`University of Chicago Medical Center,
`Chicago, IL; and Washington Univer-
`sity Barnard Cancer Center, St
`Louis, MO.
`
`Submitted June 10, 2003; accepted
`January 6, 2004.
`
`Supported by grants CA60138 (E.J.S. and
`B.I.R.), CA47577 (S.H. and E.K.),
`CA31983 (N.A.D.), CA41287 (W.M.S. and
`N.J.V.), CA77440 (J.P.), CA11789 (P.G.),
`and CA03927 (F.M.T.). The research for
`Cancer and Leukemia Group B Trial 9583
`was supported, in part, by grants from
`the National Cancer Institute (CA31946)
`to the Cancer and Leukemia Group B
`(Richard L. Schilsky, MD, Chairman). The
`research was also supported by Janssen
`Pharmaceutica Products, LP.
`
`Presented in part at the American Soci-
`ety of Clinical Oncology annual meeting,
`San Francisco, CA, May 12-15, 2001.
`
`The contents of this manuscript are
`solely the responsibility of the authors
`and do not necessarily represent
`the official views of the National
`Cancer Institute.
`
`Authors’ disclosures of potential con-
`flicts of interest are found at the end of
`this article.
`
`Address reprint requests to Eric J.
`Small, MD, UCSF Comprehensive Can-
`cer Center, University of California
`San Francisco, 1600 Divisadero St,
`Room A-718, San Francisco, CA 94115;
`e-mail: smalle@medicine.ucsf.edu.
`
`0732-183X/04/2206-1025/$20.00
`
`DOI: 10.1200/JCO.2004.06.037
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology.
`
`1025
`
`
`
`
`JANSSEN EXHIBIT 2063
`Mylan v. Janssen IPR2016-01332
`
`

`

`Small et al
`
`reported to result in a decline in PSA level of ⱖ 50% in 15%
`to 30% of patients [6-8]. The mechanism of this phenome-
`non has been attributed to alterations in the androgen sig-
`naling cascade, including mutations in the androgen recep-
`tor (AR), resulting in the antiandrogen behaving as an
`activator, not inhibitor, of the AR [9].
`The duration of decline in PSA observed with antian-
`drogen withdrawal (AAWD) is brief, with a median dura-
`tion of 3.5 to 5.0 months [6-8], after which further therapy
`is generally required. The mechanism by which prostate
`cancer patients develop disease progression after AAWD is
`not understood, but it has been postulated that persistence
`of a clone of cells with partial or full sensitivity to testoster-
`one might be provided a growth advantage by androgen
`produced by the adrenal glands. If this were the case, it could be
`anticipated that adrenal androgen suppression would demon-
`strate some anticancer activity in this setting. An early report
`suggested that the addition of aminoglutethimide (an adrenal
`steroid synthesis inhibitor) at the time of AAWD increased the
`percentage of patients with a decline in PSA over that which
`would be expected with antiandrogen alone [10].
`Ketoconazole is an azole antifungal agent which exerts
`its clinical effect through the inhibition of cytochrome P450
`14a-demethylase, a catalyst of the conversion of lanosterol
`to cholesterol. Ketoconazole has been in clinical use as an
`antifungal agent for more than 20 years. In its initial use as
`an antifungal agent, it was discovered that a proportion of
`men who used ketoconazole developed painful gynecomas-
`tia, which was later found to be due to the suppression of
`testicular and adrenal androgen production, and it was
`postulated that this adverse effect could be useful for pros-
`tate cancer therapy. Several trials have evaluated the use of
`ketoconazole in patients with androgen-independent pros-
`tate cancer (AiPCa), though most predated the use of PSA
`or an understanding of the AAWD syndrome [11-14]. Most
`recently, the use of ketoconazole after AAWD was reported
`in a trial of 48 patients, 30 (63%) of whom demonstrated a
`ⱖ 50% decline in PSA response. The decline of ⱖ 50% in
`PSA was comparable in patients who had initially re-
`sponded to AAWD and in those who had not initially re-
`sponded to AAWD at 65% v 40%, respectively. Toxicity was
`largely mild in these patients, consisting of grade 1 or 2
`nausea, fatigue, edema, hepatoxicity, and rash [15].
`It was hypothesized that the simultaneous addition of
`ketoconazole to AAWD would have additive anticancer
`activity, simultaneously targeting the stimulating effects of
`antiandrogen and adrenal androgens. Hence, the principal
`goals of this study were to prospectively evaluate and com-
`pare the effect of AAWD alone on PSA levels, compared
`with simultaneous AAWD and ketoconazole therapy. In
`addition, the effect of ketoconazole used in patients whose
`cancer had progressed after AAWD was also evaluated.
`
`PATIENTS AND METHODS
`
`Patients
`Eligible patients had histologically confirmed adeno-
`carcinoma of the prostate with progressive metastatic dis-
`ease, as defined below, despite anorchid testosterone levels
`(⬍ 50 ng/mL). Androgen deprivation therapy was required
`to include at least 4 weeks of ongoing therapy with an
`antiandrogen (flutamide, bicalutamide, or nilutamide).
`Ongoing gonadal androgen ablation with a luteinizing
`hormone-releasing hormone analog or orchiectomy was
`required. For patients with measurable disease, progression
`was defined as a greater than 25% increase in the sum of the
`products of the perpendicular diameters of all measurable
`lesions. For patients with “bone only” disease, a PSA greater
`than 5 ng/mL, which had risen from baseline on at least two
`successive occasions at least 4 weeks apart was required.
`Patients were required to have had metastatic disease dem-
`onstrated on imaging at some point during their history,
`but were not required to have demonstrated metastases on
`imaging at the time of enrollment. Patients were excluded if
`they had received prior chemotherapy, immunotherapy,
`experimental therapy, or prior treatment with ketocon-
`azole, aminoglutethimide, or corticosteroids if they had a
`Cancer and Leukemia Group B (CALGB) performance
`status of more than 2, total bilirubin level greater than 1.5⫻
`the upper levels of normal (ULN), or serum glutamic-
`oxaloacetic transaminase level greater than 3⫻ ULN. Be-
`cause of potential interactions with ketoconazole, no ongo-
`ing or concurrent use of
`terfenadine, astemizole, or
`cisapride was allowed. All participants signed an institu-
`tional review board–approved, protocol-specific, informed
`consent form in accordance with federal and institutional
`guidelines.
`
`Treatment
`After registration, patients were randomized to AAWD
`alone or AAWD ⫹ ketoconazole by the CALGB Statistical
`Center. This study was neither blinded nor placebo con-
`trolled. A conventional stratified random blocks design was
`used [16]. That is, within each stratum, patients were as-
`signed to the available two treatments in blocks of treatment
`assignments so that an equal number of patients was as-
`signed to each of the two treatment arms within each block.
`Randomization was stratified by four features that could
`potentially affect the likelihood of response, both to AAWD
`and/or ketoconazole therapy: (1) prior therapy with flut-
`amide, bicalutamide, or nilutamide; (2) continuous or in-
`termittent treatment; (3) initial or delayed antiandrogen
`therapy; and (4) imaging studies positive or negative for
`metastatic disease.
`Patient registration, subsequent randomization and
`data collection were managed by the CALGB Statistical
`Center. Furthermore, data quality was ensured by careful
`
`1026
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology. All rights reserved.
`
`

`

`AAWD and Ketoconazole for Prostate Cancer
`
`review of all data at the CALGB Statistical Center and by the
`study chairperson. Patients enrolled on the AAWD ⫹ keto-
`conazole arm received ketoconazole 400 mg tid po plus
`hydrocortisone 40 mg/d po (30 mg each morning and 10
`mg every night) continuously until disease progression or
`unacceptable toxicity, as described below. Patients random-
`ized to the AAWD-alone arm were required to cross over to
`treatment with ketoconazole on disease progression (see
`Response and Progression Criteria).
`Eligible patients were evaluated with a medical history
`and physical examination at study entry and monthly there-
`after. In addition to a complete medical history and physical
`examination at each visit, patients were evaluated for ad-
`verse events. No formal quality-of-life or pain assessment
`was undertaken. CBC, PSA, total bilirubin, alkaline phos-
`phatase, asparate transaminase, creatinine, glucose, and lac-
`tate dehydrogenase (LDH) were checked at baseline and
`then monthly. An endocrine panel including androstendi-
`one, dehydroepiandrosterone sulfate (DHEAS), dehydro-
`epiandrosterone (DHEA), and testosterone was obtained at
`baseline, at 1 month after starting therapy, at 3 months after
`starting therapy, and at the time of disease progression.
`Blood samples were obtained between 8:00 AM and 10:00 AM.
`Plasma was isolated, frozen, and shipped for analysis at a
`central laboratory to determine androstendione, DHEAS,
`DHEA, and testosterone levels. A bone scan and computed
`tomography scan of the abdomen and pelvis were obtained
`at baseline. If imaging studies were positive at baseline, they
`were repeated every 3 months.
`Replacement doses of hydrocortisone were continued
`as long as the patient was receiving ketoconazole. When the
`patient was no longer receiving ketoconazole, hydrocorti-
`sone was tapered by 5 mg every 3 days until completely
`discontinued. Antacids, H-2 blockers, and proton pump
`inhibitors were avoided but not explicitly prohibited. This
`study did not mandate that ketoconazole be taken on an
`empty stomach or with acidifying procedures. At each visit,
`toxicity was graded according to the National Cancer Insti-
`tute common toxicity criteria (CTC, version 2.0) and re-
`corded. In the event of grade 3 or higher hepatotoxicity or
`symptomatic peptic ulcer or gastritis, patients were re-
`moved from protocol treatment. Antiemetics other than
`corticosteroids were permitted. If grade 2 or 3 nausea per-
`sisted despite these measures, the patient was removed from
`therapy. Patients developing other grade 3 or higher toxicity
`had treatment held until toxicity resolved to grade 1 or
`better. Any patient developing grade 4 toxicity or grade 3
`toxicity persisting for longer than 4 weeks, except as out-
`lined previously, was removed from protocol treatment.
`
`Response and Progression Criteria
`This study was launched before publication of the Re-
`sponse Evaluation Criteria in Solid Tumors criteria or the
`PSA Consensus Criteria [17], so that a composite end point
`
`combining PSA changes with imaging changes was utilized.
`Therefore, for patients with measurable disease, a partial
`response was defined as a ⱖ 50% decrease in the sum of the
`products of the perpendicular diameters of all measurable
`lesions, together with a decline in PSA of ⱖ 75%, measured
`at least twice at least 2 weeks apart, whereas a complete
`response was complete resolution of all visible disease, and
`normalization of PSA on at least two occasions at least 2
`weeks apart. A complete response in patients with bone-
`only disease was defined as complete normalization of bone
`scan, again with a normalization of PSA, while a partial
`response in bone-only patients requires a greater than 75%
`decline in PSA with no new lesions on bone scan. Addition-
`ally, all patients had monitoring of PSA levels, and the
`percentage of patients with a decline in PSA of ⱖ 50%,
`documented on at least two successive occasions, at least 4
`weeks apart, was calculated per the PSA Consensus Criteria
`[17]. An intent-to-treat analysis was utilized. Patients who
`received less than 2 months of therapy for any reason (in-
`cluding progressive disease, toxicity, or withdrawal of con-
`sent) and who therefore did not have two sequential PSA
`values for response assessment, were nevertheless consid-
`ered to be nonresponders. Progressive disease was defined
`by a PSA increase of ⱖ 50% above nadir, on at least two
`successive occasions at least 1 month apart, with a mini-
`mum rise of 5 ng/mL [17]. Disease progression was also
`defined by new lesions on bone or computed tomography
`scan, or for patients with pre-existing measurable disease, a
`greater than 25% increase in the sum of the products of the
`perpendicular diameters of all measurable lesions. Survival
`was measured from the time of randomization to the time
`of death, and time to PSA progression was measured per
`Consensus Criteria [17].
`
`Statistical Methods
`The primary objective of this trial was to compare the
`response rates (composite end point, as defined above) of
`AAWD alone versus AAWD combined with ketoconazole
`and hydrocortisone. However, secondary objectives were to
`compare the percent of patients with a posttherapy decline
`in PSA of ⱖ 50%, documented on at least two successive
`occasions, at least 4 weeks apart, per the PSA Consensus
`Criteria [17]. Secondary objectives were also to evaluate the
`posttherapy PSA decline in patients who received ketocon-
`azole and hydrocortisone after developing progressive dis-
`ease despite AAWD. The study also sought to correlate
`posttherapy decline in PSA of ⱖ 50% PSA with survival, as
`well as to evaluate the prognostic value of several pretreat-
`ment patient characteristics. Finally, the study evaluated the
`relationship of pretreatment adrenal androgen levels, and
`changes in adrenal androgen levels with response to therapy
`and survival.
`With 119 patients per arm, and a one-sided ␣of .05, the
`independent two-group binomial test had 80% power to
`
`www.jco.org
`
`1027
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Small et al
`
`detect a difference in the percentage of patients with a
`partial or complete (composite) response from 25% in the
`AAWD group, to 40% in the AAWD ⫹ ketoconazole arm.
`Allowing for a 5% ineligibility rate, the target sample size
`was 250 patients.
`The study was monitored by the CALGB Data Safety
`and Monitoring Board. Planned interim analysis used the
`Lan-DeMets analog of the O’Brien-Fleming sequential
`boundary to maintain the overall level of significance of .05
`[18]. The Lans-DeMets stopping rule was applied to the
`composite response end point. At the final analysis, the ␣
`level was .045. An intent-to-treat approach was used in the
`analysis. Pearson ␹2 and Fisher’s exact test were used to
`compare the two arms with regard to response rates (com-
`posite end point), objective response rates, and 50% decline
`in PSA [19]. Exact confidence intervals based on the bino-
`mial distribution were used to estimate 95% CIs for the
`response rates. The Kaplan-Meier product-limit method
`was used to estimate overall survival and PSA progression-
`free survival by the two arms [20], and the log-rank test
`was used to compare the two arms on these outcomes
`(overall survival and PSA progression-free survival). The
`proportional hazards model was used to assess important
`factors for predicting survival time adjusting on the
`stratification factors.
`The relationship between overall survival and 50% de-
`cline in PSA from baseline was explored. To minimize “lead
`time bias,” landmark analyses were performed at 4, 8, 12,
`and 16 weeks postrandomization [21]. This method selects
`a fixed time point after initiation of therapy as a “landmark”
`and excludes patients who died before reaching the land-
`mark (eg, 8 weeks). Further, the patients alive at the land-
`mark were classified as responders or nonresponders de-
`pending on their 50% decline in PSA before the landmark.
`In these analyses, survival duration was defined as the time
`between the landmark (eg, 8 weeks) and death. The rela-
`tionship between survival duration, and PSA decline was
`examined. For the primary end point, a one-sided ␣⫽ .05
`was used to compute the 95% CI and the P value. For the
`secondary objectives and analyses, tests were performed
`using a two-sided ␣⫽ .05.
`
`RESULTS
`
`Patient Characteristics
`Two hundred sixty patients were enrolled onto this
`study. No consistent approach to screening patients for this
`trial was mandated. One hundred thirty-two were random-
`ized to AAWD alone and 128 were randomized to AAWD
`and ketoconazole. Patient characteristics, including strati-
`fication variables, are summarized in Table 1. With regard
`to stratification variables, approximately 36% of patients
`(on both arms) received prior flutamide, 59% had prior
`bicalutamide, and 4% to 5% had received nilutamide. Ap-
`
`Table 1. Baseline Characteristics
`
`Age, years
`Median
`Interquartile range
`Race, % white
`Sites of disease, %ⴱ
`Bone metastases
`Measurable disease
`Lymph node involvement
`Lung metastases
`Liver metastases
`Lymph node metastases only
`Performance status (0 to 1), %
`Opioid analgesic use, %
`Hemoglobin, g/dL
`Median
`Interquartile range
`PSA, ng/mL1
`Median
`Interquartile range
`Alkaline phosphatase, U/L
`Median
`Interquartile range
`LDH U/L
`Median
`Interquartile range
`Creatinine, mg/dL
`Median
`Interquartile range
`Prior therapy, %
`Flutamide
`Bicalutamide
`Nilutamide
`Androgen deprivation, %
`Intermittent
`Continuous
`Initial therapy for advanced
`disease, %
`CAB
`Monotherapy/ later antiandrogen
`
`AAWD
`(n ⫽ 132)
`
`AAWD and
`Ketoconazole
`(n ⫽ 128)
`
`Total
`(N ⫽ 260)
`
`71
`66-76
`78
`
`72
`64-76
`81
`
`72
`65-76
`79
`
`86
`31
`28
`5
`5
`5
`93
`30
`
`84
`40
`34
`6
`10
`8
`93
`28
`
`84
`35
`31
`5
`6
`7
`93
`29
`
`12.6
`11.7-13.3
`
`12.6
`11.1-13.6
`
`12.6
`11.6-13.5
`
`58
`17-162
`
`125
`91-239
`
`58
`20-137
`
`120
`85-225
`
`58
`20-151
`
`124
`90-235
`
`200
`171-405
`
`215
`189-409
`
`210
`176-405
`
`1.0
`0.9-1.2
`
`1.1
`1.0-1.3
`
`1.1
`0.9-1.3
`
`35.6
`59.1
`5.3
`
`12.9
`87.2
`
`59.9
`40.1
`
`37.5
`58.6
`3.9
`
`16.4
`83.6
`
`59.4
`40.6
`
`36.5
`58.9
`4.6
`
`14.6
`85.4
`
`59.6
`40.4
`
`Abbreviations: AAWD, antiandrogen withdrawal; PSA prostate-specific
`antigen; LDH, lactate dehydrogenase; CAB, combined androgen blockade.
`ⴱPatients may have more than one metastasis.
`
`proximately 15% of patients had received prior intermittent
`androgen deprivation, and approximately 60% had re-
`ceived initial combined androgen blockade (luteinizing
`hormone-releasing hormone analog plus antiandrogen).
`Imaging studies were positive for metastatic disease in 97%
`and 94% of patients in the AAWD and AAWD and keto-
`conazole arms, respectively. The two arms were similar
`regarding age, sites of disease, requirement for opioid anal-
`gesics, and a variety of pretreatment prognostic factors,
`including performance status, hemoglobin, PSA, alkaline
`phosphatase, LDH, and creatinine. The median age of pa-
`tients in both arms was just older than 70 years, and 93%
`
`1028
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology. All rights reserved.
`
`

`

`AAWD and Ketoconazole for Prostate Cancer
`
`Table 2. Summary of Clinical Outcome
`
`PSA decline ⱖ 50%
`No. of patients
`%
`95% CI
`Objective response rate
`No. of patients
`%
`95% CI
`Survival time, months
`Median
`95% CI
`Time to PSA progression in
`PSA responders,
`months
`Median
`95% CI
`No. of patients
`
`AAWD Alone
`(n ⫽ 132)
`
`AAWD and
`Ketoconazole
`(n ⫽ 128)
`
`34/128
`15/132
`27
`11
`7% to 17% 20% to 35%
`
`10/50
`1/41
`20%
`2%
`0.1% to 11% 11% to 32%
`
`P
`
`.002
`
`.020
`
`16.7
`14.3 to 21.5
`
`15.3
`13.40 to 19.5
`
`.936
`
`5.9
`5.3 to 10.1
`15
`
`8.6
`5.7 to 20.4
`34
`
`.063
`
`Abbreviations: AAWD, antiandrogen withdrawal; PSA, prostate-
`specific antigen.
`
`had a performance status of 0 or 1. The median serum PSA
`levels at study entry was 58 ng/mL. Thirty-one percent of
`patients in the AAWD arm and 39% of patients in the
`AAWD and ketoconazole arms had measurable disease.
`More than 80% in both arms had bone metastases, and
`approximately one-third had lymph node involvement.
`Approximately 30% of patients were using opioid analge-
`sics at the time of study entry.
`Clinical Outcome
`Relevant clinical outcomes, including PSA decline, ob-
`jective response rate, overall survival, and time to PSA pro-
`gression are summarized in Table 2. Overall, 15 (11%; 95%
`CI, 7% to 17%) of 132 patients undergoing antiandrogen
`withdrawal alone experienced a ⱖ 50% decline in PSA. By
`contrast, 34 (27%; 95% CI, 20% to 35%) of 128 patients
`who underwent AAWD and received simultaneous keto-
`conazole had a ⱖ 50% decline in PSA (P ⫽ .002). In patients
`with a ⱖ 50% decline in PSA, the subsequent median time
`to PSA progression was 5.9 months (95% CI, 5.3 to 10.1
`months) and 8.6 months (95% CI, 5.7 to 20.4 months) in
`the AAWD alone and AAWD and ketoconazole arms, re-
`spectively (log-rank P ⫽ .063). Figure 1 demonstrates the
`overall PSA progression-free survival by treatment arm for
`those patients who had a 50% decline in PSA. Objective
`responses in measurable disease were observed in 1 (2%;
`95% CI, 0.13% to 11%) of 41 of patients treated with
`AAWD alone, compared with 10 (20%; 95% CI, 11% to
`32%) of 50 in the AAWD and ketoconazole arm (P ⫽ .02).
`While no longer currently in use, when the composite end
`points described were applied, 8 (6%) of 132 of patients
`treated with AAWD had a response, compared with 22
`
`Fig 1. Overall prostate-specific antigen (PSA) progression-free survival by
`treatment arm in patients with 50% decline in PSA. AAWD, antiandrogen
`withdrawal.
`
`(17%) of 128 of patients on the AAWD/ketoconazole arm
`(one-sided P ⫽ .004).
`Eighty-two percent of patients (108 of 132) treated
`with AAWD alone ultimately received “deferred” ketocon-
`azole/hydrocortisone therapy. Twenty-four patients as-
`signed to initial AAWD alone never received deferred keto-
`conazole for a variety of reasons,
`including disease
`progression and withdrawal of consent. Of the 108 patients
`receiving deferred ketoconazole, a decline in PSA of ⱖ 50%
`was observed in 35 (32%) of 108 patients. An objective
`response was seen in 3 (7%) of 41 patients treated with
`AAWD followed by deferred ketoconazole. Although the
`study was not designed to compare the aggregate PSA de-
`clines and objective responses in patients treated with si-
`multaneous versus sequential AAWD and ketoconazole,
`there did not seem to be an advantage of either approach
`over the other. In aggregate, 40 (30%) of 132 patients
`treated with AAWD followed by ketoconazole had a PSA
`decline of ⱖ 50% compared with 34 (27%) of 128 of pa-
`tients treated with simultaneous AAWD and ketoconazole.
`The median survival time was 16.7 months (95% CI, 14.3 to
`21.5 months) in the AAWD-alone arm, and 15.3 months
`(95% CI, 13.4 to 19.5 months) in the group of patients who
`received simultaneous AAWD and ketoconazole therapy
`(two-sided P ⫽ .936; Fig 2).
`For those patients receiving sequential AAWD fol-
`lowed by ketoconazole, prior PSA “response” (decline of ⱖ
`50%) after AAWD seemed to be associated with a higher
`likelihood of PSA response to subsequent ketoconazole.
`Overall, 35 patients had a PSA response when treated with
`deferred ketoconazole. Ten of these PSA responses oc-
`curred in the group of 15 patients who had obtained a prior
`PSA response to AAWD (10 of 15; 67%), whereas an addi-
`tional 25 patients had a response to deferred ketoconazole
`from among a group of 117 patients (25 of 117; 21%) who
`had failed to achieve a PSA response after AAWD.
`
`www.jco.org
`
`1029
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Small et al
`
`Fig 2. Overall survival by treatment arm. AAWD, antiandrogen
`withdrawal.
`
`A proportional hazards model was used to identify
`predictors of overall survival. Pretreatment PSA, alkaline
`phosphatase, LDH, and hemoglobin levels, each dichoto-
`mized at the median; the presence or absence of weight loss
`at the time of study entry; and prior therapy with flutamide
`but not treatment arm, were predictors of survival (Table
`3). In addition, using a 4-week landmark analysis, a ⱖ 50%
`decline in PSA was a strong predictor of survival— 41
`months in patients with a ⱖ 50% PSA decline versus 13
`months in patients without a PSA decline (P ⬍ .0001; Fig 3).
`Toxicity
`Overall, 7% of patients undergoing AAWD alone had a
`grade 3 or 4 toxicity (all presumably attributed to causes
`
`Table 3. Predictors of Survival Time in Proportional Hazards Model
`(all patients)
`
`Variable
`
`Weight loss at baseline, ⬎ 5%
`(yes or no)
`Pretreatment PSA, ⬎ 58 v
`58 mg/mL
`Alkaline phosphatase, ⬎ 124 v
`124 U/L
`LDH, ⬎ 210 v 210
`Performance status, 2 v 0 or 1
`Hemoglobin, ⬎ 12.6 v 12.6
`Prior therapy, flutamide v
`nilutamide or bicalutamide
`Androgen deprivation, continuous v
`intermittent
`Initial therapy for advanced disease,
`monothrapy v CAB ⫹
`antiandrogen
`Treatment arm, AAWD ⫹
`ketoconazole v AAWD
`
`Hazard
`Ratio
`
`95% CI
`
`P
`
`2.63
`
`1.48 to 4.65 ⬍ .001
`
`1.60
`
`1.17 to 2.19
`
`1.50
`
`1.10 to 2.03
`
`1.33
`1.30
`0.64
`1.53
`
`1.00 to 1.78
`0.70 to 2.44
`0.47 to 0.86
`1.12 to 2.08
`
`1.29
`
`0.88 to 1.93
`
`1.18
`
`0.88 to 1.57
`
`.003
`
`.010
`
`.051
`.402
`.004
`.008
`
`.208
`
`.273
`
`1.09
`
`0.82 to 1.43
`
`.555
`
`Abbreviations: PSA, prostate specific antigen; LDH, lactate dehydroge-
`nase; CAB, combined androgen blockade; AAWD, antiandrogen with-
`drawal.
`
`Fig 3. Kaplan-Meier plot of overall survival as a function of ⱖ 50% decline
`in prostate-specific antigen (PSA), using a 1-month landmark analysis.
`
`other than AAWD itself), but no one toxicity occurred in
`more than 3% of patients. Twenty-one percent of patients
`receiving ketoconazole had grade 3 and 4 toxicities. The
`most common toxicities with ketoconazole were neurologic
`toxicity (4%), which consisted of motor neuropathy and
`ototoxicity, and malaise or fatigue (3%; Table 4). Grade 3 or
`4 hepatic toxicity was observed in 2% of patients receiving
`ketoconazole, and was not more common than in the
`AAWD arm.
`Adrenal Androgen Levels
`Baseline plasma adrenal androgen levels are available
`on 213 patients, 113 in the AAWD-alone arm, and 100 in
`the AAWD and ketoconazole arm. One hundred sixty-four
`patients had levels drawn after 1 month of therapy; 81, after
`3 months; and 111, at progression, which occurred at a
`median of 3.1 months after starting therapy (95% CI, 2.1 to
`4.9 months). Baseline median DHEAS and androstendione
`levels were within the normal range, while baseline DHEA
`levels were slightly elevated (Table 5). Baseline median
`DHEA, DHEAS, and androstendione levels were similar in
`both arms (data not shown). The median baseline testoster-
`
`Table 4. Treatment-Related Grade 3 and 4 Toxicities
`
`% of Patients
`
`AAWD Alone
`(n ⫽ 124)
`
`AAWD and
`Ketoconazole
`(n ⫽ 124)
`
`Hepatic toxicity
`Anorexia
`Neurotoxicity
`Cardiotoxicity
`Pulmonary
`Coagulation
`Nausea/vomiting
`Malaise/fatigue
`
`4
`0
`0
`0
`0
`0
`0
`0
`
`Abbreviation: AAWD, antiandrogen withdrawal.
`
`2
`2
`4
`1
`2
`1
`1
`4
`
`1030
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from jco.ascopubs.org on July 20, 2016. For personal use only. No other uses without permission.
`Copyright © 2004 American Society of Clinical Oncology. All rights reserved.
`
`

`

`AAWD and Ketoconazole for Prostate Cancer
`
`Table 5. Median Adrenal Androgen Levels in AAWD Plus Ketoconazole-Treated Patients
`
`Baseline (n ⫽ 100)
`
`Month 1 (n ⫽ 73)
`
`At Progression (n ⫽ 56)
`
`Adrenal Androgen
`
`Normal Range
`
`Median
`
`25th-75th Percentile
`
`Median
`
`25th-75th Percentile
`
`Median
`
`25th-75th Percentile
`
`DHEA
`DHEAS
`
`Androstendione
`
`(0.2-1.5 ng/mL)
`(31-4,668 ng/
`mL)
`(0.3-3.1 ng/mL)
`
`2.1
`317
`
`0.6
`
`1.4-3.3
`144-696
`
`0.5-1.1
`
`1.0
`30
`
`0.30
`
`0.8-1.4
`1-70
`
`0.2-0.4
`
`1.3
`116ⴱ
`
`0.45ⴱ
`
`0.8-2.0
`18-270
`
`0.3-0.6
`
`Abbreviations: AAWD, antiandrogen withdrawal; DHEA, dehydroeplandrosterone; DHEAS, DHEA sulfate.
`ⴱDifference between levels at month 1 and levels at progression are significant at P ⫽ .0001.
`
`one level was 13 ng/mL, and did not change over time in
`either treatment arm. DHEA, DHEAS, and androstendione
`levels did not change appreciably over time in the AAWD
`group (data not shown). By contrast, in the AAWD and
`ketoconazole group, there was a decline in levels of all three
`adrenal androgens (DHEA, DHEAS, androstendione), ac-
`counting for declines from baseline of 54%, 90%, and 58%,
`respectively, at 1 month. There was a rise in all three adrenal
`androgen levels at the time of disease progression, though
`not back to baseline (Table 5). The difference between
`DHEAS and androstendione levels at month 1 and at the
`time of progressive disease are significant (P ⫽ .0001).
`
`DISCUSSION
`
`This prospective, randomized phase III trial has compared
`changes in PSA levels as well as objective responses in met-
`astatic AiPCa patients treated with either AAWD alone or
`with AAWD plus simultaneous ketoconazole and hydrocorti-
`sone replacement therapy. No difference in survival was ob-
`served, though the planned use of deferred ketoconazole in the
`AAWD-alone arm may have blunted a treatment effect.
`This study has demonstrated that PSA declines and
`objective responses after AAWD are uncommon. A PSA
`decline of ⱖ 50% was observed in 11% of patients under-
`going AAWD, while an objective response proportion of
`2% was noted. The median time to PSA progression was
`brief, at 5.9 months. The AAWD phenomenon has been
`appreciated for many years [6-8], though there are few
`prospective trials carefully assessing its frequency. To our
`knowledge, this is the single largest prospective study of
`AAWD. These data indicat

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