throbber
Urol Clin N Am 33 (2006) 227–236
`
`Changing Perspectives of the Role of Chemotherapy
`in Advanced Prostate Cancer
`Earle F. Burgess, MD, Bruce J. Roth, MD*
`Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center,
`Vanderbilt-Ingram Cancer Center, 777 Preston Research Building, Nashville, TN 37232-6307, USA
`
`The use of cytotoxic chemotherapy in ad-
`vanced prostate adenocarcinoma has been vali-
`dated by the recent demonstration of survival
`benefit in two large randomized phase III trials
`[1,2]. Before publication of these landmark trials,
`SWOG 9916 and TAX 327, no chemotherapeutic
`regimen had shown survival benefit in the treat-
`ment of androgen independent prostate cancer
`(AIPC). These trials provide new encouragement
`for the use of chemotherapy in all stages of dis-
`ease. Improved communication between medical
`and urologic oncologists and early patient referral
`for clinical trial participation remains essential for
`identifying new chemotherapeutic regimens with
`improved activity in AIPC and for defining the
`role of chemotherapy in earlier-stage disease.
`This article discusses the role of chemotherapy
`as the current standard of care for the treatment
`of AIPC and provides a historical perspective
`of the trials that preceded the development of
`current docetaxel-based regimens.
`
`Chemotherapy for AIPC
`
`Trials from the National Prostatic Cancer
`Project era
`
`In the 1980s, the National Prostatic Cancer
`Project (NPCP) conducted the largest series to
`date of phase II trials involving single and
`combinational cytotoxic agents in AIPC. The
`results of the NPCP and other early trials have
`
`* Corresponding author.
`E-mail address: bruce.roth@vanderbilt.edu
`(B.J. Roth).
`
`previously been reviewed and heavily criticized for
`erroneously concluding that many of the tested
`agents possessed clinically significant activity
`[3–6]. Although the trials were not designed to
`study impact on survival, patient median survival
`was generally no better than best supportive care,
`even though the response rates often exceeded
`50% with some regimens. The underlying biology
`and difficulties associated with studying advanced
`prostate cancer can explain the misleading conclu-
`sions drawn from some of the trials. The classic
`objective criteria for gauging tumor response in
`phase II trials continue to be bidimensional mea-
`surement of solid tumor lesions before and after
`therapy. Because prostate cancer often metasta-
`sizes to bone only, assessment of tumor response
`after therapy in AIPC has been difficult to achieve
`in an objective and reproducible manner. Imaging
`modalities do not provide accurate bidimensional
`measurement of skeletal lesions, and, before the
`routine use of serum prostate specific antigen
`(PSA) as a surrogate marker, the methodologic
`challenges of measuring tumor response were
`well recognized. Faced with these limitations, the
`NPCP-era trial investigators used response crite-
`ria that were often subjective and that lacked the
`specificity to provide accurate assessment of
`drug activity. For example, the presence of ‘‘sta-
`ble’’ disease, as defined by the absence of progres-
`sion on bone scan and acid phosphatase levels
`after 12 weeks of therapy, was included as one cri-
`terion for drug response in the NPCP guidelines
`[7]. The trials did show that patients who had
`‘‘stable’’ disease had longer median survival than
`those who had progressed, but they did not distin-
`guish whether disease stability was a result of
`treatment or reflected the underlying biology of
`
`0094-0143/06/$ - see front matter Ó 2006 Elsevier Inc. All rights reserved.
`doi:10.1016/j.ucl.2005.12.006
`
`urologic.theclinics.com
`
`JANSSEN EXHIBIT 2031
`Mylan v. Janssen IPR2016-01332
`
`

`

`228
`
`BURGESS & ROTH
`
`disease in those patients [8]. In the NPCP trials,
`approximately 90% of
`the patient
`responses
`were classified as ‘‘stable disease’’
`[4]. Because
`this response criterion is hardly specific for drug
`effect, false ‘‘activity’’ was incorrectly attributed
`to drugs that ultimately were shown to lack clini-
`cal benefit.
`
`Mitoxantrone
`
`After the NPCP and other trials of the 1980s,
`much debate existed over how best to measure
`tumor response in AIPC clinical trials. At the
`time, the general attitude regarding the use of
`chemotherapy in this setting was pessimistic and
`shared by urologists and oncologists alike. How-
`ever, in the 1990s, the use of post-treatment serum
`PSA decline was validated as a surrogate marker
`for drug activity, and trialists were hopeful that
`PSA response could provide a much needed
`objective criterion that was reproducible and
`could be used in patients who had isolated skeletal
`disease [9,10]. Measurement of PSA response, as
`defined by a O50% decline from pretreatment
`baseline, solved key problems from earlier trials
`and established an end point in subsequent trials
`that more accurately reflected drug activity.
`After acknowledgment of
`limited response
`criteria before the routine use of PSA decline,
`investigators considered palliation of symptoms as
`a primary endpoint in trials of cytotoxic agents.
`The idea originated in the AIPC setting after low-
`dose prednisone was shown to improve pain
`symptoms and quality of life [11]. Although PSA
`response has limitations, its superiority over his-
`torical standards quickly established this criterion
`as a gold standard for drug activity in phase II
`trials. Mitoxantrone, a member of the anthra-
`cenedione
`class
`that
`interacts with DNA
`topoisomerase II, was selected for evaluation of
`palliative ability based on a favorable side effect
`profile in elderly patients from an earlier phase
`II study [12–14]. Based on palliative endpoints,
`two phase III trials established that the addition
`of mitoxantrone provided superior response over
`glucocorticoids alone [15–17]. Mitoxantrone sub-
`sequently became the first cytotoxic agent ap-
`proved by the FDA for use in AIPC and was
`approved for palliative treatment based on these
`trials. After FDA approval, mitoxantrone re-
`mained first-line standard of care in symptomatic
`patients before the approval of docetaxel.
`Mitoxantrone provides effective palliative ther-
`apy. Although PSA response rates ranged from
`
`33% to 48% in three phase III trials, a survival
`advantage over best supportive care has never
`been shown [16–18]. Additional phase II trials
`have explored the feasibility of mitoxantrone in
`combination with other cytotoxic agents or as se-
`quential therapy with docetaxel-based regimens
`[19–23]. However, many of these studies accrued
`taxane-naive patients and need to be re-evaluated
`because docetaxel has been approved as first-line
`therapy. Recent trials have also studied the use
`of mitoxantrone in the second-line setting after
`patients have failed initial taxane therapy, al-
`though data suggest
`limited activity [24–26].
`Based on the promise of docetaxel-based regimens
`(see below), the role of mitoxantrone will likely
`continue to diminish.
`
`Estramustine
`
`Estramustine is a synthetic conjugate of estra-
`diol and a nitrogen mustard that was rigorously
`studied in the NPCP trials and found to have
`modest activity [4,27]. Although the drug was de-
`signed to target delivery of the mustard conjugate
`to malignant cells that overexpressed sex hormone
`receptors, estramustine acts through binding mi-
`crotubule-associated proteins and inducing micro-
`tubule destabilization [27,28]. Preclinical studies
`suggest that combination of estramustine with
`other antimicrotubule agents could potentiate
`a cytotoxic effect in vivo [29,30]. These observa-
`tions provided the scientific rationale for a large
`number of clinical studies of estramustine com-
`bined with other antimicrotubule agents. Many
`of these trials consistently showed that the addi-
`tion of estramustine improved PSA response rates
`over single-agent therapy in the phase II setting
`[31–49]. Finally, the activity of an estramustine-
`containing doublet was definitively established
`by the SWOG 9916 phase III trial, which reported
`a prolongation of survival in patients who had
`AIPC treated with docetaxel and estramustine
`(D/E). SWOG 9916 was not designed to evaluate
`the contribution of each drug to the regimen. The
`TAX 327 phase III trial evaluating the docetaxel/
`prednisone (D/P) doublet showed a similar sur-
`vival benefit in the docetaxel-treated arm. The
`results from the TAX 327 trial (see below) suggest
`that
`the efficacy of estramustine in the D/E
`doublet is limited and that the survival benefit is
`primarily due to docetaxel.
`Estramustine is associated with the develop-
`ment of thromboembolic events in up to 25% of
`treated individuals [50]. Despite this risk, interest
`
`

`

`CHEMOTHERAPY IN ADVANCED PROSTATE CANCER
`
`229
`
`in combining the drug with other agents persists
`[19,51–53]. Many of these trials were written be-
`fore the results of TAX 327 and SWOG 9916
`were known. Given the risk for thromboembolism
`and limited contribution to docetaxel-based regi-
`mens, the continued use of estramustine must be
`questioned.
`
`Taxanes
`
`In the combination phase II studies involving
`estramustine, the taxanes had some of the highest
`activity among the classes tested. The early-phase
`trials involving paclitaxel and docetaxel have
`previously been reviewed in detail and are sum-
`marized here [54]. Single-agent paclitaxel has
`shown limited activity in AIPC, with PSA re-
`sponse ranging from 0% to 39% and measurable
`disease responses ranging from 4% to 50% de-
`pending on the administration schedule [55,56].
`In these studies, weekly dosing provided better re-
`sponse rates than every-3-week administration, al-
`though 33% of patients experienced grade 3
`neuropathy [55].
`Based on the synergism observed in preclinical
`models [29], multiple phase II studies evaluated re-
`sponse rates of the paclitaxel/estramustine (P/E)
`doublet [35–38]. One study combined 96-hour
`paclitaxel infusion every 3 weeks with oral estra-
`mustine and reported a 53% PSA response rate
`without any grade 3 neuropathy [35]. This study
`provided evidence that optimized paclitaxel dos-
`ing combined with estramustine could produce
`meaningful
`responses without excess
`toxicity.
`Other trials using lower doses of weekly paclitaxel
`in combination with estramustine observed PSA
`response rates ranging 42% to 60% with signifi-
`cantly less grade 3 neuropathy than the original
`single-agent study [37,38]. Higher-dose weekly
`paclitaxel in combination with estramustine failed
`to improve outcome in one trial that reported
`a 62% PSA response accompanied by grade 3
`neuropathy in 21% of patients [36].
`Docetaxel is a semisynthetic taxane that has
`been shown to have greater preclinical activity than
`paclitaxel [57]. Single-agent phase II studies have
`been conducted using weekly and every-3-week
`dosing schedules. In two studies, every-3-week dos-
`ing resulted in PSA response rates of 38% to 46%,
`and one of the studies reported a median survival
`of 27 months [58,59]. Three trials examined weekly
`administration of docetaxel and reported PSA
`response rates ranging from 41% to 48%, with me-
`dian survival ranging from 9 to 20 months [60–62].
`
`In general, single-agent docetaxel phase II
`studies showed improved activity and toxicity
`profiles over single-agent paclitaxel. Because the
`addition of estramustine to paclitaxel improved
`response rates in phase I/II trials, the combination
`of estramustine and docetaxel has also been
`examined. One small trial based on a weekly
`dosing schedule reported a 72% PSA response
`rate with median survival of 16 months, which
`suggested that,
`like paclitaxel, the addition of
`estramustine to docetaxel may provide better
`response rates than docetaxel alone [63]. Grade
`1–3 neutropenia was noted to be 17% in this study.
`Alternatively, phase I/II D/E doublets based on
`every-3-week dosing have shown PSA responses
`of 45% to 74%, measurable soft tissue response
`rates from 20% to 57%, and a median survival
`reported in two trials to be 20 to 23 months
`[32,33,64–66]. Grade 3–4 neutropenia ranged
`from 56% to 75% in these studies. Although
`greater myelosuppression was observed in studies
`based on every-3-week docetaxel administration,
`rates of neutropenic fever were low. These early
`docetaxel trials seemed to favor every-3-week ad-
`ministration, and the superiority of every-3-week
`dosing was clearly established in the two pivotal
`phase III trials discussed below.
`
`SWOG 9916
`
`In October 2004, two randomized phase III
`trials concurrently reported a modest survival
`advantage
`from docetaxel-based therapy
`in
`AIPC [1,2]. These trials ushered in a new era of ex-
`citement for the use of cytotoxic regimens because
`they marked the first time any chemotherapeutic
`agent has demonstrated a survival advantage in
`this disease setting. SWOG 9916 [2] was the first
`of these trials that compared D/E against the pre-
`vious standard of care mitoxantrone and predni-
`sone (M/P). The significant activity of D/E
`noted in combination phase I/II trials mentioned
`previously served as the basis for this trial. Overall
`survival was the primary endpoint, and the trial
`was powered to detect a 33% survival improve-
`ment in the D/E arm over the M/P arm. Progres-
`sion-free survival, PSA, and measurable response
`rates were secondary endpoints. Patients were
`randomized to receive one of the following 21-
`day regimens: (1) estramustine 280 mg three times
`a day on days 1 through 5, docetaxel 60 mg/m2 on
`day 1 and 60 mg of dexamethasone over three di-
`vided doses starting the night before docetaxel;
`or (2) mitoxantrone 12 mg/m2 on day 1 plus
`
`

`

`230
`
`BURGESS & ROTH
`
`prednisone 5 mg twice daily. Doses were increased
`to docetaxel 70 mg/m2 and mitoxantrone 14 mg/m2
`on subsequent cycles if no grade 3–4 toxicities
`were noted in the initial cycle.
`Of the 674 eligible patients in the study, 338
`received D/E, and 336 received M/P. The median
`patient age was 70 years, and approximately 90%
`of both arms were SWOG performance status
`0 or 1. The median PSA (ng/ml) was 84 and 90 in
`the D/E and M/P arms, respectively. Bone pain
`less than grade 2 was reported in 64% of both
`arms. Although the study did not achieve the
`primary endpoint of demonstrating a 33% im-
`provement in survival between the two treatment
`arms, intention-to-treat analysis revealed a median
`survival of 17.5 months among patients receiving
`D/E and 15.6 months among patients receiving
`M/P (P ¼ .02). Along with the TAX 327 trial dis-
`cussed below, the 2-month median survival ad-
`vantage noted in this study represented the first
`survival benefit from chemotherapy in patients
`with AIPC. The hazard ratio for death in this trial
`was 0.8 (95% confidence interval, 0.67–0.97), so
`during the study, the risk of death was reduced
`by 20% in the docetaxel-treated group. Analysis
`of secondary endpoints further supported the
`advantage of D/E therapy: Median time to pro-
`gression was 6.3 months in the D/E arm versus
`3.2 months in the M/P arm (P !.001). PSA
`response rates were 50% in D/E group and 27%
`in M/P group (P ! .001). Partial response of mea-
`surable disease and subjective pain relief rates
`were not statistically different between the two
`treatment groups. Withdrawal rates for adverse
`events were 16% in the D/E arm and 10% in
`the M/P arm. Eight and four treatment-related
`deaths occurred in the D/E and M/P arms, respec-
`tively. Grade 3–4 neutropenia occurred in 12.5%
`versus 16.1% of patients in the M/P and D/E
`arms, respectively, although the difference was
`not statistically significant (P ¼ .22). The rate of
`severe neutropenia in patients treated with D/E
`was markedly lower than seen in the earlier-phase
`trials, and the rate of neutropenic fever was only
`5% in this group.
`The results from SWOG 9916 showed that
`combination of docetaxel and estramustine pro-
`longs median survival by 2 months and reduces
`the risk for death relative to mitoxantrone and
`prednisone. The median survival of 15.6 months in
`the control group was approximately 3.5 months
`longer than reported in earlier phase III trials that
`studied a similar regimen of mitoxantrone and
`prednisone [16,17]. Higher tumor burden in the
`
`patients of the earlier trials may account for this
`difference because the previously studied patients
`had symptomatic disease and slightly higher me-
`dian PSA values (150–158 ng/ml). Nonetheless,
`the SWOG 9916 trial definitively established that
`docetaxel-based therapy every 3 weeks prolongs
`median survival and reduces the risk of death.
`These results were complemented by the TAX
`327 trial and immediately set a new standard of
`care for first-line therapy in patients who have
`AIPC.
`
`TAX 327
`
`The TAX 327 trial, reported concurrently with
`SWOG 9916, was a randomized phase III study
`that compared dose-equivalent docetaxel given on
`a weekly basis or every 3 weeks with prednisone
`against mitoxantrone given every 3 weeks [1].
`Notable differences of this trial compared with
`SWOG 9916 include the addition of a weekly do-
`cetaxel arm and the absence of estramustine in the
`docetaxel arms. As in the SWOG trial, the pri-
`mary endpoint of this study was to detect an over-
`all
`survival advantage in the docetaxel arm
`compared with the mitoxantrone control arm.
`Secondary endpoints in this trial differed slightly
`and included measurement of pain levels, quality
`of life (measured by the FACT-P questionnaire),
`PSA, and measurable soft tissue responses. Like
`the SWOG trial, inclusion criteria required pro-
`gressive AIPC measured radiographically and
`biochemically, ongoing androgen-ablation ther-
`apy, and prior cessation of antiandrogen ther-
`apy. Patients were randomized to receive one of
`the following treatment regimens: (1) docetaxel
`75 mg/m2 every 3 weeks (D/P), (2) docetaxel
`30 mg/m2 every week (wD/P), or (3) mitoxantrone
`12 mg/m2 every 3 weeks (M/P). All patients
`received prednisone 5 mg twice daily, and patients
`receiving docetaxel were premedicated with dexa-
`methasone. The impact of dexamethasone on
`treatment response in the docetaxel arms has
`previously been suggested not to contribute to
`the treatment response [67].
`Of the 1006 patients enrolled in the trial, 332,
`330, and 335 were treated with D/P, wD/P, and
`M/P, respectively. In each of the three arms, the
`median patient age was 68 to 69 years, and 12%
`to 14% of patients had Karnofsky performance
`status !80%. The median serum PSA value in
`the three treatment arms ranged from 108 to
`123 ng/ml, and approximately 45% of patients
`had pain. Ninety percent of patients had known
`
`

`

`CHEMOTHERAPY IN ADVANCED PROSTATE CANCER
`
`231
`
`bone metastases, and 40% had measurable soft-
`tissue lesions. Based on intention-to-treat analysis,
`the median durations of
`survival were 18.9
`months in the every-3-week docetaxel arm, 17.4
`months in the weekly docetaxel arm, and 16.5
`months in the mitoxantrone arm. Only the every-
`3-week docetaxel regimen was demonstrated to
`have a statistically significant survival benefit
`compared with mitoxantrone. The hazard ratio
`for death using this regimen was 0.76 (95%
`confidence interval, 0.62–0.94), which confirmed
`the mortality reduction seen in the SWOG trial
`with every-3-week docetaxel/estramustine. Analy-
`sis of secondary endpoints showed reduced pain in
`35% (P ¼ .01), 31% (P ¼ .08), and 22% of pa-
`tients treated with D/P, wD/P, and M/P, respec-
`tively. Quality of life improvement was noted in
`22% (P ¼ .009), 23% (P ¼ .005), and 13% of pa-
`tients treated with D/P, wD/P, and M/P, respec-
`tively. D/P provided superior palliation relative
`to the prior standard M/P, whereas, pain reduc-
`tion in patients treated with weekly D/P was not
`different from those treated with M/P. Based on
`these results, the use of weekly D/P for palliative
`intent only may be appropriate if the toxicity
`profile precludes use of every-3-week docetaxel.
`However, based on the survival advantage, every-
`3-week docetaxel is the preferred first-line standard
`regimen. The PSA response to treatment was
`45% (P ! .001) in the D/P, 48% (P ! .001) in
`the wD/P, and 32% in the M/P arms (see below).
`Grade 3–4 neutropenia was seen in 32% (P %
`.05) of patients treated with D/P, in 2% (P %
`.0015) of patients treated with wD/P, and in 35%
`of patients treated with M/P, although only 3%
`of D/P-treated patients had febrile neutropenia.
`Common adverse events with D/P that occurred
`more frequently with every-3-week administration
`included fatigue, diarrhea, alopecia, and neuropa-
`thy. Three of five treatment-related deaths occurred
`in the mitoxantrone group.
`The results of TAX 327 confirmed the findings
`of SWOG 9916 by demonstrating a 2-month
`survival advantage and a 20% mortality reduction
`over the study period in patients who had AIPC
`treated with docetaxel every 3 weeks. Weekly
`docetaxel was not associated with a statistically
`significant survival advantage, although this regi-
`men demonstrated pain reduction equivalent to
`the historical standard of mitoxantrone and pred-
`nisone. Every-3-week docetaxel was accompanied
`by a slightly worse toxicity profile than mitoxan-
`trone and prednisone, although treatment-related
`deaths were essentially the same in all arms. The
`
`survival advantage associated with every-3-week
`docetaxel reported in the TAX 327 and SWOG
`9916 trials represent independent confirmation
`from two large, randomized phase III trials that
`cytotoxic chemotherapy with an acceptable toxic-
`ity profile can prolong life in patients with AIPC.
`These two trials provide new excitement for the use
`of cytotoxic chemotherapy in patients who have
`AIPC and establish a foundation on which to
`develop improved regimens. The urologist will
`continue to play a critical role in defining better
`cytotoxic regimens by early referral to medical
`oncologists for participation in clinical
`trials.
`Although these trials report a modest survival
`improvement, they herald greater potential activ-
`ity with future regimens. Many trials have been
`designed based on the results of TAX 327 and
`SWOG 9916, and expectations for future survival
`gains in AIPC are optimistic.
`
`PSA response as surrogate marker
`for clinical benefit
`
`Post-treatment serum PSA decline of at least
`50% of pretreatment baseline has been suggested
`as a surrogate marker of survival benefit in phase
`II trials [9,10,68,69]. One interesting observation
`from the TAX 327 trial is the failure of PSA re-
`sponse to predict the proven survival advantage.
`Only 45% of patients in the every-3-week doce-
`taxel arm experienced a post-therapy PSA decline
`of at least 50% despite a 2.4-month prolongation
`of survival. Follow-up analysis by the TAX 327
`investigators further confirmed that the PSA re-
`sponse only partly explained the observed survival
`benefit [70]. Because 50% of patients treated with
`D/E in SWOG trial had at least a 50% reduction
`in post-treatment PSA, these results suggest that
`the addition of estramustine serves only to im-
`prove PSA response rates without additive sur-
`vival benefit. This suggestion is supported by
`a recent randomized phase II study comparing
`every-3-week docetaxel with docetaxel plus estra-
`mustine that showed a PSA response in 68% of
`patients treated with D/E compared with only
`29% of those treated with docetaxel alone [71].
`Based on PSA response, results from this phase
`II trial imply that every-3-week D/E should have
`far greater clinical benefit than docetaxel alone
`and that every-3-week docetaxel alone should
`not produce any survival benefit. The results of
`TAX 327 prove otherwise and highlight an impor-
`tant limitation with the use of post-treatment PSA
`decline as a surrogate marker in phase II studies.
`
`

`

`232
`
`BURGESS & ROTH
`
`The TAX 327 trial showed that PSA response
`lacks sufficient sensitivity as a marker of survival
`benefit in phase II trials to exclude drugs from ad-
`ditional phase III testing. Because a better phase
`II surrogate marker does not exist, the use of
`post-treatment PSA response should not be aban-
`doned in future trials at this time. However, this
`marker should not serve as the sole criteria for ex-
`cluding drugs from further phase III analysis.
`
`Future directions in AIPC
`
`Current and future clinical trials in AIPC are
`based on two fundamental paradigms. The search
`for novel agents with improved activity continues,
`and such agents have been recently reviewed [72].
`A larger number of trials seek to use docetaxel as
`a component of doublet therapy in combination
`with an investigational agent. An account of all
`docetaxel-based combinations under investigation
`is beyond the scope of this article; however, a few
`examples are mentioned. Bevacizumab is an anti-
`vascular endothelial growth factor monoclonal
`antibody that has shown synergistic activity in
`combination with traditional cytotoxic agents in
`other solid tumors [73,74]. Encouraging phase II
`data from patients treated with docetaxel, estra-
`mustine, and bevacizumab [75] led to the creation
`of cooperative group CALGB 90401 trial, which
`is an open phase III protocol randomizing partic-
`ipants to docetaxel/prednisone with or without
`bevacizumab. Synergy between docetaxel and
`antiangiogenic therapies has been further sug-
`gested by results from a phase II trial of docetaxel
`and thalidomide in which 51% of patients receiv-
`ing combined treatment had a PSA response ver-
`sus 37% of patients receiving docetaxel alone [76].
`The addition of high-dose calcitriol to docetaxel
`has recently garnered much interest based on fa-
`vorable preclinical studies and phase II results of
`weekly docetaxel plus high-dose pulse calcitriol
`[77,78]. These data led to the creation of the ran-
`domized, double-blind, placebo-controlled AS-
`CENT trial, which asked whether calcitriol adds
`to the effect of weekly docetaxel on serum PSA re-
`sponse [79]. Recent analysis of the trial showed
`a failure to achieve its primary goal [80]. Altho-
`ugh the data indicated a trend toward improved
`survival, the study was not powered to detect
`a survival advantage. Additional studies have
`examined the impact of calcitriol on alternate
`docetaxel-based regimens [51], although no benefit
`has been confirmed in a randomized phase III
`trial, so clinical use should be limited to the
`
`investigational setting. Given the large number
`of open trials seeking to improve docetaxel-based
`therapy with novel combinations, oncologists are
`likely to have multiple active options for the treat-
`ment of AIPC in the coming years.
`
`Chemotherapy in the nonmetastatic setting
`
`Strong interest lies in whether earlier stages of
`disease ranging from the neoadjuvant setting to
`biochemical failure can gain survival benefit from
`cytotoxic therapy. Multiple pilot studies have
`shown that docetaxel-based therapy is safe and
`active in earlier stages of disease [81–87]. These
`and other trials have served as the basis for larger
`randomized phase III trials seeking to define the
`activity of chemotherapy in earlier stages of dis-
`ease. For example, the adjuvant TAX 3501 (AT-
`LAS) trial scheduled to open this fall is a large
`international phase III trial planning to enroll
`over 2000 patients with high risk for relapse after
`radical prostatectomy. Patients with a predicted
`risk of biochemical failure within 3 years of sur-
`gery will be randomized to receive 18 months of
`leuprolide with or without docetaxel or receive de-
`ferred treatment after radical prostatectomy. The
`primary endpoint of the trial is progression-free
`survival, and secondary endpoints include overall
`survival and cancer-specific survival.
`Progress in the nonmetastatic setting has been
`hindered by poor accrual to the large, cooperative
`group studies, which has resulted in the premature
`termination of the RTOG 0014 and ECOG 1899
`trials. These two phase III trials were designed to
`examine the role of chemotherapy in the setting of
`PSA relapse after local therapy but only enrolled
`2% of target accrual. The anemic participation
`recently seen in the large cooperative group trials
`has been a subject of much debate [88]. It is unfor-
`tunate that lack of physician interest has plagued
`the nonmetastatic trials because the pilot studies
`have shown tolerable drug activity and raise critical
`clinical questions that require large phase III trials
`for proper answers. As patients become more savvy
`and seek other therapies for earlier-stage disease,
`medical and urologic oncologists have an unprece-
`dented need to communicate and to facilitate
`enrollment in clinical trials to obtain the much-
`needed answers to pressing clinical questions.
`
`Summary
`
`This article summarizes the historical perspec-
`tive of clinical trials in androgen independent
`
`

`

`CHEMOTHERAPY IN ADVANCED PROSTATE CANCER
`
`233
`
`prostate cancer. In October of 2004, the results
`from TAX 327 and SWOG 9916, two large,
`randomized, phase III trials, independently con-
`firmed that every-3-week docetaxel-based therapy
`produces a modest survival benefit and immedi-
`ately established docetaxel as first-line standard
`treatment. These trials reported the first survival
`benefit from cytotoxic therapy seen in the treat-
`ment of AIPC and led to the FDA approval of
`docetaxel in this setting. Multiple clinical trials are
`ongoing in attempts to extend the survival benefits
`gained from docetaxel in AIPC and to define the
`role of cytotoxic therapy in earlier stages of
`disease. Improved collaboration between medical
`and urologic oncologists is essential to facilitate
`clinical trial enrollment so that answers can be
`learned to important clinical questions regarding
`optimal management of all stages of disease.
`
`References
`
`[1] Tannock IF, de Wit R, Berry WR, et al. Docetaxel
`plus prednisone or mitoxantrone plus prednisone
`for advanced prostate cancer. N Engl J Med 2004;
`351:1502–12.
`[2] Petrylak DP, Tangen CM, Hussain MH, et al. Doce-
`taxel and estramustine compared with mitoxantrone
`and prednisone for advanced refractory prostate
`cancer. N Engl J Med 2004;351:1513–20.
`[3] Elder JS, Gibbons RP. Results of trials of the USA
`National Prostatic Cancer Project. Prog Clin Biol
`Res 1985;185A:221–42.
`[4] Eisenberger MA, Simon R, O’Dwyer PJ, et al. A
`reevaluation of nonhormonal cytotoxic chemother-
`apy in the treatment of prostatic carcinoma. J Clin
`Oncol 1985;3:827–41.
`[5] Tannock IF. Is there evidence that chemotherapy is
`of benefit to patients with carcinoma of the prostate?
`J Clin Oncol 1985;3:1013–21.
`[6] Yagoda A, Petrylak D. Cytotoxic chemotherapy for
`advanced hormone-resistant prostate cancer. Can-
`cer 1993;71(Suppl):1098–109.
`[7] Murphy GP, Slack NH. Response criteria for the
`prostate of the USA National Prostatic Cancer Pro-
`ject. Prostate 1980;1:375–82.
`[8] Slack NH, Brady MF, Murphy GP. A reexamina-
`tion of the stable category for evaluating response
`in patients with advanced prostate cancer. Cancer
`1984;54:564–74.
`[9] Kelly WK, Scher HI, Mazumdar M, et al. Prostate-
`specific antigen as a measure of disease outcome in
`metastatic hormone-refractory prostate
`cancer.
`J Clin Oncol 1993;11:607–15.
`[10] Bubley GJ, Carducci M, Dahut W, et al. Eligibil-
`ity and response guidelines for phase II clinical
`trials in androgen-independent prostate cancer:
`recommendations
`from the
`Prostate-Specific
`
`Antigen Working Group. J Clin Oncol 1999;17:
`3461–7.
`[11] Tannock I, Gospodarowicz M, Meakin W, et al.
`Treatment of metastatic prostatic cancer with low-
`dose prednisone: evaluation of pain and quality of
`life as pragmatic indices of response. J Clin Oncol
`1989;7:590–7.
`[12] Osborne CK, Drelichman A, Von Hoff DD, et al.
`Mitoxantrone: modest activity in a phase II trial in
`advanced prostate cancer. Cancer Treat Rep 1983;
`67:1133–5.
`[13] Koeller J, Eble M. Mitoxantrone: a novel anthracy-
`cline derivative. Clin Pharm 1988;7:574–81.
`[14] Moore MJ, Osoba D, Murphy K, et al. Use of palli-
`ative end points to evaluate the effects of mitoxan-
`trone and low-dose prednisone in patients with
`hormonally resistant prostate cancer. J Clin Oncol
`1994;12:689–94.
`[15] Osoba D, Tannock IF, Ernst DS, Neville AJ.
`Health-related quality of life in men with metastatic
`prostate cancer treated with prednisone alone or
`mitoxantrone and prednisone. J Clin Oncol 1999;
`17:1654–63.
`[16] Tannock IF, Osoba D, Stockler MR, et al. Chemo-
`therapy with mitoxantrone plus prednisone or pred-
`nisone alone for symptomatic hormone-resistant
`prostate cancer: a Canadian randomized trial with
`palliative end points. J Clin Oncol 1996;14:1756–64.
`[17] Kantoff PW, Halabi S, Conaway M, et al. Hydro-
`cortisone with or without mitoxantrone in men
`with hormone-refractory prostate cancer: results of
`the cancer and leukemia group B 9182 study.
`J Clin Oncol 1999;17:2506–13.
`[18] Berry W, Dakhil S, Modiano M, et al. Phase III
`study of mitoxantrone plus low dose prednisone ver-
`sus low dose prednisone alone in patients with
`asymptomatic hormone refractory prostate cancer.
`J Urol 2002;168:2439–43.
`[19] Samelis GF, Kalofonos H, Adamou A, et al. The
`combination of estramustine, vinorelbine, and
`mitoxantrone
`in
`hormone-refractory
`prostate
`cancer: a Phase II feasibility study conducted by
`the Hellenic Cooperative Oncology Group. Urology
`20

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