throbber
Novel cytotoxic and biological agents for prostate cancer: Where
`will the money be in 2005?
`
`John M. Strother a, Tomasz M. Beer a,*, Robert Dreicer b
`
`a Division of Hematology and Medical Oncology, Oregon Health and Science University, Mail Code CR-145,
`3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
`b Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
`
`Received 18 August 2004; received in revised form 7 February 2005; accepted 8 February 2005
`Available online 10 March 2005
`
`Abstract
`
`In 2004, docetaxel-based chemotherapy became the first treatment capable of extending life in androgen-independent prostate
`cancer. The era of therapeutic nihilism in this disease has thus been put to rest and a broad range of agents is being tested with
`the goal of improving on the successes of 2004. Lessons learned from other tumour types will need to be applied to prostate cancer
`in order to harness the bounty of available ideas. Target amplification or activating mutations and not merely the presence of a
`target are likely to be important to the success of targeted agents. Thus, the promise of the current crop of targeted agents is most
`likely to be realised when pursued in the context of well-credentialed targets and tested in highly translational clinical trials that are
`capable not only of assessing tumour response, but also of evaluating the status of the targeted pathway. The most promising agents
`in clinical development are reviewed.
`Ó 2005 Elsevier Ltd. All rights reserved.
`
`Keywords: Prostate cancer; Chemotherapy; Targeted therapy; Novel agents
`
`1. Introduction
`
`2. Current cytotoxic agents
`
`In 2004, docetaxel became the first drug to prolong sur-
`vival of patients with androgen-independent prostate
`cancer (AIPC) and the long-held view that AIPC is an
`untreatable disease was put to rest. The advance seen with
`docetaxel is critically important because it demonstrates
`that progress in this disease is possible and it establishes
`a clear standard of care upon which future studies will
`be built. A substantial number of investigational agents,
`belonging to a wide range of drug classes and targeting
`a broad range of cancer pathways are in clinical develop-
`ment. Thus, the future has never been brighter in treating
`this disease, which is the second-leading cancer killer of
`men in the United States of America (USA) and in 2000
`was the cause of more than 200,000 deaths worldwide.
`
`0959-8049/$ - see front matter Ó 2005 Elsevier Ltd. All rights reserved.
`
`The range of currently available cytotoxic agents has
`been summarised in detail by Bhandari, Petrylak and
`Hussain elsewhere in this volume. Active agents include
`mitoxantrone, the taxanes (paclitaxel and docetaxel,
`which seem to illustrate some schedule dependency)
`and estramustine [1–24]. The level of reported anti-can-
`cer efficacy has depended somewhat on the case selection
`characteristics, the nature of ÔresponseÕ (subjective or
`objective) and the use of surrogate markers of response.
`
`3. New cytotoxic agents
`
`3.1. SB-715992
`
`Mitotic kinesins, which play essential roles in the
`assembly and function of
`the mitotic spindle, are
`
`JANSSEN EXHIBIT 2032
`Mylan v. Janssen IPR2016-01332
`
`

`

`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`955
`
`expressed preferentially in neoplastic cells. They repre-
`sent novel targets for cancer treatment [25]. Inhibition
`of the mitotic kinesin spindle protein (KSP) results in
`formation of monopolar spindles within single cells
`and the induction of apoptosis in pre-clinical studies.
`SB-715992 is the first KSP inhibitor to enter clinical tri-
`als and has a broad spectrum of activity in pre-clinical
`models of cancer, including models that are refractory
`to chemotherapy and prostate cancer models. This agent
`has completed phase I evaluation and is entering phase
`II trials. The Southwest Oncology Group (SWOG) will
`be evaluating this agent in patients who have progressed
`on docetaxel-containing therapy.
`
`3.2. Epothilones
`
`The epothilones are a new class of cytotoxic agents
`with anti-neoplastic activity in pre-clinical models of a
`range of tumours insensitive or resistant to paclitaxel;
`their mechanism of action is microtubular stabilisation
`resulting in mitotic arrest [26]. EPO906 is a novel epothi-
`lone that is not a substrate for multidrug-resistance pro-
`tein. A phase IIa trial of weekly EPO906 in 37 patients
`with previously-treated AIPC found that it was well tol-
`erated; the most common adverse events were gastroin-
`testinal. Twenty-two percent of patients responded by
`prostate-specific antigen (PSA) criteria and 4 out of 20
`patients with measurable disease responded [27]. Two
`recent phase II studies demonstrate that the epothilone
`analogue BMS-247550 has activity in chemotherapy-
`naı¨ve patients with metastatic AIPC. SWOG 0111
`showed 41% PSA and 30% measurable disease response
`rates in patients with chemotherapy-naı¨ve AIPC [28].
`Another multi-institutional phase II study reported
`56% PSA and 23% measurable disease response rates
`for BMS-247550 alone and 69% PSA and 44% measur-
`able disease response rates for the combination of BMS-
`247550 with estramustine [29]. The most frequent grade
`3 toxicities in both of these studies were fatigue and sen-
`sory neuropathy. These studies demonstrate activity for
`this agent, although its place in the treatment of AIPC is
`unclear in light of current docetaxel results. With the
`establishment of docetaxel-based chemotherapy as
`front-line therapy, suggestive evidence that there is some
`degree of non-cross-resistance with epothilones becomes
`increasingly relevant [30].
`
`3.3. Satraplatin
`
`Satraplatin is a novel oral platinum complex that has
`shown activity against AIPC in cisplatin-resistant human
`tumour lines and in phase I trials. Satraplatin plus pred-
`nisone was compared with prednisone alone in 50 chemo-
`therapy-naı¨ve AIPC patients. The satraplatin arm had a
`better progression-free survival (median 5.2 months
`versus 2.5 months, P = 0.023) and a higher frequency of
`
`PSA response (33.3% versus 8.7%; P = 0.046) [31]. A
`phase III trial of satraplatin plus prednisone versus
`prednisone as second-line chemotherapy is underway.
`
`3.4. Amonafide
`
`Amonafide is a synthetic imide anti-neoplastic agent
`with DNA intercalative properties that demonstrated
`significant activity in pre-clinical studies and some activ-
`ity in phase I trials, including at least one partial re-
`sponse in a patient with prostate cancer [32]. The drug
`is extensively metabolised and detected in plasma and
`urine. Its toxicity has previously been correlated to the
`formation of an active metabolite, N-acetyl-amonafide.
`In phase I studies using various administration sched-
`ules of amonafide, myelosuppression was the dose limit-
`ing toxicity. A phase II trial of amonafide at a dose of
`225 mg/m2 i.v. daily for 5 d was conducted by SWOG.
`Forty-three evaluable patients with measurable AIPC
`were treated. The most common toxicities were haema-
`tological including leucopaenia (72%), granulocytopae-
`nia (32.6%) and thrombocytopaenia (44.2%). There
`were no complete responses and 5 partial responses, giv-
`ing an overall response rate of 12% [33].
`Subsequent work determined that N-acetyltransfer-
`ase enzyme polymorphisms play an important role in
`the metabolism of amonafide (fast acetylators are at
`great risk of haematological toxicity from amonafide
`therapy). Ratain and colleagues [34] proposed a phar-
`macodynamic model based on acetylator phenotype
`using caffeine as a probe to optimise amonafide dosing.
`Observations from a phase II trial of amonafide in pa-
`tients with metastatic breast cancer that response rates
`appeared higher in patients experiencing more severe
`myelosuppression led to the hypothesis that optimisa-
`tion of amonafide dosing might yield evidence of higher
`anti-tumour activity [35]. Using a modification of Ra-
`tainÕs pharmacodynamic model, a phase I/II trial of
`NAT2 phenotype-based dosing of amonafide is ongoing
`in patients with metastatic AIPC treated with up to one
`prior systemic chemotherapy regimen.
`
`4. Nuclear receptor ligands
`
`4.1. Peroxisome proliferators-activated receptor gamma
`(PPARc) ligands
`
`Peroxisome proliferators-activated receptor gamma
`(PPARc) ligands are members of the nuclear receptor
`superfamily of ligand-dependent transcription factors
`and play a central role in lipid metabolism and adipo-
`cyte regulation. PPARs are widely expressed by prostate
`cancer cells. Synthetic PPARc agonists inhibit prostate
`cancer growth in vitro [36]. The thiazolidinediones
`(including troglitazone, rosiglitazone and piaglitazone)
`
`

`

`956
`
`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`are a class of synthetic ligands for PPARc that have
`been developed primarily for the treatment of insulin-
`resistant type II diabetes mellitus. A phase II study of
`troglitazone in patients with asymptomatic advanced
`prostate cancer included 12 men with androgen-depen-
`dent disease and 29 men with AIPC. In the androgen-
`dependent group, 1 of 12 patients had a >50% decrease
`in PSA and 3 of 12 patients had a <50% decrease. In the
`AIPC group, no patient had a >50% decrease in PSA
`but 4 of 29 patients had a <50% decrease [37]. A second
`randomised, placebo-controlled trial of rosiglitazone
`was initiated. A total of 105 men with rising PSA after
`local therapy were randomised with a primary end-point
`of post-treatment
`change
`in PSA doubling time
`(PSADT). There was no difference between the 2 arms
`in the primary efficacy analysis. Complicating analysis
`was the higher-than-expected (40%) response rate in
`the placebo arm [38,39]. Given these results, it is unlikely
`that PPARc ligands alone will play a role in prostate
`cancer therapy. However, as the role of this receptor
`in prostate cancer progression is better understood,
`rationally designed combination of drugs that include
`PPARc ligands may emerge.
`
`5. Calcitriol
`
`Calcitriol, the most active metabolite of vitamin D,
`has significant anti-neoplastic activity in a broad range
`of pre-clinical cancer models including in vitro and
`in vivo activity in several models of prostate cancer
`[40–46]. Several mechanisms of activity have been pro-
`posed. These include inhibition of proliferation associ-
`ated with cell cycle arrest and,
`in some models,
`differentiation, reduction in invasiveness and angiogene-
`sis, and induction of apoptosis. Further synergistic and/
`or additive effects with cytotoxic chemotherapy, radia-
`tion and other cancer drugs have been reported. A con-
`temporary comprehensive review of
`spectrum and
`mechanisms of activity is available [47].
`Significantly supraphysiological concentrations of
`calcitriol are required for anti-neoplastic effects. The
`development of high-dose regimens, made possible by
`intermittent dosing of calcitriol [48,49], has led to several
`clinical trials in prostate cancer. Single agent weekly cal-
`citriol in hormone-naı¨ve prostate cancer appeared to
`lengthen PSADT in an uncontrolled study [50]. Weekly
`calcitriol yielded encouraging phase II results when
`added to docetaxel. Eighty-one percent of 37 patients
`had confirmed >50% reduction in serum PSA in a single
`institution trial [51]. These results were encouraging rel-
`ative to historical experience with single agent docetaxel
`and led to the development of a randomised trial of
`docetaxel with calcitriol or placebo. This trial (dubbed
`ASCENT) is using DN-101, a highly concentrated for-
`mulation of calcitriol developed specifically for oncol-
`
`ogy. ASCENT accrual has been completed and results
`should provide robust information about the potential
`of calcitriol to enhance chemotherapy for prostate can-
`cer. Studies of calcitriol with dexamethasone, paclitaxel
`and carboplatin are underway as well [52].
`The development of calcitriol analogues that might
`have anti-neoplastic activity but cause less hypercalca-
`emia has been proposed as another strategy to target
`the vitamin D receptor for cancer treatment. Several
`hundred such analogues have been synthesised [53–55].
`Several analogues, dosed daily, have entered clinical tri-
`als. Two of 25 AIPC patients had objective partial re-
`sponses to 1-alpha-hydroxyvitamin D2 [56]. Further
`studies of calcitriol analogues in prostate cancer are
`underway.
`
`6. Growth factor receptor antagonists
`
`6.1. Epidermal growth factor receptor
`
`The epidermal growth factor (EGF) system has been
`implicated as one of the mitogenic signals that may drive
`progression of AIPC. EGF receptor (EGFR) signalling
`pathways may be involved in prostate cancer invasion
`and angiogenesis [57,58]. Cetuximab is an anti-EGFR
`monoclonal antibody with high affinity that prevents
`activation of receptor tyrosine kinase. In pre-clinical
`studies cetuximab has demonstrated activity in a PC-
`3M-LN4 orthotopic model of AIPC, both as a single
`agent and in combination with paclitaxel [59]. A phase
`Ib/IIa study of cetuximab and doxorubicin in 18 patients
`with AIPC documented stabilisation in PSA for a med-
`ian of 4 months in 5 patients (28%) [60]. Gefitinib (Ires-
`sa) is an oral small molecule EGFR tyrosine kinase
`inhibitor that has demonstrated clinical utility in non-
`small cell lung cancer [59,61]. However, a phase II trial
`of gefitinib in AIPC has preliminarily found infrequent
`PSA responses and early progression in many patients
`[62]. Thus the EGF receptor is a target that has yet to
`be vetted in a clinical trial and its role in the future of
`prostate cancer therapy remains to be determined.
`
`6.2. HER-2
`
`Trastuzumab is a humanised monoclonal antibody
`that targets the extracellular domain of HER-2 and con-
`fers a survival benefit when combined with chemother-
`apy in patients with metastatic breast carcinoma that
`overexpresses HER-2 [63]. Subsequent analyses have
`suggested that most of the activity of this agent is seen
`in patients with HER-2 gene amplification and fluores-
`cent in situ hybridisation has largely replaced immuno-
`histochemistry as a tool for identifying breast cancer
`patients who are likely to benefit from trastuzumab.
`HER-2 expression in prostate cancer, as determined by
`
`

`

`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`957
`
`immunohistochemistry, has produced very heteroge-
`neous results [64,65]. In one series of 70 patients with
`metastatic disease, the overall rate of prostatic-tissue
`sections presenting positive HER-2 immunostaining
`was 64.3% [66] and in another study all 39 patients with
`prostate cancer showed positive staining by immunohis-
`tochemistry [67]. Conversely, two other series identified
`HER-2 positivity in 40% (32 of 79) and 0% (0 of 40) of
`patients with prostate cancer [68,69]. A phase II trial in
`patients with progressive prostate cancer found that
`trastuzumab alone produced no responses in 23 patients
`with either HER-2 negative (17 patients) or HER-2 po-
`sitive (6 patients) disease as determined by immunohis-
`tochemistry. Additionally, four patients with HER-2
`positive disease received combination therapy with trast-
`uzumab and paclitaxel;
`two patients had a partial
`response by PSA and two patients had disease progres-
`sion [65]. A second phase II trial of trastuzumab and
`docetaxel in HRPC was closed early when only 7/72 pa-
`tients were found to have HER-2 overexpression by
`immunohistochemistry and none of the 4 patients trea-
`ted with trastuzumab alone demonstrated response
`[64]. Thus it appears that trastuzumab is unlikely to gain
`a foothold in prostate cancer therapy.
`
`6.3. PDGF receptor
`
`Platelet-derived growth factor (PDGF) and its recep-
`tor (PDGF-R) have been implicated as both paracrine
`and autocrine mediators of prostate cancer progression
`[70,71]. The PDGF-R is expressed in 80% of AIPC le-
`sions [72]. Imatinib (Gleevec) is a specific inhibitor of
`the BCR-ABL, c-kit and PDGF receptor tyrosine
`kinases. In a mouse model of prostate cancer bone
`metastases, PDGF-R inhibition by imatinib resulted in
`preservation of bone structure,
`significant
`tumour
`growth inhibition, a significant increase in apoptosis of
`malignant cells and a decrease in lymph node metastases
`[61]. Therefore, PDGF-R would appear to be a promis-
`ing target in prostate cancer therapy. A small study of 15
`patients with AIPC and painful bony metastases treated
`with imatinib and zoledronic acid failed to demonstrate
`PSA response and had no palliative or clinical activity
`[73]. A phase II trial of imatinib as a single agent in 17
`patients with hormone-naı¨ve PSA progression after lo-
`cal therapy demonstrated biochemical stable disease in
`6 patients (35%) [74]. At the present time, it is not clear
`whether this class of agents will prove useful. Although
`little single agent activity was seen, imatinib may prove
`more useful in combination therapy. A study at MD
`Anderson is examining imatinib in combination with
`docetaxel. As with all targeted therapies,
`it will be
`important that future studies include determination of
`PDGF-R expression whenever feasible, so that clinical
`outcomes can be interpreted in the context of knowledge
`about the status of the target.
`
`6.4. IGF receptor
`
`Interest in the IGF system and prostate cancer was
`stimulated by epidemiological investigations that sug-
`gest a relationship between elevated IGF-1 levels and
`prostate cancer risk [75,76] and recognition that signal-
`ling through the IGF-1 receptor inhibits apoptosis. Tar-
`geting the IGF signalling system is therefore a promising
`strategy in prostate cancer prevention and therapy.
`Agents that target the IGF system are in pre-clinical
`development.
`
`7. Anti-angiogenic agents
`
`7.1. Thalidomide
`
`As is the case for most solid tumours, the recruit-
`ment of blood vessels is an important step in progres-
`sion and metastasis of prostate cancer. Inhibition of
`this complex process is an attractive approach to treat-
`ment and many anti-angiogenic agents are currently in
`clinical development. Thalidomide is a sedative, anti-
`inflammatory
`and immunosuppressive
`agent
`that
`blocks
`the activity of angiogenic agents
`including
`bFGF, VEGF and IL-6. The limb defects attributed
`to its teratogenic effects have been postulated to be
`secondary to an inhibition of blood vessel growth in
`the developing foetal limb bud. A recent clinical trial
`of thalidomide for AIPC randomised 63 patients to a
`low-dose arm of 200 mg daily or a high-dose arm,
`escalating to the highest tolerated dose (up to 1200
`mg) [77]. Only 13 patients were treated on the high
`dose arm that was terminated early due to a combina-
`tion of lack of efficacy and poor tolerance. In all, 9
`patients (15%) showed a PSA decline of over 50%,
`all of whom were in the low-dose arm. A second phase
`II study of low-dose thalidomide in 20 patients with
`AIPC identified 3 men (15%) with a decline in serum
`PSA of at least 50% [78]. A randomised phase II trial
`of docetaxel with or without thalidomide 200 mg daily
`in 53 patients with chemotherapy-naı¨ve AIPC reported
`that 19 of 36 patients (53%) in the combination arm
`and 6 of 17 (35%) receiving docetaxel alone had a
`PSA decrease of at least 50%. Combination therapy
`also resulted in improved progression-free survival
`(5.9 versus 3.7 months) and overall survival at 18
`months (68.2% versus 42.9%) [79]. Therapy was rela-
`tively well tolerated, although 12 of the first 43 pa-
`tients in the combined treatment group developed
`thromboembolic events; all subsequent patients re-
`ceived prophylactic anticoagulation with low molecular
`weight heparin and no further thromboembolic events
`were noted. Thus thalidomide appears to have modest
`single-agent activity and its activity with docetaxel
`appears additive.
`
`

`

`958
`
`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`7.2. Lenalidomide
`
`CC-5013 (lenalidomide, Revlimid) is an immuno-
`modulatory thalidomide analogue, which in animal
`models is both anti-angiogenic and non-teratogenic [80].
`A phase I trial in patients with melanoma and other
`solid tumours provided evidence that CC-5013 can
`boost Th1-type cellular immunity. Figg and colleagues
`are currently conducting a phase I trial of CC-5013 in
`AIPC patients [81].
`
`7.3. Bevacizumab
`
`Bevacizumab (Avastin) is a recombinant, humanised
`anti-vascular endothelial growth factor (VEGF) mono-
`clonal antibody that blocks the binding of VEGF to
`its receptors. Recently approved in the United States
`for first-line use in metastatic colorectal cancer, bev-
`acizumab is also being examined in prostate cancer.
`CALGB 90006 is an ongoing phase II trial of bev-
`acizumab, docetaxel and estramustine in patients with
`AIPC. Initial results include a confirmed >50% decline
`in PSA in 13 of 20 patients (65%) with sufficient data
`and a partial response in 9 of 17 patients (53%) with
`measurable disease [82].
`
`7.4. Other
`
`PTK787/ZK 222584 is an oral angiogenesis antago-
`nist that inhibits all known VEGF receptors. A phase
`I study in 23 patients with AIPC showed that 3 (13%)
`had >40% reductions in PSA [83]. A recent randomised
`phase II trial of two doses of the matrix metalloprotein-
`ase inhibitor BMS-275291 in 80 patients with AIPC re-
`sulted in no responders [84].
`
`8. Proapoptotic agents
`
`8.1. Bcl-2 antisense
`
`The mitochondrial-associated protein Bcl-2 confers
`resistance to apoptosis and is overexpressed in AIPC
`[85,86]. G3139 (oblimersen sodium) is an 18-mer phosp-
`horothioate antisense oligonucleotide directed to the
`first six codons of the initiating sequence of the human
`bcl-2 mRNA [87]. In pre-clinical prostate cancer models,
`Bcl-2 antisense oligonucleotides have inhibited expres-
`sion of Bcl-2, delayed development of androgen inde-
`pendence and enhanced the effects of chemotherapy
`[88–91]. A phase II study of G3139 and docetaxel in
`31 men with metastatic AIPC (8 had received prior che-
`motherapy) yielded a confirmed >50% PSA reduction in
`15 patients (48%) and a PR in 4 out of 15 patients (27%)
`with measurable disease. Toxicities included grade 3–4
`neutropaenia in 42% of patients [92]. As this was a small
`
`study, it is difficult to determine whether this level of
`activity represents an improvement from that expected
`with docetaxel alone.
`
`8.2. Selective apoptotic anti-neoplastic drugs (SAANDs)
`
`Exisulind (sulindac sulphone), the oxidative metabo-
`lite of sulindac, is a member of a class of novel drugs
`that inhibit growth and induce apoptosis in prostate
`cancer cell lines by specifically inhibiting cyclic GMP
`phosphodiesterases but not cyclooxygenase-1 or -2
`[93,94]. Goluboff and colleagues [95] randomised 96 pa-
`tients with rising PSA levels after radical prostatectomy
`to 12 months of exisulind 250 mg orally or placebo twice
`daily and demonstrated that exisulind significantly sup-
`pressed the increase in PSA in all patients and prolonged
`the PSA doubling times (PSADT) in high-risk patients.
`Reduction in the PSADT persisted through 24-month
`follow-up on an open-label extension [96]. Additionally,
`pre-clinical studies have suggested synergistic interac-
`tions between exisulind and several chemotherapeutic
`agents [97–100] and a phase II study examining the com-
`bination of exisulind and docetaxel in men with AIPC
`has completed accrual [101].
`CP-461 is an exisulind analogue with broad anti-
`tumour activity. In a phase I study, 21 patients with a
`range of solid tumours (not including prostate cancer)
`received CP-461 twice daily for 28 d. Therapy was well
`tolerated and 4 patients exhibited disease stability after
`two cycles of treatment [93]. A phase II study of
`CP-461 in prostate cancer is underway.
`
`8.3. Clusterin antisense oligonucleotide
`
`Clusterin is a survival gene whose expression in-
`creases markedly in response to androgen-deprivation
`therapy [102]. The expression of clusterin confers a che-
`motherapy-resistant phenotype, probably due to reduc-
`tion
`in
`treatment-induced
`apoptosis
`[103,104].
`Oligonucleotide antisense molecules to clusterin have
`been developed and have entered clinical
`trials in
`prostate cancer [105]. Development in combination with
`chemotherapy is expected.
`
`9. Other novel targeted agents
`
`9.1. Bortezomib
`
`Bortezomib (PS-341, Velcade) is a novel boronic acid
`dipeptide
`that
`inhibits
`26S proteasome
`activity.
`Although this agent has been most extensively studied
`in multiple myeloma, there is some intriguing evidence
`of its potential utility in prostate cancer. Several investi-
`gators have demonstrated that bortezomib is active in
`
`

`

`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`959
`
`pre-clinical models of human prostate cancer. Ikezoe
`and colleagues demonstrated that bortezomib blocks
`the androgen receptor signalling pathway and induces
`growth arrest and apoptosis in LNCaP cells [106]. Wil-
`liams and colleagues found that bortezomib appeared
`to have anti-tumour activity in both LNCaP and PC-3
`lines, but with distinctly different underlying mecha-
`nisms. The LNCaP lines demonstrated reduced micro-
`vessel density and VEGF secretion and high levels of
`apoptosis, while the PC-3 lines had direct increases in tu-
`mour cell death [107].
`Papandreou and colleagues conducted a phase I trial
`of bortezomib in 48 patients with AIPC. Doses ranging
`from 0.13 to 2 mg/m2 were administered weekly · 4,
`with cycles repeated every 5 weeks. Dose-limiting toxic-
`ity was noted in 2 of 5 patients treated at the 2 mg/m2
`dose level (grade 3 diarrhoea in both patients, grade 3
`syncope and hypotension in 1 patient). The maximally
`tolerated dose (MTD) recommended with this schedule
`was 1.6 mg/m2. Twenty-four patients were treated at
`the MTD (P1.45 mg/m2).
`dosages in the range of
`Two patients (8%) demonstrated a P50% decline in
`PSA and 1 of 9 (11%) patients with measurable nodal
`disease achieved a partial response [108]. Based on pre-
`clinical data suggesting potential
`synergism with
`chemotherapy, phase II trials of bortezomib in combi-
`nation with docetaxel were initiated [109–112]. Dreicer
`and colleagues conducted a phase I/II trial (the phase
`II portion consisted of three separate cohorts of pa-
`tients treated at various dosages) of bortezomib plus
`docetaxel in 100 patients with metastatic AIPC. Data
`from the phase I portion of the study and the low ex-
`panded cohort were presented recently. Docetaxel was
`given at 25, 30, 35 and 40 mg/m2 (cohorts 1–4) i.v. over
`30 min on days 1 and 8. For the first 4 cohorts, bort-
`ezomib was given at a fixed dose of 1.3 mg/m2 i.v. push
`on days 2 and 9, while a fifth cohort evaluated a higher
`dose of bortezomib at 1.6 mg/m2 with 40 mg/m2 of
`docetaxel, with treatment in all cohorts repeated every
`21 d. In the phase I portion of the study no maximally
`tolerated dose was determined. A total of 32 patients
`were enrolled in the low expanded cohort. Twenty-
`one patients had prior chemotherapy, 11 with a tax-
`ane-containing regimen. Therapy was well tolerated;
`anaemia was the most common grade 3 event (2 if 32
`patients). Six of 25 patients (24%) evaluable for re-
`sponse had a confirmed PSA decrease of P50% from
`baseline. Three of 13 (23%) patients with measurable
`disease achieved a partial response [113].
`
`9.2. Atrasentan
`
`Endothelin-1 is a potent vasoconstrictor produced
`by prostate cancer and appears to have a role in pros-
`tate cancer progression and morbidity. Atrasentan is
`an oral, highly selective endothelin receptor antagonist.
`
`In a phase III, placebo-controlled trial that involved
`809 patients with metastatic AIPC, atrasentan did
`not prolong time to clinical progression (HR 1.14,
`95% CI 0.98–1.34) in an intent-to-treat analysis. Atras-
`entan therapy was associated with smaller increases in
`serum PSA and markers of bone turnover. A modest
`delay in time to clinical progression (HR 1.26, 95%
`CI 1.06–1.50) was noted in the protocol-compliant pa-
`tients [114].
`An intent-to-treat meta-analysis that included this
`study as well as a prior randomised phase II study dem-
`onstrated a delay in disease progression (HR 1.19,
`P = 0.013), but the overall clinical benefit was small.
`These studies are characterised by rapid early progres-
`sion of a subset of patients, which contributed to the dif-
`ficulty detecting a benefit in the individual trials. A
`phase III study of this agent in earlier stages of AIPC
`will probably determine its fate.
`
`10. Immunotherapeutic agents
`
`10.1. GM-CSF
`
`Granulocyte-macrophage colony stimulating factor
`(GM-CSF) is a cytokine that regulates the proliferation
`and differentiation of myeloid precursor cells and en-
`hances the function of both mature granulocytes and
`mononuclear phagocytes. GM-CSF can influence the
`recruitment, activation and survival of macrophages
`and dendritic cells. Additionally, experimental evidence
`suggests that GM-CSF may increase the efficiency of
`tumour antigen presentation and the subsequent acti-
`vation of
`tumour-specific cytotoxic T lymphocytes
`[115,116]. Small and colleagues administered GM-
`CSF (250 lg/d daily · 2 weeks followed by three times
`a week administration)
`to patients with metastatic
`AIPC to evaluate their hypothesis that apoptotic tu-
`mour cells could provide a source of tumour antigen,
`which might lead to T-cell cross-priming by dendritic
`cells. Therapy was well tolerated and a small subset
`of the patients experienced a greater than 50% decline
`in PSA, with one patient demonstrating both a pro-
`longed PSA response and objective improvement in
`bone scan imaging [117]. Dreicer and colleagues treated
`16 patients with advanced prostate cancer (7 hormon-
`ally-naı¨ve and 9 androgen-independent) with GM-
`CSF administered subcutaneously at 250 lg thrice
`weekly for up to 6 months. Although no patient
`achieved an objective response, 6 patients demon-
`strated a 10–15% decline in their baseline PSA that
`was maintained during the entire treatment period.
`Five of these 6 patients demonstrated a rise in their
`PSA following study completion. Therapy was well
`tolerated, with only one grade 3 event, which was
`probably not treatment-related [118].
`
`

`

`960
`
`J.M. Strother et al. / European Journal of Cancer 41 (2005) 954–964
`
`10.2. GVAX
`
`10.5. MVA-MUC1-IL2 vaccine
`
`GVAX cancer vaccines are comprised of tumour cells
`that have been genetically modified to secrete GM-CSF.
`GVAX was studied in a phase II dose escalation trial.
`Nineteen patients were treated at the highest dose. Of
`these patients, 6 (32%) had modest declines in serum
`PSA and 87% demonstrated immune response [119].
`Phase III studies of GVAX are planned.
`
`10.3. Provenge
`
`Provenge consists of autologous dendritic cells
`loaded ex vivo with a recombinant fusion protein of
`prostatic acid phosphatase (PAP) linked to GM-CSF.
`In early studies, Provenge was shown to induce immune
`responses to PAP in 38% of patients and occasional PSA
`reductions in excess of 50% [120]. A single durable re-
`sponse has been reported [121]. In the initial randomised
`study in AIPC, Provenge failed to improve progression-
`free survival, but a benefit in progression-free survival
`and overall survival was seen in a subset of patients
`whose Gleason score was less than 8 [122]. As a result,
`Provenge is now being evaluated in patients with meta-
`static AIPC that at initial diagnosis did not exceed Glea-
`son 7. It is also being tested in a placebo-controlled
`randomised study in patients with a rising serum PSA
`after prostatectomy.
`
`Over-expression, non-polarity and under-glycosyla-
`tion of the mucin glycoprotein MUC1 is associated
`with many epithelial neoplasms, making MUC1 a po-
`tential
`target of vaccine
`immunotherapy. MUC1
`expression has been reported in 60–90% of prostate
`cancer specimens evaluated [124,125]. Modified vac-
`cinia ankara (MVA) is a highly attenuated vaccinia
`virus that is non-propagative in most mammalian cells.
`TG4010 is a recombinant MVA expressing MUC1 and
`IL-2 that demonstrated an excellent safety profile in
`phase I evaluations. Interim results from a randomised
`phase II trial of two different schedules of TG4010 in
`patients with hormone-naı¨ve biochemical failure have
`been reported recently. Eligible patients were required
`to have a PSA doubling time of less than 10 months,
`with an absolute value of 2.0 ng/ml or greater without
`evidence of metastatic disease. Patients were randomly
`assigned to receive either weekly injections of 108 pla-
`que-forming units for the first 6 weeks then injections
`every 3 weeks until week 36 or progressive disease, or
`the same dosage administered every 3 weeks until week
`36 or progressive disease. At the time of the interim
`assessment, 29 patients had been treated on study,
`with a significant change in PSA doubling time from
`pre-treatment assessment
`favouring arm 1 (weekly
`therapy) [126].
`
`10.4. Vaccinia virus/fowlpox virus
`
`10.6. J591 monoclonal antibody
`
`Various investigators have pursued immunotherapy
`with viral vectors since they can mimic natural infection
`and produce potent immune responses. Investigators
`from the Eastern Cooperative Oncology Group
`(ECOG) have recently reported a phase II trial designed
`to evaluate the tolerability and feasib

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