throbber
New drugs in prostate cancer
`Andrew J. Armstrong and Michael A. Carducci
`
`Purpose of review
`The survival of hormone-refractory metastatic prostate
`cancer patients has improved with the use of docetaxel-
`based chemotherapy. The survival benefits, however, are
`modest suggesting that rationally designed therapeutic
`approaches are needed. We discuss recent
`developments in the therapeutic approach to advanced
`metastatic hormone-refractory prostate cancer, including
`molecularly targeted therapy, signal transduction
`inhibitors, stem-cell targeted therapy, anti-angiogenic
`compounds, vaccines and immunomodulating agents,
`differentiation agents, cytotoxics, and pro-apoptotic
`agents.
`Recent findings
`Over 200 compounds have entered clinical development
`for use in advanced prostate cancer, alone or in
`combination with cytotoxic agents such as docetaxel, or in
`other combinations. This article will review the results of
`emerging targets since the approval of docetaxel in 2004,
`concentrating on some of those compounds that, in our
`opinion, have the greatest potential and rationale for use.
`Summary
`The growing field of targeted molecular therapy of prostate
`cancer has opened up numerous opportunities for
`therapeutic impact. Knowledge of the molecular
`determinants of progression, relapse after local therapy,
`chemotherapeutic resistance, and hormone refractoriness
`remains essential in the rational design of clinical trials of
`these agents. Given the complexity, heterogeneity, and
`crosstalk of molecular pathways and the molecular lesions
`in prostate cancer, combination or sequential therapy may
`be a necessary step towards significant therapeutic
`progress. Novel translational clinical trial methodologies
`may assist in a more rapid identification of active
`compounds at biologically active doses for phase-III testing.
`
`Keywords
`angiogenesis, apoptosis, hormone refractory, metastatic
`prostate cancer, novel agents, prostate cancer, targeted
`therapy
`
`Curr Opin Urol 16:138–145. # 2006 Lippincott Williams & Wilkins.
`
`Prostate Cancer Research Program, Sidney Kimmel Comprehensive Cancer
`Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
`
`Correspondence to Michael A. Carducci MD, Associate Professor of Oncology
`and Urology, Prostate Cancer Research Program, Sidney Kimmel Comprehensive
`Cancer Center, Johns Hopkins School of Medicine, CRB 1M51, 1650 Orleans
`Street, Baltimore, MD 21231, USA
`Tel: +1 410 502 9746; fax: +1 410 614 9006
`
`Current Opinion in Urology 2006, 16:138–145
`
`138
`
`Abbreviations
`
`EGFR
`epidermal growth factor receptor
`HRPC
`hormone-refractory prostate cancer
`PDGFR platelet-derived growth factor receptor
`PSA
`prostate-specific antigen
`VEGF
`vascular endothelial growth factor
`
`# 2006 Lippincott Williams & Wilkins
`0963-0643
`
`Introduction
`In 2004, docetaxel (Taxotere, Sanofi-Aventis, Bridge-
`water, NJ) with prednisone was US Food and Drug
`Administration (FDA) approved for the treatment of
`metastatic, progressive, hormone-refractory prostate can-
`cer based on findings from TAX327 and SWOG 9916
`[1(cid:127)(cid:127),2]. Survival was increased in TAX327 by a median
`of 2.5 months from 17.4 to 18.9 months as compared
`with mitoxantrone and prednisone, the previous stan-
`dard of care, and was similar to results seen with doce-
`taxel and estramustine in SWOG 9916. Importantly,
`quality-of-life and pain measures were improved despite
`the higher risk for severe neutropenia, fatigue, alopecia,
`diarrhea, stomatitis, nail changes, tearing, peripheral
`edema, and neuropathy. While docetaxel represents an
`improvement in care and the first demonstrable survival
`advantage in this patient population, treatment remains
`palliative and survival benefits are modest. The identi-
`fication of molecular lesions that reproduce the aggres-
`sive cancer phenotype, which are not present in normal
`tissues, is essential to further the field beyond the cur-
`rent cytotoxic plateau and to move towards the develop-
`ment of targeted novel agents.
`
`Growth factor and signal transduction
`pathways
`The last decade has witnessed the identification of hun-
`dreds of cell-signaling molecules important in the devel-
`opment of prostate cancer. Crosstalk between hormonal
`and growth factor signaling pathways creates a redun-
`dancy and complexity to drug development efforts [3].
`Abnormal expression of a target protein may be insuffi-
`cient as a surrogate marker for a therapeutic target, with-
`out independent validation of that target’s role in repro-
`ducing the cancer phenotype and in mediating the
`response to a drug [4]. Indeed, the androgen receptor
`represents the most valid target to date in prostate can-
`cer, and novel strategies to more potently target the
`androgen axis are in development [5(cid:127)(cid:127)]. Unfortunately,
`it is often the expensive failure of a certain drug class to
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`JANSSEN EXHIBIT 2043
`Mylan v. Janssen IPR2016-01332
`
`

`

`impact response, time-to-progression, or survival in pros-
`tate cancer that triggers a reconsideration of the under-
`lying biological model for a known potential target.
`Table 1 provides an overview of the most advanced
`novel agents in clinical trials in prostate cancer.
`
`cancer
`prostate
`hormone-refractory
`In metastatic
`(HRPC), several classes of agents that modify growth
`factor pathways hold some promise and are in early
`phase trials. These include rapamycin analogs that tar-
`get the Akt/PI3 kinase/mTOR pathway, monoclonal
`antibodies, and inhibitors of upstream growth factor
`receptors
`such as HER-2/3,
`IGF-R,
`IL-6R,
`and
`platelet-derived growth factor
`receptor
`(PDGFR).
`PTEN is a tumor suppressor that is lost in a majority
`of metastatic prostate cancers, leading to unrestrained
`activation of the Akt/PI3 kinase pathway and autono-
`mous growth and survival [6,7]. Akt activation and/or
`PTEN loss have been linked to hormonal resistance,
`chemotherapeutic insensitivity, biologically aggressive
`behavior, high Gleason sum, and relapse after local ther-
`apy [8,9]. Mouse models of PTEN loss or Akt activation
`have demonstrated growth inhibition, restoration of che-
`mosensitivity, and improved time-to-progression with
`inhibitors of this pathway, including rapamycin analogs
`[10–12]. Mechanistic dose-finding pre-prostatectomy
`studies are in progress in prostate cancer with three
`rapamycin analogs: CCI-779 (Temsirolimus, Wyeth,
`Collegeville, PA), RAD001 (Everolimus, Novartis, Cam-
`bridge, MA), and rapamycin itself [13–16]. The devel-
`opment of these agents will be dependent on the estab-
`lishment of surrogate markers of biologic effect, the
`identification of subgroups of responders, and drug char-
`acteristics such as dose, pharmacokinetic variability, and
`tolerability. Everolimus,
`in combination with the
`upstream epidermal growth factor receptor (EGFR) tyr-
`osine kinase inhibitor gefitinib, is in phase-II develop-
`ment and the Ariad agent AP23573 (Cambridge, MA) is
`being evaluated currently as a single agent in metastatic
`
`New drugs in prostate cancer Armstrong and Carducci 139
`
`HRPC. All agents in this class have a well described
`pattern of toxicity, including stomatitis, acneform rash,
`glucose intolerance, nausea, fatigue, mild thrombocyto-
`penia, arthralgias, electrolyte abnormalities, and possibly
`increased risks of infection [17]. Their long-term immu-
`nosuppressive safety in prostate cancer patients has yet
`to be tested, but they have been evaluated favorably in
`renal cell carcinoma and other tumor types [18(cid:127)]. The
`use of these agents in combination with docetaxel is
`under
`investigation and requires careful monitoring
`owing to metabolic and myelosuppressive interactions.
`As these agents are primarily cytostatic in prostate can-
`cer, combination therapy with other biologic agents will
`likely be necessary.
`
`While agents targeting the EGFRs such as gefitinib and
`trastuzumab have not been successful in metastatic pros-
`tate cancer, it is likely that the EGFR and HER-2 targets
`for these drugs may not be causal in prostate cancer [19–
`21]. Indeed, HER-2 overexpression in prostate cancer is
`rare, unlike the case in subsets of breast cancer [21].
`Recent studies, however, may point to other growth fac-
`tors and paracrine signals that may be of greater impor-
`tance, such as HER-3, IGF-R, and cytokine receptors,
`such as the receptors for TGF-β and IL-6. Mellinghoff
`et al. [22(cid:127)] found that HER-2/HER-3 dimerization and
`activation led to optimization of androgen-receptor sig-
`naling in the setting of androgen depletion, pointing to
`a potential novel therapeutic target. The ligand for
`HER-3 is neuregulin, and potential therapeutic agents
`that may target HER-2/HER-3 signaling include the
`monoclonal antibody pertuzumab (Omnitarg, Genen-
`tech, San Francisco, CA) and the intracellular inhibitor
`lapatinib (GlaxoSmithKline, Philadelphia, PA) [23,24].
`
`The endothelin axis has been proposed as an important
`mediator of the bone–prostate cancer interface and para-
`crine signaling target. Endothelin receptors are overex-
`pressed in metastatic HRPC and higher
`levels of
`
`Table 1 Selected novel agents in phase-II/III development in metastatic hormone-refractory prostate cancer
`
`Target
`
`Agent, sponsor
`
`Phase
`
`Brief eligibility overview
`
`Vitamin D receptor
`
`Calcitriol (DN101), Novacea
`
`Endothelin axis
`Vascular endothelial growth factor
`
`Atrasentan (Xinlay), SWOG
`Bevacizumab (Avastin), CALGB
`
`Vaccine strategies
`
`Prostate GVAX (Cell Genesys), Provenge
`(Dendreon), TRICOM (NCI, ECOG)
`
`Epidermal growth factor receptor
`(HER-2/HER-3)
`PTEN/Akt and mTOR pathway
`
`Lapatinib, ECOG
`
`Temsirolimus (Wyeth), Everolimus (No-
`vartis), Rapamycin (Johns Hopkins),
`AP23573 (Ariad)
`
`HRPC, hormone-refractory prostate cancer.
`
`III
`
`III
`III
`
`III
`
`II
`
`I–II
`
`ASCENT II: docetaxel ± DN101 in metastatic
`HRPC
`Docetaxel ± atrasentan in metastatic HRPC
`CALGB 90401: docetaxel ± bevacizumab in me-
`tastatic HRPC, first line
`GVAX (VITAL-1), Provenge (D9902B) – first-line
`metastatic, asymptomatic HRPC VITAL-2: doc-
`etaxel ± GVAX in symptomatic HRPC
`Rising PSA setting (nonmetastatic)
`
`Preoperative pharmacodynamic and dose-finding
`studies; combination with docetaxel in meta-
`static HRPC (Dana Farber and MSKCC)
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`

`

`140 Prostate cancer
`
`endothelin correlate with progressive disease [25]. While
`endothelin is a potent vasoconstrictive agent, it may also
`regulate cellular mitogenic pathways in prostate cancer
`and osteoblasts, and may play a role in the mediation of
`bone-related pain from metastatic prostate cancer. Atra-
`sentan (Xinlay, Abbott Labs, Abbott Park, IL) has been
`developed as highly selective ET-A receptor antagonist
`and is the most clinically developed agent of this class in
`prostate cancer [26]. In the phase-III trial, 809 patients
`with metastatic HRPC were randomized to placebo or
`10 mg of oral atrasentan, with the primary clinical end-
`point being time to progression (TTP) [27]. Although
`TTP was not found to be statistically significantly dif-
`ferent from placebo in the intent-to-treat analysis, sev-
`eral secondary endpoints indicated clinical activity,
`including improvements in quality-of-life scores, pain
`scores, and reductions in the rise of laboratory markers
`including alkaline phosphatase and prostate-specific
`antigen (PSA). While atrasentan was not approvable on
`the basis of these data and out of concerns for cardiovas-
`cular toxicity, these results clearly point to biologic
`activity of the endothelin axis in modulating osteoblastic
`metastases, and underscore the importance of
`trial
`design in this population. Further development of this
`agent in combination with docetaxel or in select bone-
`only subgroups of patients may continue. Another ET-A
`receptor antagonist, ZD4054 (Astra Zeneca, Waltham
`MA), is also in phase-II development at this time [28].
`
`Prostate cancer cells express high levels of platelet-
`derived growth factor receptor (PDGFR), and this sig-
`naling pathway utilizes the PI3 kinase/Akt pathway,
`which has been implicated in prostate cancer progres-
`sion [29]. A phase-I study of imatinib (Gleevec, Novar-
`tis, Cambridge, MA) a small-molecule PDGFR tyrosine
`kinase inhibitor used to treat chronic myeloid leukemia
`and gastrointestinal stromal tumor, has been conducted
`in advanced prostate cancer alone and in combination
`with weekly docetaxel. The combination of imatinib
`600 mg and docetaxel 30 mg/m2 weekly for four out of
`six weeks demonstrated a more than 50% PSA reduction
`in eight of 21 patients (38%), with several durable (>18
`month) responses [29]. A second-line randomized trial of
`this combination sponsored by the National Cancer
`Institute Prostate Cancer SPORE clinical consortium is
`underway.
`
`Prostate cancer stem-cell targeted therapy
`While a true prostate cancer stem-cell phenotype has
`yet to be identified, several molecular stem-cell targets
`overexpressed in prostate cancer have been described
`and include elements of the hedgehog signaling path-
`way, human telomerase, and CD133 [30(cid:127),31–33]. Tar-
`geting the hedgehog embryonic pathway, which is over-
`expressed in many metastatic prostate cancers, has been
`
`shown to prevent prostate regeneration after androgen
`withdrawal and to lead to prolonged responses in PC-3
`xenograft models [30(cid:127)]. Novel cyclopamine analogs,
`which inhibit a downstream hedgehog signal, are in pre-
`clinical development.
`
`Human telomerase is responsible for the maintenance of
`chromosome stability and length during cell division,
`and is overexpressed in nearly all cancers. In prostate
`cancer, telomerase may be regulated by androgen or
`growth factors, and is overexpressed as compared with
`normal and benign prostatic hypertrophy tissue [31,32].
`Strategies targeting telomerase have been reviewed in
`detail by Biroccio and Leonetti [31]. Other markers of
`basal epithelial prostate cells that may indicate a subset
`of cancers derived from putative prostate stem cells
`include lack of androgen receptor, CD133, α2β1 integrin
`overexpression, Bcl-2, TGF-β signaling, high-molecular-
`weight cytokeratins, hepsin, and potentially TMPRSS2-
`ETS fusion proteins [30(cid:127),31–33,34(cid:127)(cid:127),35,36]. The identi-
`fication of clonogenic populations of cells in prostate
`cancer with the ability to self-renew and differentiate
`would allow the testing of novel agents directed at
`these cells, provided there was tolerability to normal
`stem-cell populations.
`
`Anti-angiogenic agents
`Prostate cancer
`is known to overexpress vascular
`endothelial growth factor (VEGF) and its receptors,
`and VEGF levels correlate with disease stage and per-
`haps survival in the metastatic setting, with levels falling
`after surgical resection of primary tumors [37,38]. The
`mechanism of action of anti-VEGF therapies is unclear,
`and potentially includes a normalization of vasculature
`for facilitated delivery of chemotherapy, a decrease in
`oncotic interstitial pressure from leaky capillary mem-
`branes, improved recruitment of mature dendritic cells
`and immunostimulation, and true antineovascularization
`effects [39–42]. A phase-III study CALGB 90401 that
`will randomize patients in a phase-III trial to docetaxel
`compared with docetaxel and bevacizumab (Avastin,
`Genentech, San Francisco, CA), including prednisone
`in each arm, is now open with a goal accrual of 1020
`patients over 3 years. This follows on the heels of a
`large phase-II study, which demonstrated the safety
`and efficacy of docetaxel, estramustine, and bevacizu-
`mab in combination for HRPC [43].
`
`agents with anti-angiogenic properties
`Additional
`include thalidomide and its analogs, Revlimid and Acti-
`mid (Celgene, Summitt, NJ), as well as small-molecule
`inhibitors of the VEGF receptor tyrosine kinase such as
`Sorafenib (Onyx, Emeryville CA and Bayer, West
`Haven CT) and Sutent (SU11248, Pfizer, Cambridge,
`MA) [44–46]. These agents are in phase-II development
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`

`

`in HRPC. Thalidomide analogs likely have multiple
`mechanisms of action, including inhibition of VEGF as
`well as improved T-cell costimulatory function, TNF-α
`inhibition, and a decrease in IL-6 levels [44,45]. A
`phase-II randomized study of thalidomide and docetaxel
`in HRPC demonstrated impressive PSA declines, TTP,
`and overall survival but was complicated by a high rate
`of thrombosis, sedation, and neuropathy in the experi-
`mental arm, necessitating the introduction of therapeu-
`tic
`low-molecular-weight heparin prophylaxis
`[47].
`Novel thalidomide analogs are in phase-II development
`in prostate cancer, and are expected to have a lower inci-
`dence of vascular and neurotoxic adverse events [48].
`The high potency of these agents in terms of T-cell sti-
`mulation, anti-angiogenic properties, and oral availabil-
`ity make them attractive as therapeutic agents.
`
`Immunologic approaches
`Several vaccination strategies have progressed beyond
`phase-II testing in prostate cancer, including Provenge,
`a PAP-activated dendritic-cell-based vaccine, and Pros-
`tate GVAX, a whole-cell allogeneic vaccine. Both of
`these agents are under evaluation in the phase-III set-
`ting [49(cid:127)]. Provenge (Sipuleucel-T, Dendreon, San
`Francisco, CA) is a proprietary process of antigen deliv-
`ery to activated antigen-presenting cells, collected from
`patients through leukapheresis, stimulated with fusion
`PAP-GMCSF protein, and reinjected intradermally
`every 4 weeks [50]. Initial results from small phase-III
`studies (D9901 and D9902A) involving a total of 225
`patients with asymptomatic metastatic HRPC did not
`significantly demonstrate improved time to disease or
`pain progression, the primary endpoints [51]. While not
`originally powered to detect a survival benefit, overall
`survival was improved by an average of 4 months in
`each study. Analyses based on chance imbalances in
`prognostic factors and use of chemotherapy after vacci-
`nation did not seem to reduce this survival finding. Den-
`dreon is submitting a Biologics License Application to
`the FDA for marketing approval.
`
`Prostate GVAX (Cell Genesys, San Francisco, CA) is a
`form of active immunotherapy using whole-cell allo-
`geneic prostate cell
`lines (PC-3 and LnCaP) virally
`transduced to express an immune adjuvant GM-CSF,
`lethally irradiated, and injected intradermally. Given
`that GM-CSF likely facilitates the maturation and acti-
`vation of dendritic cells, initial work extrapolated early
`melanoma studies to mouse models of prostate cancer
`with results showing prolonged survival and tumor
`regression [52,53]. A phase-II study of prostate GVAX
`was conducted in 34 patients with metastatic HRPC.
`Median survival in this trial was 26 months, historically
`very favorable, but again, observed in select, asympto-
`matic patients [54]. A further evaluation of 80 patients
`
`New drugs in prostate cancer Armstrong and Carducci 141
`
`with metastatic HRPC treated at higher doses demon-
`strated one partial PSA response and improvement in
`markers of bone turnover, with survival analysis still
`ongoing [55]. A phase-II trial of GVAX compared with
`docetaxel (VITAL-1) in 600 men with minimally symp-
`tomatic metastatic HRPC is currently accruing patients,
`and a second phase-III study examining docetaxel and
`prednisone with or without GVAX has been initiated
`(VITAL-2).
`
`Finally, two other vaccine approaches are in early testing
`and include the Prostvac-VF recombinant vaccinia–
`fowlpox PSA vaccine (TRICOM) and the BLP25
`MUC1 liposomal vaccine MUC-1 [56,57]. The use of
`vaccines alone or in combination with chemotherapy or
`biologic agents, such as CTLA4 blocking antibodies, is
`an exciting area of preclinical and clinical development
`[58].
`
`Differentiation and apoptotic therapy
`Two strategies to exploit the latent and resistant nature
`of tumor growth in prostate cancer are differentiation
`therapy and the use of agents that target the apoptotic
`machinery of cancer cells. In prostate cancer, epidemio-
`logic data suggest that the vitamin D receptor is a poten-
`tially valid target, given the link of vitamin D deficiency
`with prostate cancer development, reviewed elsewhere
`[59]. In vitro, calcitriol may have growth inhibitory, pro-
`apoptotic, and differentiating properties in prostate can-
`cer, as well as potential chemosensitizing properties,
`thus leading to a rationale for clinical trials [60,61].
`Based on favorable phase-II results of calcitriol and doc-
`et al.
`[62,63(cid:127)]
`etaxel
`in combination, Beer
`recently
`reported interim results from a phase-II randomized
`trial (ASCENT) of docetaxel and prednisone with or
`without DN-101 (Novacea, San Francisco, CA), a pro-
`prietary oral calcitriol analog. In this randomized multi-
`institutional study of 250 men with progressive meta-
`static HRPC treated with weekly docetaxel with or
`without DN101,
`the primary endpoint was PSA
`response rate, a typical phase-II endpoint. With a med-
`ian follow-up of 18.3 months, neither PSA nor clinical
`response endpoints were met (6-month PSA response
`58% compared with 49%, P = 0.07, and measurable dis-
`ease response rate 29% compared with 24%, P = 0.58 in
`the placebo compared with treated subjects, respec-
`tively). Despite being underpowered to detect a differ-
`ence in survival, however, the estimated median survival
`was nonsignificantly prolonged,
`from 16.4 to 23.5
`months [hazard ratio 0.70, 95% confidence interval (CI)
`0.48–1.03, P = 0.07], and better tolerated than docetaxel
`alone [63(cid:127)]. This difference became significant with a
`prespecified multivariate adjustment based on chance
`imbalances in this small sample size. A larger study
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`

`

`142 Prostate cancer
`
`with typical phase-III survival endpoints is planned
`(ASCENT II) to confirm these findings.
`
`Other differentiation strategies include inhibitors of his-
`tone deacetylase and DNA methyltransferase, enzymes
`responsible for the epigenetic silencing of gene expres-
`sion. Histones maintain DNA in a closed, coiled config-
`uration, and this activity is mediated by a reversible
`acetylation process, in which acetylation of lysine resi-
`dues on select histone tails favors transcription. Aberrant
`hypermethylation and gene silencing of specific promo-
`ter regions in prostate cancer has been described, such
`as the antioxidant enzyme GST-II and the tumor sup-
`pressor p21 [64]. The rationale behind agents that target
`histone deacetylase or DNA methyltransferase is their
`ability to induce broad gene re-expression in preclinical
`models of prostate cancer, which may induce apoptosis,
`upregulate p21 signaling, and arrest growth [65(cid:127)]. Phase-
`II studies are planned using SAHA (Merck, Whitehouse
`Station, NJ), an orally bioavailable histone deacetylase
`inhibitor,
`in metastatic HRPC. The development of
`these agents in prostate cancer will require careful atten-
`tion to pharmacodynamic endpoints, toxicity, potential
`differentiating effects such as transient PSA rises, and
`the potential for interactions with chemotherapy and
`other biologic agents [66,67].
`
`As the anti-apoptotic Bcl-2 protein is overexpressed in
`metastatic HRPC, and resistance to cell death seems to
`be dominant over proliferation in these tumors, pro-
`apoptotic strategies are attractive therapeutically [68].
`The mechanism of Bcl-2 overexpression is unclear, how-
`ever, and could be related to PTEN loss and/or Akt
`pathway activation, thus providing a common mechan-
`ism for the observed joint occurrences with tumor pro-
`gression [69(cid:127),70]. Two agents that have progressed to
`the phase-II setting include the antisense Bcl-2 mole-
`cule oblimersen sodium (Genasense, Berkeley Heights,
`NJ) and the proteasome inhibitor bortezomib (Velcade,
`Millenium Pharmaceuticals, Cambridge, MA) [71,72].
`Current results have demonstrated feasibility and safety
`but unclear efficacy or target validation in prostate can-
`cer.
`
`Novel cytotoxics
`The dose limitations of docetaxel therapy in metastatic
`HRPC are predominantly those of peripheral neurotoxi-
`city and myelotoxicity, and the search for well tolerated
`novel cytotoxic compounds continues, both in the front
`line and in the second-line setting. One agent in devel-
`opment for second-line therapy is satraplatin (Spectrum
`Pharmaceuticals, Irvine, CA), a novel oral platinum ana-
`log that may fulfill a niche in second-line therapy if it is
`well tolerated and shown to improve survival over corti-
`costeroids in the ongoing SPARC phase-III trial [73].
`The epothilones are a class of microtubule targeting
`cytotoxic agents in development for second-line and
`relapsed HRPC. While sharing a common mechanism
`of action with the taxanes, they are not apparently sus-
`ceptible to P-glycoprotein induced drug efflux [74]. The
`epothilone-B analog BMS-247550 (Ixabepilone, Ingenta,
`Cambridge, MA) has been studied in a phase-II trial of
`men with HRPC [75]. Initial results demonstrated com-
`parable PSA declines and progression-free survival to
`that seen with docetaxel-based therapy. Use of these
`drugs may be limited by dose-limiting neurotoxicity
`similar to that seen with the taxanes. The use of BMS-
`247550 in taxane-resistant HRPC is being investigated
`currently in the second-line setting as compared with
`mitoxantrone and prednisone, and has shown similar
`PSA declines of about 20% [76]. Finally, monoclonal
`antibodies (mAbs)
`targeted to prostate cancer cells
`with tagged cytotoxic agents represent a novel approach
`to therapy. One agent, MLN2704 (Millenium Pharma-
`ceuticals, Cambridge, MA), is a prostate-specific mem-
`brane antigen conjugated maytansinoid agent and is in
`phase-I/II trials currently in HRPC [77]. Radiopharma-
`ceuticals tagged to mAbs have also shown some promise
`and are also in phase-II trials [78–81]. Table 2 provides
`an overview of second-line clinical trials that are ongoing
`in metastatic HRPC.
`
`Conclusion
`Metastatic HRPC in 2006 remains an incurable disease
`with a median survival of 18–20 months with current
`docetaxel-based chemotherapy regimens. Patients that
`are asymptomatic or have slow PSA doubling times
`
`Table 2 Agents in phase-II/III trials for use in second-line therapy after docetaxel failure in metastatic hormone refractory
`prostate cancer
`
`Agent
`
`Satraplatin
`
`Epothilone B Analog BMS-247550 (Ixabepilone)
`Targeted cytotoxics and radiopharmaceuticals: MLN2704,
`177Lu/ 90Y J591,
`PDGFR: Imatinib(Gleevec)
`
`Phase
`
`Trial, sponsor
`
`III
`
`II
`II
`
`II
`
`SPARC trial (Spectrum Pharmaceuticals):
`prednisone ± satraplatin
`ECOG: BMS compared with mitoxantrone and prednisone
`MLN2704: MSKCC, Millenium Pharmaceuticals; J591:
`Cornell University
`MD Anderson and Prostate Cancer Foundation with docetaxel
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`

`

`New drugs in prostate cancer Armstrong and Carducci 143
`
`10 Neshat MS, Mellinghoff IK, Tran C, et al. Enhanced sensitivity of PTEN-defi-
`cient tumors to inhibition of FRAP-mTOR. Proc Natl Acad Sci USA 2001;
`98:10314–10319.
`
`11 Podsypanina K, Lee RT, Politis C, et al. An inhibitor of mTOR reduces neo-
`plasia and normalizes p70/s6 kinase activity in PTEN +/- mice. Proc Natl
`Acad Sci USA 2001; 98:10320–10325.
`
`12 Grunwald V, DeGraffenried L, Russel D, et al. Inhibitors of mTOR reverse
`doxorubicin resistance conferred by PTEN status in prostate cancer cells.
`Cancer Res 2002; 62:6141–6145.
`
`13 Gera JF, Mellinghoff IK, Shi Y, et al. Akt activity determines sensitivity to
`mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1
`and c-myc expression. J Biol Chem 2004; 279:2737–2746.
`
`14 Tabernero J. A phase I study with tumor pharmacodynamic evaluation of
`dose and schedule of the oral mTOR inhibitor Rapamycin (RAD001) in
`patients with advanced solid tumors. Proc Am Soc Clin Oncol 2005
`(abstract 3007).
`
`15 Thomas G, Speicher L, Reiter R, et al. Demonstration that temsirolimus pre-
`ferentially inhibits the mTOR pathway in the tumors of prostate cancer
`patients with PTEN deficiencies. AACR-NCI-EORTC International Confer-
`ence on Molecular Targets and Cancer Therapeutics, Philadelphia, PA
`2005; (abstract C131).
`
`16 Hidalgo M, Rowinsky EK. The rapamycin-sensitive signal transduction path-
`way as a target for cancer therapy. Oncogene 2000; 19:6680–6686.
`
`17 Raymond E, Alexandre J, Faivre S, et al. Safety and pharmacokinetics of
`escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR
`inhibitor, in patients with cancer. J Clin Oncol 2004; 22:2336–2347.
`18 Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase II study of mul-
`tiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase
`inhibitor,
`in patients with advanced refractory renal cell carcinoma. J Clin
`Oncol 2004; 22:909–918.
`This is the largest study to date reported of an mTOR inhibitor, CCI-779, in can-
`cer patients, and provides an idea of the tolerability of these agents in patients
`with genitourinary malignancies.
`
`19 Schro¨ der FH, Wildhagen MFZD. 1839(gefitinib) and hormone resistant (HR)
`prostate cancer - final results of a double blind randomized placebo-con-
`trolled phase II study. Proc Am Soc Clin Oncol 2004; 22 (14S) (Abstract
`4698).
`
`20 Moore M, Winquist E, Pollak M, et al. A randomised phase II study of two
`doses of ZD1839 in patients (pts) with hormone refractory prostate cancer
`(HRPC): A NCI Canada Clinical Trials Group Study. Ann Oncol 2002; 13
`(Suppl 5) (abstract 326).
`
`21 Ziada A, Barqawi A, Glode LM, et al. The use of trastuzumab in the treat-
`ment of hormone refractory prostate cancer: phase II trial. Prostate 2004;
`60:332–337.
`22 Mellinghoff IK, Vivanco I, Kwon A, et al. Her2/neu kinase-dependent modula-
`tion of androgen receptor function through effects on DNA binding and sta-
`bility. Cancer Cell 2004; 6:517–527.
`This recent report characterizes the signaling crosstalk between cell membrane
`growth factor receptors and the androgen receptor axis, and raises questions
`about the downstream targets of HER-2/3 in prostate cancer.
`
`23 Agus DB, Gordon MS, Taylor C, et al. Phase I clinical study of pertuzumab, a
`novel HER dimerization inhibitor, in patients with advanced cancer. J Clin
`Oncol 2005; 23:2534–2543.
`
`24 Liu Y, Majumder S, McCall W, et al. Inhibition of HER-2/neu kinase impairs
`androgen receptor recruitment to the androgen responsive enhancer. Can-
`cer Res 2005; 65:3404–3409.
`
`25 Nelson J, Bagnato A, Battistini B, et al. The endothelin axis: emerging role in
`cancer. Nat Rev Cancer 2003; 3:110–116.
`
`26 Yin JJ, Mohammad KS, Kakonen SM, et al. A causal role for endothelin-1 in
`the pathogenesis of osteoblastic bone metastases. Proc Natl Acad Sci USA
`2003; 100:10954–10959.
`
`27 Carducci M, Nelson JB, Saad F, et al. Effects of atrasentan on disease pro-
`gression and biological markers in men with metastatic hormone-refractory
`prostate cancer: phase 3 study. Proc Am Soc Clin Oncol 2004 (abstract
`4508).
`
`28 Lassiter LK, Carducci MA. Endothelin receptor antagonists in the treatment
`of prostate cancer. Sem Oncol 2003; 30:678–688.
`
`may have a survival of over 2 years; thus, it is important
`to stratify patients based on symptoms, disease sites,
`PSA kinetics, performance status, and other prognostic
`markers to facilitate adequate comparisons of different
`agents in clinical trials [82]. As the number of novel
`agents has increased dramatically in recent years, and
`combination therapy in phase-III trials is accelerating,
`attention to prognostic factors, survival, and quality-of-
`life endpoints, and the mechanism of action is essential
`in the rational development of these drugs. Early studies
`of surrogate assays for survival and the mechanism of
`action of these targeted agents are essential for their
`future development.
`
`Disclosures
`Funding for some of the studies described in this pre-
`sentation was provided by Aventis and Abbott Labora-
`tories. Dr Carducci is on the Speakers Bureau for Aven-
`tis and is a consultant to Abbott Laboratories. He has
`participated as an investigator in studies described in
`this presentation. The terms of this arrangement are
`being managed by the Johns Hopkins University in
`accordance with its conflict of interest policies.
`
`References and recommended reading
`apers of particular interest, published within the annual period of review, have
`been highlighted as:
`(cid:127) of special interest
`(cid:127)(cid:127) of outstanding interest
`Additional references related to this topic can also be found in the Current
`World Literature section in this issue (pp. 197–198).
`1 Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitox-
`antrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;
`351:1502–1512.
`This largest phase-III trial ever conducted in meta

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