throbber
NIH Public Access
`Author Manuscript
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`Published in final edited form as:
`Cancer Lett. 2010 May 1; 291(1): 1–13. doi:10.1016/j.canlet.2009.08.012.
`
`Novel targeted therapeutics for metastatic castration-resistant
`prostate cancer
`
`Emmanuel S. Antonarakis*, Michael A. Carducci, and Mario A. Eisenberger
`Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, United
`States
`
`Abstract
`Virtually all patients that succumb to prostate cancer die of metastatic castration-resistant disease.
`Although docetaxel is the standard of care for these patients and is associated with a modest
`prolongation of survival, there is an urgent need for novel treatment strategies for metastatic
`prostate cancer. In the last several years, great strides have been made in our understanding of the
`biological and molecular mechanisms driving prostate cancer growth and progression, and this has
`resulted in widespread clinical testing of numerous new targeted therapies. This review discusses
`some of the key therapeutic agents that have emerged for the treatment of metastatic castration-
`resistant prostate cancer in the last 5 years, with an emphasis on both molecular targets and
`clinical trial design. These agents include mammalian target of rapamycin (mTOR) pathway
`inhibitors, anti-angiogenic drugs, epidermal growth factor receptor (EGFR) inhibitors, insulin-like
`growth factor (IGF) pathway inhibitors, apoptosis-inducing drugs, endothelin receptor antagonists,
`receptor activator of nuclear factor κB (RANK) ligand inhibitors, vitamin D analogues,
`cytochrome P17 enzyme inhibitors, androgen receptor modulators, epigenetic therapies, vaccine
`therapies, and cytotoxic T lymphocyte-associated antigen (CTLA)-4 blocking agents.
`
`Keywords
`Metastatic castration-resistant prostate; cancer; Targeted therapies; Immune therapies; Molecular
`targets; Clinical trials; Drug development
`
`1. Introduction
`
`Prostate cancer is the most common malignancy in men worldwide. In the United States,
`there were an estimated 186,300 new diagnoses of prostate cancer and 28,700 prostate
`cancer deaths in 2008, representing 25% of new cancer cases and 10% of male cancer deaths
`
`© 2009 Elsevier Ireland Ltd. All rights reserved.
`*Corresponding author. Address: Prostate Cancer Research Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins
`University, 1650 Orleans Street, CRB1-191, Baltimore, MD 21231, United States. Tel.: +1 410 614 4459; fax: +1 410 955 0125.
`eantona1@jhmi.edu (E.S. Antonarakis).
`Conflict of interest
`ESA indicates no financial or other conflicts of interest. MAC has served as a consultant for Sanofi-Aventis, Abbott, Pfizer,
`Genentech, AstraZeneca, Novartis, Wyeth, and Cougar Biotechnology (now Johnson & Johnson); he has received honoraria from
`Sanofi-Aventis and Abbott. MAE has served as a consultant for Sanofi-Aventis, Ipsen, Celgene, and Centocor (now Centocor Ortho
`Biotech).
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`AVENTIS EXHIBIT 2232
`Mylan v. Aventis, IPR2016-00712
`
`

`
`Antonarakis et al.
`
`Page 2
`
`[1]. This makes prostate cancer the second leading cause of cancer death in men. While the
`disease can potentially be cured when localized, metastatic prostate cancer remains
`incurable.
`
`Treatment of localized prostate cancer is usually centered around surgery and/or radiation
`therapy. However, even after definitive local therapy, approximately 30–50% of patients
`will have a local or distant recurrence [2,3]. Patients with metastatic prostate cancer have a
`median survival of 3–7 years, and most men die of it [2]. Treatment for metastatic disease
`involves surgical castration or hormonal manipulation using gonadotropin-releasing
`hormone (GnRH) agonists, antiandrogens, or both. Although the majority of these patients
`initially respond to androgen deprivation therapy, all eventually progress to a state of
`castration-resistant prostate cancer (CRPC). Treatment options for these men are limited.
`Secondary hormonal manipulations, such as ketoconazole, are often used but these produce
`short-lived responses and do not improve survival [4]. Other acceptable approaches in these
`men include watchful waiting until the development of symptoms, or the initiation of
`cytotoxic chemotherapy. In this regard, the chemotherapeutic agent docetaxel has been
`shown to improve overall survival in patients with CRPC, but only by a median of 2.9
`months (median survival 19.2 months versus 16.3 months using mitoxantrone, P = .004)
`[5,6]. Novel therapies for this patient population are urgently needed.
`
`Since the approval of docetaxel by the Food and Drug Administration (FDA) in 2004, no
`new anti-cancer therapies have entered the market for the treatment of metastatic CRPC. On
`the other hand, the number of agents for CRPC in various stages of clinical development is
`higher than ever before. This has been made possible due to our accelerated understanding
`of the biological and molecular mechanisms underpinning prostate cancer growth and
`spread, which has fueled an expansion in research on new therapeutic approaches. This
`review will highlight novel targeted therapies that have emerged for CRPC in the last 5
`years, focusing on the mechanism of action and developmental status of some key clinical
`compounds that have reached phase II and III clinical trials (Table 1). Advances in
`chemotherapeutic drugs, hormonal agents, and bisphosphonates will not be discussed herein.
`
`2. Targeted treatments
`
`Although a prostate cancer stem cell has yet to be conclusively demonstrated, prostate
`cancer clearly progresses from an androgen-dependent tumor (with features similar to the
`luminal differentiated glands of the prostate) to a castration-resistant tumor that has features
`of adult stem cells, including anti-apoptotic mechanisms, chemotherapy resistance, and
`reliance on nonhormonal signaling pathways [7]. Candidate prostate cancer progression
`pathways under investigation include epidermal growth factor receptor (EGFR) signaling,
`vascular endothelial growth factor (VEGF) receptor-mediated pathways,
`phosphatidylinositol 3-kinase (PI3K)/Akt signaling, the insulin-like growth factor (IGF)
`axis, Hedgehog signaling, mitogen-activated protein (MAP) kinase signaling, the endothelin
`axis, and others. Given the molecular complexity of these pathways in the prostate cancer
`cell and the potential redundancy of individual pathways in the process of cancer
`progression, the simultaneous inhibition of multiple pathways remains a common strategy to
`induce sustained and clinically meaningful responses in metastatic CRPC.
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 3
`
`The major biologic processes under therapeutic investigation in CRPC involve growth and
`survival, nutrition, apoptosis, chemotherapy and hormone therapy resistance, extra-gonadal
`androgen production, modulation of the androgen receptor, angiogenesis, the bone interface,
`epigenetic regulation, immune surveillance and escape, and stem cell renewal. This article
`provides an overview of these pathways as they pertain to prostate cancer rational targets
`and the approaches that are currently being developed for therapeutic purposes (Table 1).
`
`2.1. PI3K/Akt/mTOR pathway
`
`In advanced prostate cancer, loss of the tumor suppressor gene PTEN occurs in more than
`50% of metastatic lesions and in approximately 20% of locally advanced lesions [8,9]. Loss
`of PTEN correlates with high Gleason score and stage, chemotherapy resistance, and other
`features of advanced prostate cancers [8]. PTEN is a negative regulator of the
`phosphatidylinositol 3-kinase (PI3K)/Akt survival pathway, and advanced prostate cancers
`frequently have elevated levels of phosphorylated (activated) Akt [10]. The Akt pathway is
`involved in signal transduction from multiple cell surface receptors, including the insulin
`receptor, epidermal growth factor receptor, insulin-like growth factor receptor, platelet-
`derived growth factor receptor, and interleukin-6 receptor, and it is likely to function as a
`cellular sensor for nutrient and growth signals [11]. In addition to promoting cell survival
`through the inhibition of apoptosis, the Akt pathway regulates cell growth, proliferation, and
`angiogenesis through the mTOR (mammalian target of rapamycin) pathway and the
`facilitated translation of signals such as c-Myc, cyclin D, and vascular endothelial growth
`factor [10]. Restoration of functional PTEN activity or inhibition of mTOR activity can
`block the growth of PTEN−/− prostate cancer xenografts in mice and restore chemotherapy
`(and possibly hormonal) sensitivity [12,13].
`
`Rapamycin is a natural compound derived from soil samples containing the bacterium
`Streptomyces hygroscopicus, and has been used as a potent immunosuppressive agent in
`solid organ transplantation. Its antiproliferative properties and anti-tumor activity in cell
`lines also led to its clinical development in cardiology as a means of preventing stent
`restenosis and in oncology, in which a wide variety of tumors were found to exhibit
`sensitivity to this agent and its analogue, temsirolimus [14–16]. Temsirolimus has now been
`approved for the treatment of metastatic renal cell carcinoma [17]. Toxicities with
`rapamycin and its analogues are predictable and are not dose-related; they include
`maculopapular rash, hypertriglyceridemia, hyperglycemia, allergic reactions, pedal edema,
`mucositis, and thrombocytopenia [14,17–19].
`
`Although mTOR inhibitors probably have little single-agent activity in advanced CRPC
`[20], the combination of these agents with docetaxel is an attractive option given their ability
`to reverse chemotherapy resistance in prostate cancer cell lines [21]. In addition, these
`agents induce apoptosis when they are given in combination with chemotherapy in patients
`who have demonstrable activation of the Akt pathway as a result of PTEN mutation/loss or
`other genetic alterations [22]. To this end, the mTOR inhibitor, everolimus, is currently
`being evaluated in combination with docetaxel for the first-line treatment of metastatic
`CRPC in a phase I/II clinical trial [23]. Everolimus is already approved for the treatment of
`advanced renal cell carcinoma [24]. A new mTOR inhibitor, deforolimus (AP23573), is also
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 4
`
`being investigated in the phase II setting as single-agent therapy for men with advanced
`taxanerefractory CRPC.
`
`2.2. Angiogenesis targets
`
`Tumor angiogenesis is likely to be an important biologic component of prostate cancer
`metastasis, and elevated levels of the potent angiogenic molecule, vascular endothelial
`growth factor (VEGF), have been shown to correlate with advanced clinical stage and
`survival [25,26]. In a retrospective study of archived serum samples, VEGF levels were
`independently associated with survival from prostate cancer [27]. Similarly, antibodies to
`VEGF have slowed prostate xenograft growth rates, especially in combination with
`chemotherapy [28,29].
`
`These findings led to the phase II CALGB 90006 trial, which added bevacizumab to
`docetaxel and estramustine in men with metastatic CRPC. Among 79 treated patients in this
`study, a fall in PSA of 50% or more was seen in 65% of men, median time to progression
`was 9.7 months, and overall median survival was 21 months [30]. Other phase II trials
`combining docetaxel and bevacizumab have also shown promising results [31,32]. These
`favorable trials have led to the design of a phase III randomized study (CALGB 90401)
`evaluating docetaxel 75 mg/m2 every 3 weeks and prednisone 10 mg daily plus either
`bevacizumab 15 mg/kg or placebo given every 3 weeks. The primary endpoint of this trial is
`overall survival, and accrual of 1020 patients with metastatic CRPC has been completed.
`The initial results of this pivotal trial are awaited.
`
`Thalidomide was originally developed in the 1960s for treatment of morning sickness and
`subsequently linked to teratogenic effects resulting in phocomelia and dysmelia. Whereas
`the exact mechanism of teratogenesis is unproven, the metabolites of thalidomide have been
`shown to inhibit angiogenesis through multiple potential mechanisms, including inhibition
`of pro-angiogenic signals such as VEGF, basic fibroblast growth factor (bFGF),
`interleukin-6, and tumor necrosis factor-α [33,34]. Preclinical studies suggest that
`thalidomide also has T-cell co-stimulatory activity and immunomodulatory properties. Phase
`I/II studies using high doses of thalidomide as a single agent have yielded low PSA response
`rates in the order of 20% [33,35]. However, in a randomized phase II trial of weekly
`docetaxel and low-dose thalidomide versus docetaxel alone, PSA responses, time to disease
`progression, and overall survival were greater in the combination arm [36]. Although this
`trial was underpowered to detect a difference from the standard arm, the clinical activity and
`manageable toxicity of this agent have led to the development of more potent thalidomide
`analogues for combination therapy, and these are currently undergoing clinical evaluation.
`Finally, a recent report of a phase II trial using a three-drug combination of docetaxel,
`thalidomide and bevacizumab showed PSA responses in approximately 80% of patients;
`however neurotoxicity was significant with this combination [37].
`
`Toxicities with thalidomide include deep venous thrombosis, sedation, neuropathy,
`constipation, and fatigue. Newer thalidomide analogues with immunomodulatory features
`have been developed that lack the neurotoxicity of thalidomide but retain many of the T-cell
`modulatory effects, anti-angiogenic properties, and even direct pro-apoptotic functions [34].
`Lenalidomide and CC-4047 are second-generation compounds with much more potent
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 5
`
`tumor necrosis factor-α inhibition than the parent compound, and clinical testing with these
`agents has begun. For example, several phase I and II studies have revealed PSA responses
`and partial radiological responses with lenalidomide, both when used alone and when
`combined with ketoconazole or docetaxel [38–40]. However, phase III trials using
`thalidomide or lenalidomide in CRPC have not yet been conducted.
`
`There has been a recent interest in the evaluation of tyrosine kinase inhibitors (TKIs), agents
`which block angiogenic growth factor targets such as the VEGF and PDGF receptors. The
`drug sorafenib is an oral inhibitor of RAF kinase, VEGFR, and PDGFR, which has been
`approved for use in metastatic renal cell carcinoma and hepatocellular carcinoma [41,42]. In
`phase II studies using sorafenib in men with metastatic CRPC, this agent was shown to
`prevent radiological progression and cause regression of bone metastases in some patients,
`but without affecting PSA levels [43,44]. The agent sunitinib and a novel multi-kinase
`inhibitor, vatalinib, are currently being tested in phase II studies in combination with
`docetaxel for chemotherapy-naïve CRPC; radiological responses rather than PSA responses
`have been chosen as primary endpoints in these trials. Finally, single-agent sunitinib is being
`evaluated in a phase III study of patients with docetaxel-refractory disease.
`
`An alternative anti-angiogenic strategy is the use of VEGF decoy receptors (VEGF-Trap) to
`saturate circulating VEGF and prevent it from binding to its natural transmembrane receptor.
`One such agent is aflibercept (AVE0005), a novel recombinant decoy fusion protein of
`VEGFR and the Fc fragment of IgG1 [45]. In a phase I/II study of intravenous aflibercept
`combined with docetaxel in 54 heavily-pretreated patients with advanced solid tumors, the
`optimal dose of aflibercept was determined to be 6 mg/kg given every 3 weeks [46].
`Toxicities from this combination regimen included neutropenia, hypertension, proteinuria,
`epistaxis, and dysphonia. Five patients (9%) achieved partial radiological responses, and 32
`(59%) had stable disease. A multicenter, randomized, placebo-controlled phase III study of
`docetaxel with or without aflibercept in men with chemotherapy-naïve metastatic CRPC is
`now accruing patients.
`
`A final approach to angiogenesis inhibition involves the use of tumor-vascular disrupting
`agents, drugs that primarily act against established tumor blood vessels by disrupting
`vascular endothelial cells and causing a range of subsequent antivascular effects [47]. The
`prototype in this class of agents is 5,6-dimethylxanthenoine-4-acetic acid (DMXAA).
`Motivated by experiments showing that DMXAA functioned synergistically with docetaxel
`in human prostate cancer xenografts [48], a multicenter randomized phase II trial of
`docetaxel plus or minus DMXAA (1200 mg/m2 intravenously every 3 weeks) was
`conducted for men with metastatic CRPC in the first-line setting. In that study of 71 patients,
`PSA responses (>30% PSA reduction) at 3 months were 47% and 63% in the docetaxel-
`alone and docetaxel-DMXAA arms, respectively, and radiological response rates were 9%
`and 23% in the monotherapy-arm and the combination-arm, respectively [49]. Adverse
`events with DMXAA-docetaxel included neutropenia/febrile neutropenia, cardiac toxicities
`(supraventricular tachycardia, myocardial ischemia), gastrointestinal effects, and infectious
`complications.
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 6
`
`2.3. EGFR and PDGFR pathways
`
`The rapid development in the last several years of small molecules that inhibit tyrosine
`kinases has yielded encouraging results in a host of cancers. Demonstration of tumor
`response has sometimes correlated with mutation in the target tyrosine kinase, such as
`epidermal growth factor receptor (EGFR), Bcr-Abl, and c-Kit. In these cases, the target
`mutation has played a central role in the pathogenesis of these tumors. In prostate cancer, no
`such mutations have been identified, perhaps explaining why early trials of tyrosine kinase
`inhibitors in prostate cancer have been disappointing.
`
`EGFR is overexpressed in 40–80% of prostate cancer cells, and over-expression may be
`more common in African American men with prostate cancer [50]. Furthermore, preclinical
`data suggested a correlation of EGFR expression with Gleason sum and androgen
`independence [51]. In phase II studies of approximately 100 patients with castration-
`resistant disease evaluating the EGFR tyrosine kinase inhibitor gefitinib, minimal activity
`and no PSA responses were reported [52,53]. Gefitinib resistance may be related to
`overactivity of the PI3K/Akt pathway in prostate cancer, and thus combinations of agents
`that target multiple pathways may be more beneficial [54]. In an effort to overcome this
`resistance, trials combining EGFR or dual kinase inhibitors with other novel agents are in
`development. For example, gefitinib is currently being tested in combination with the Akt/
`mTOR inhibitor, everolimus, in a phase II trial as first-line therapy for metastatic CRPC.
`
`Prostate cancer cells express high levels of platelet-derived growth factor receptor
`(PDGFR), and signaling through this mechanism converges with the PI3K/Akt pathway
`which has been implicated in prostate cancer progression. Single-agent activity with the
`PDGFR inhibitor imatinib has been disappointing [55]; however, encouraging results in
`combination with weekly docetaxel have been reported in the phase I setting [56]. A phase
`II randomized trial of this combination compared with docetaxel alone is in progress, but
`early results from this study suggested a lack of improvement in median progression-free
`survival in men receiving docetaxel plus imatinib [57]. Plans to move forward with imatinib
`as a potential therapeutic agent for prostate cancer in the future are lacking.
`
`Another potential target in this family of receptors is the HER2/neu tyrosine kinase, whose
`expression has been shown to increase androgen receptor activation leading to prostate
`cancer growth and survival [58]. However, phase II studies using the anti-HER2 monoclonal
`antibody trastuzumab showed minimal efficacy in CRPC, perhaps due to a low frequency of
`HER2 over-expression [59]. Studies using the dual EGFR/HER2 small molecule inhibitor
`lapatinib in asymptomatic CRPC are now being conducted, but early results have shown
`PSA responses in only about 10% of participants [60].
`
`2.4. Apoptosis
`
`Apoptosis is regulated by pro-apoptotic and anti-apoptotic proteins that are recruited as a
`result of apoptotic stimuli such as DNA damage, chemo- and hormonal therapy, and
`irradiation. Bcl-2, an anti-apoptotic factor, is an attractive molecular target in the treatment
`of CRPC. In human prostate carcinoma cell lines as well as in clinical prostate cancer
`specimens, increased Bcl-2 expression induces the transition to androgen-independent cell
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 7
`
`growth [61], and confers resistance to many antineoplastic agents including taxanes [62].
`These findings suggest that Bcl-2 over-expression may mediate clinical resistance to both
`androgen deprivation and chemotherapy in prostate cancer patients.
`
`Oblimersen is a synthetic antisense oligonucleotide that hybridizes to bcl-2 mRNA and
`inhibits Bcl-2 protein expression [63]. In mice bearing xenograft tumors from androgen-
`independent human prostate cancer cell lines, oblimersen markedly enhanced the anti-tumor
`activity of docetaxel resulting in increased rates of complete tumor regression compared
`with animals treated with docetaxel alone [64]. Because docetaxel itself partially inactivates
`the Bcl-2 protein (by phosphorylation), the addition of oblimersen to docetaxel is a rational
`therapeutic strategy. To this end, a phase I/II study using oblimersen (given by continuous
`intravenous infusion on days 1–8) with docetaxel (on day 6) every 3 weeks in patients with
`CRPC showed that 14/27 men (52%) achieved PSA responses while 4/12 men (33%) with
`measurable disease achieved partial radiological responses [65]. Adverse events with this
`combination were myelosuppression (including febrile neutropenia), alopecia, fatigue,
`diarrhea, and nausea/vomiting. Toxicities specifically attributed to oblimersen were fever
`(beginning 2–3 days after drug initiation), aspartate aminotransferase elevations,
`hypophosphatemia, and deep vein thrombosis. A randomized phase II trial evaluating
`docetaxel (given on day 5) with or without oblimersen (by continuous intravenous infusion
`on days 1–7) in patients with metastatic CRPC was recently reported. Discouragingly, this
`study revealed that PSA responses were similar in the docetaxel–oblimersen arm and in the
`docetaxel-alone arm (46% and 37%, respectively), and partial radiological responses were
`also similar (18% and 24%, respectively) [66]. In addition, docetaxel–oblimersen was
`associated with an increased incidence of grade 3–4 fatigue, mucositis, and
`thrombocytopenia; and caused more major toxic events (40.7% versus 22.8%, respectively).
`
`AT-101 (R-gossypol acetate) is a polyphenolic compound derived from the cottonseed plant
`that inhibits the function of all Bcl-2 – related proteins (Bcl-2, Bcl-xL, Mcl-1, and Bcl-w)
`[67]. By blocking the binding of Bcl-2 family members with pro-apoptotic proteins and up-
`regulating specific pro-apoptotic factors, AT-101 lowers the threshold for cancer cells to
`undergo apoptosis [68]. Preclinically, AT-101 has shown anti-tumor activity in a variety of
`tumor types including prostate cancer [69]. A phase I/II study of oral AT-101 used alone
`was conducted in men with CRPC and no prior chemotherapy. In that study, the optimal
`dose was determined to be 20 mg/day for 21 out of 28 days, and common toxicities included
`diarrhea, fatigue, nausea, anorexia, and small bowel obstruction [70]. Two of 23 patients
`(9%) had a ≥50% PSA decline, but no patient achieved a radiological response. A second
`phase I/II study was performed by combining AT-101 (on days 1–3 of each cycle) with
`docetaxel (given every 3 weeks) in men with docetaxel-naïve CRPC. In that study, the
`optimal dose of AT-101 was found to be 40 mg twice daily on days 1–3 of each
`chemotherapy cycle, and adverse events of this combination included neutropenia,
`lymphopenia, fatigue, nausea, diarrhea, and hypophosphatemia [71]. Eight of nine patients
`treated at the optimal dose (89%) had a ≥50% PSA decline, and 2 of 6 patients with
`measurable disease (33%) had a partial radiologic response. A multicenter randomized
`phase II study evaluating docetaxel plus or minus oral AT-101 in the first-line treatment of
`metastatic CRPC is now underway.
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`2.5. IGF pathway
`
`Page 8
`
`The insulin-like growth factor type-1 receptor (IGF-1R) and its ligands may also play a key
`role in prostate cancer carcinogenesis through mechanisms that involve mitogenesis, anti-
`apoptosis, and cellular transformation. Epidemiological studies have shown that increased
`circulating insulin-like growth factor type-1 (IGF-1) levels and decreased insulin-like
`growth factor binding protein-3 (IGFBP-3) levels are associated with higher risk of
`developing prostate cancer [72]. Conversely, partial inactivation of the IGF-1R seems well
`tolerated and may protect individuals from prostate cancer. In addition, IGF-1R is often
`overexpressed in prostate tumors and can mediate prostate cancer cell proliferation and
`resistance to androgen ablation therapy [73,74]. A promising strategy to inhibit the function
`of the IGF-1R is the use of monoclonal antibodies that bind to the extracellular domain of
`this transmembrane receptor [75]. In prostate cancer cell lines as well as in xenograft
`models, such antibodies can inhibit growth of both androgen-dependent and -independent
`tumors [76,77].
`
`IMC-A12 (cixutumumab) is a fully human IgG1 monoclonal antibody that specifically
`targets the IGF-1R, inhibiting ligand binding and IGF signaling [78]. A phase II study of
`intravenous IMC-A12 (10 mg/kg every 2 weeks) used as monotherapy in men with
`asymptomatic metastatic CRPC was recently reported. In that study, 9 of 31 patients (29%)
`demonstrated lack of radiographic progression after 6 months of treatment, and an even
`greater number had PSA responses [79]. Adverse events related to this agent were fatigue,
`hyperglycemia (usually asymptomatic), thrombocytopenia, hyperkalemia, and pneumonia. A
`phase II study combining IMC-A12 (6 mg/kg on days 1, 8, and 15 of a 3-week cycle) with
`mitoxantrone in the second-line treatment of docetaxel-refractory metastatic CRPC is
`currently underway.
`
`CP-751,871 (figitumumab) is the second fully human anti-IGF-1R IgG2 monoclonal
`antibody to enter clinical trials [80]. A phase I study of intravenous CP-751,871 given in
`combination with docetaxel to men with metastatic CRPC has been completed. In that study,
`4 of 18 patients (22%) had a radiographic partial response to therapy, and an additional 2
`men (11%) had disease stabilization for >6 months [81]. Toxicities of this combination
`regimen were neutropenia (including neutropenic fever), diarrhea, and transient
`hyperglycemia. A phase II study of CP-751,871 (20 mg/kg on day 1 of a 21-day cycle)
`combined with docetaxel in men with chemotherapy-naïve (arm A) and docetaxel-resistant
`(arm B) CRPC is now open.
`
`2.6. Bone interface
`
`An emerging target with a prominent role in prostate cancer progression and development of
`bone metastases is endothelin-1, a peptide that also plays an important role in vascular tone
`[82]. Preclinical studies suggest that endothelin A receptors are overexpressed in prostate
`cancer, and higher tissue endothelin receptor levels in patients with prostate cancer correlate
`with advanced tumor stage, grade, and metastases [83]. Endothelin-1 is a potent
`vasoconstrictor, and antagonists have been developed for the treatment of pulmonary
`hypertension. In oncology, endothelin is likely to be involved in the paracrine signals
`between osteoblasts and prostate cancer cells that regulate the development of bone
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 9
`
`metastases and have been shown to influence cell growth and proliferation, regulate
`osteoblast activity, and inhibit apoptosis [84–86]. These observations suggest that this
`pathway may be a rational target for the interference of tumor-stromal interactions.
`
`Atrasentan is a highly selective endothelin A receptor antagonist and has been extensively
`tested in prostate cancer [87]. In phase II trials, a 10-mg dose of oral atrasentan was found to
`prolong time to progression compared with placebo in men with metastatic CRPC (196
`versus 129 days, respectively; P = .02) [88]. Adverse events with atrasentan were mild and
`related to vasomotor reactions including headache, rhinitis, flushing, and peripheral edema.
`In addition, favorable effects were seen in markers of bone deposition and resorption.
`However, in a placebo-controlled double-blind phase III trial involving 809 patients with
`metastatic CRPC, atrasentan (10 mg/day) did not reduce the risk of disease progression (P
`= .14), despite evidence of biologic effects on PSA and bone alkaline phosphatase [89]. A
`second phase III trial in non-metastatic CRPC that randomized 467 men to atrasentan and
`474 to placebo also failed to improve time to metastatic progression (P = .29) or overall
`survival [90]. A large cooperative group phase III clinical trial evaluating docetaxel with or
`without atrasentan as first-line therapy for metastatic CRPC is now underway. This study
`was fueled by promising results of early phase II trials evaluating this combination [91], and
`from preclinical data showing synergism between docetaxel and atrasentan in vitro and in
`vivo [92].
`
`A novel small molecule endothelin receptor inhibitor, zibotentan (ZD4054), has shown
`initial promising results [93]. In a phase II trial of zibotentan versus placebo in men with
`metastatic CRPC, this agent did not improve time to disease progression (the primary study
`endpoint) (P = .55); however overall survival was longer on the zibotentan arm (P = .01)
`[94]. Although survival was a secondary endpoint in that trial, this has led to the design of
`several ongoing placebo-controlled phase III clinical studies evaluating zibotentan either
`alone or in combination with docetaxel in patients with metastatic CRPC. A further phase III
`trial is investigating single-agent zibotentan in men with non-metastatic castration-resistant
`disease.
`
`2.7. RANK ligand inhibitors
`
`Interactions between tumor cells and the bone marrow microenvironment have been
`postulated as an additional important mechanism in the pathogenesis of bone metastasis.
`Tumor-associated cytokines have been shown to induce the expression of RANKL (the
`receptor activator of nuclear factor κB ligand), which binds and activates RANK which is
`found in osteoclasts [95]. Inhibition of the RANKL system has recently been the focus of
`much research and represents an evolving bone-targeted strategy. Among the approaches
`employed are monoclonal antibodies to RANKL and the use of recombinant osteoprotegerin
`(the natural decoy receptor of RANKL), both of which significantly inhibit osteoclastic
`function in vitro and in vivo [96].
`
`Denosumab, a fully human monoclonal antibody against RANKL, has entered clinical trials
`in prostate and breast cancers [97]. In a phase II randomized study evaluating 50 patients
`with metastatic prostate cancer, denosumab (180 mg subcutaneously every 4 weeks)
`produced a reduction in bone resorption over that of zoledronate as indicated by a lowering
`
`Cancer Lett. Author manuscript; available in PMC 2014 May 21.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`
`Antonarakis et al.
`
`Page 10
`
`of urinary N-telopeptide levels, and also resulted in less skeletal-related events (SREs) [98].
`A multi-center phase III double-blind study comparing denosumab with zoledronate in the
`prevention of SREs in patients with metastatic CRPC has recently completed accrual of 745
`men.
`
`2.8. Vitamin D analogue

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