213
Views
4
CrossRef citations to date
0
Altmetric
Review

The tumor microenvironment in prostate cancer: elucidating molecular pathways for therapy development

Pages 183-193 | Published online: 25 Jul 2012

Abstract

Mechanisms leading to the development of virulent prostate cancer are not confined to the cancer epithelial cell, but also involve the tumor microenvironment. Multiple signaling pathways exist between epithelial cells, stromal cells, and the extracellular matrix to support tumor progression from the primary site to regional lymph nodes and distant metastases. Prostate cancers preferentially metastasize to the skeleton, prompting considerable research effort into understanding the unique interaction between prostate cancer epithelial cells and the bone microenvironment. This effort has led to the discovery that signaling pathways involved in normal prostate and bone development become dysregulated in cancer. These pathways stimulate excessive cell growth and neovascularization, impart more invasive properties to epithelial cells, weaken antitumor immune surveillance, and promote the emergence of castrate-resistant disease. An improved understanding of the complex relationship between cancer epithelial cells and the organ-specific microenvironments with which they interact has created a powerful opportunity to develop novel therapies.

Introduction

For men in the United States and Europe, prostate cancer is the most commonly diagnosed nonskin cancer.Citation1Citation3 In the United States, prostate cancer accounts for 11% of cancer-related deaths in males, ranks second as a cause of cancer mortality in men, and incurs significant healthcare costs.Citation1,Citation4,Citation5 If the tumor is organ-confined at the time of diagnosis, prognosis is often favorable and treatment is curative for many patients.Citation6,Citation7 In contrast, metastatic prostate cancer portends a much worse prognosis.Citation7 Prostate cancers preferentially metastasize to the skeleton, and associated complications including bone pain, pathological fractures, and spinal cord compression are responsible for much of the morbidity of the disease.Citation8 The presence of bone metastases also confers a 5-year survival rate of only 25% and a median survival of approximately 40 months.Citation8 Prostate cancer cells are typically derived from the prostate epithelium and receive stimulation via the androgen receptor (AR) for continued proliferation and survival.Citation9,Citation10 For this reason, the initial treatment for metastatic or advanced prostate cancer is androgen deprivation therapy, consisting of either surgical (ie, orchiectomy) or medical castration (with luteinizing hormone-releasing analogs or antagonists).Citation11 Although androgen deprivation therapy is usually effective when initially used, relapse occurs after a median of 18–24 months with the appearance of castrate-resistant prostate cancer (CRPC).Citation12

A notable feature of the castrate-resistant phenotype is that, paradoxically, tumors continue to rely on androgen signaling for growth despite undetectable levels of circulating testosterone in blood.Citation13,Citation14 This occurs through a shift in reliance of the tumor on androgen endocrine sources (ie, gonads and adrenal glands) to autocrine/paracrine sources produced locally within the tumor microenvironment.Citation15 The detection of intratumoral androgen levels is now being incorporated into clinical research studies, but is not yet a standardized test. Importantly, intratumoral androgens can be inhibited by novel small-molecule therapeutics. For example, the drug abiraterone, which potently inhibits both endocrine and autocrine/paracrine testosterone biosynthesis by blocking the activity of the enzyme cytochrome P450 CYP17,Citation16 has been shown to improve survival in men with metastatic CRPC (mCRPC) after failure of the chemotherapeutic agent docetaxel.Citation17

Other treatment options for patients with mCRPC include cytotoxic chemotherapies and vaccine-based approaches. Both docetaxel and cabazitaxel have been shown to prolong survival in patients with mCRPC in randomized phase III studies.Citation18,Citation19 Presently, docetaxel remains the standard first-line therapy, followed by cabazitaxel in the second-line setting for patients whose cancers have progressed after receiving docetaxel therapy.Citation11 The autologous cancer vaccine, sipuleucel-T, was also approved in 2010 after it was shown to improve survival in patients with minimally symptomatic mCRPC.Citation11,Citation20

Despite considerable progress in the development of novel androgen-ablative, cytotoxic, and immunologic therapies, mCRPC remains incurable, and better treatments are needed. An improved understanding of the cellular and molecular mechanisms involved in prostate cancer progression has led to an increasing number of new anticancer drugs under development for the treatment of this disease.Citation21 Within the cellular microenvironment, molecular signaling between the tumor cells and surrounding cells that contribute to cancer growth are emerging as critical therapy targets. This review discusses the three principal tumor microenvironments involved in prostate cancer progression: the prostate gland, lymph node, and bone microenvironments; signaling pathways that impact the tumor microenvironment; and novel agents that modulate these pathways for potential therapy.

Tumor microenvironments

Tumors are no longer regarded as isolated masses of aberrantly proliferating epithelial cells. Rather, their properties depend on complex interactions between cancer epithelial cells and the surrounding stromal compartment within the tumor microenvironment. The stromal compartment is comprised of multiple nonmalignant cells such as fibroblasts, myofibroblasts, endothelial cells, and immune cells; growth factors; chemokines; cytokines; extracellular matrices (ECMs); and matrix-degrading enzymes.Citation22,Citation23 Crosstalk between the epithelial and stromal compartments promotes tumor progression by mechanisms such as remodeling ECM to enhance invasion, releasing soluble growth factors necessary for castrate-resistant growth, and stimulating angiogenesis.Citation24,Citation25 Surrounding tumor stromal cells, while not having the marked genetic instability of tumors, have been shown to undergo genetic alteration in response to the presence of a tumor; these may further sustain the malignant phenotype.Citation26Citation28

Prostate cancer has a prolonged natural history that can be conceptually divided into three stages. The first and most prolonged stage involves the initiation and development of organ-confined disease, estimated to take more than 15 years.Citation29 The second stage involves spreading into regional lymph nodes, and the third stage involves metastatic dissemination via both the lymphatic and blood circulations to the skeleton.Citation29 Each of these stages represents a unique tumor microenvironment in which the prostate cancer epithelial cells survive, adapt, and proliferate.Citation29

Prostate gland microenvironment

The normal prostate gland consists of prostatic ducts lined with epithelium and a stroma that consists mainly of smooth muscle cells with smaller numbers of fibroblasts, endothelial cells, and nerve cells.Citation22 In addition to their expression in epithelial cells, ARs are also abundantly expressed on multiple cell types in the normal prostate stroma, including smooth muscle cells and endothelial cells.Citation30 During normal development, prostate epithelial cells depend on signals from the stroma for migration and organ homeostasis.Citation30 In turn, signals from epithelial cells to prostate stromal cells appear to be required to maintain smooth muscle differentiation.Citation30,Citation31

In prostate cancer, stromal cells demonstrate an altered phenotype in which there is increased ECM remodeling, increased protease activity, increased angiogenesis, and an influx of inflammatory cells.Citation32 These cancer-activated stromal changes resemble the tissue alterations that accompany normal wound healing.Citation32 Altered stromal fibroblasts (referred to as myofibroblasts) present in both cancer and wound healing are morphologically distinct from normal fibroblasts and acquire some of the properties of smooth muscle cells,Citation23,Citation25,Citation33 including expression of vimentin and smooth muscle α-actin.Citation24 While myofibroblasts participate in the formation of granulation tissue during normal wound healing,Citation24,Citation34,Citation35 in tumors, they promote stromal reactivity and tumor cell proliferation.Citation35,Citation36 The importance of myofibroblasts is evidenced by their appearance within carcinoma in situ lesions of different epithelial tumors, suggesting they participate in early tumorigenesis.Citation25 In prostate cancer, myofibroblasts are detectable in the tissues surrounding the foci of prostate intraepithelial neoplasia, and myofibroblast activity increases with increasing prostate tumor grade.Citation37

Experimental studies demonstrate that myofibroblasts are not merely a passive reaction to the tumor, but also a source of signals that enhance tumorigenicity. For example, coculture experiments have shown that tumor-associated myofibroblasts stimulate growth and tumor formation from nontumorigenic prostate cell lines.Citation38,Citation39 Conversely, it has been found that nonactivated fibroblasts can promote the reversion of tumor cells to a more differentiated and slower-growing phenotype.Citation40 Other effects of myofibroblasts relevant to tumor progression include remodeling of the ECM, tumor neovascularization (angiogenesis), and the development of immunological tolerance to the tumor.Citation34

Lymph node microenvironment

Based on personal experience with numerous clinical trials at University of Texas MD Anderson Cancer Center (Houston, TX), radiographically enlarged lymph nodes are detected in only roughly 20% of patients with metastatic disease. Interestingly, however, molecular-pathologic analysis of lymph nodes from surgical specimens and autopsy studies suggest that nodal involvement without enlargement occurs much more frequently.Citation41,Citation42 The fact that a majority of prostate cancers spread to regional lymph nodes, but only a minority of nodes are pathologically enlarged, suggests that lymph nodes are not a preferred microenvironment for prostate cancer growth. Thus, while lymphadenopathy is an independent adverse prognostic indicator in prostate cancer, clinical sequelae from lymphadenopathy are relatively uncommon.Citation43

However, it has been postulated that because tumor cells within lymph nodes have disseminated from the original site of the cancer, these cells may possess a higher metastatic potential. In support of this, lymph node metastases show a relatively greater loss of cell adhesion molecules than primary tumors.Citation44 This phenomenon may be explained by epithelial-to-mesenchymal transition, a biologic process that commonly occurs during both normal embryogenesis and cancer progression when epithelial cells assume characteristics of mesenchymal cells, including greater migratory and invasive properties.Citation45,Citation46 A significant association also exists between vascular endothelial growth factor (VEGF) activity and the presence of lymph node metastases, suggesting a requirement for angiogenesis during prostate cancer tumor dissemination.Citation47 Interestingly, although the association between lymphangiogenesis (formation of new lymph vessels) and lymph node metastases is less clear,Citation47 lymphangiogenesis can occur in the absence of tumor involvement.Citation48 These data suggest that the primary tumor acts as a source of soluble stimuli (mainly growth factors in the VEGF family) that “prepare” the lymph node microenvironment for subsequent tumor involvement.Citation47

Bone microenvironment

The propensity of prostate cancer to metastasize to bone is one of the most striking examples of microenvironment-dependent tumor progression in human cancer.Citation8 The bone microenvironment has become an important focus of basic and clinical research efforts. In contrast to most other solid tumors that demonstrate osteolytic lesions (eg, breast and lung), prostate cancer bone metastases are typically osteoblastic.Citation49 Osteoblastic lesions are “bone-forming” lesions that occur through the remodeling of bone matrix by osteoblasts and osteoclasts. In normal bone, a continuous process of bone turnover maintains structural homeostasis through a dynamic balance between osteoblastic and osteoclastic activities. In prostate cancer, this balance is disrupted when prostate cancer epithelial cells express bone-specific proteins and soluble osteoblastic growth stimulatory factors that lead to the production of abnormal, unstructured bone.Citation49 For example, prostate cancer cells produce bone morphogenetic protein, β2 microglobulin, osteocalcin, bone sialoprotein, endothelin-1 (ET-1), transforming growth factor-β (TGF-β), VEGF, parathyroid hormone-related protein, and insulin-like growth factor-1.Citation49 Expression of many of these proteins is typically restricted to bone cells, but during prostate cancer progression, these proteins become aberrantly expressed by cancer epithelial cells, a phenomenon referred to as “osteomimicry.”Citation49Citation51

Another stromal pathway usurped by prostate cancers is the receptor activator of nuclear factor κβ (RANK) ligand pathway.Citation22 RANK ligand is normally expressed by osteoblasts and other stromal cells and functions in order to regulate osteoclastic activity by binding to RANK receptors on preosteoclasts, and stimulating them to mature into osteoclasts.Citation8,Citation52,Citation53 In prostate tumors, RANK ligand is either overexpressed directly by the tumor or induced indirectly through tumor activation of osteoblasts.Citation22 This event prompts an endless loop of increased bone turnover and increased tumor metastatic ability that may account for the central role of bone metastases in advanced prostate cancer.Citation22,Citation52 At the same time that prostate cancer epithelial cells induce tumor-promoting changes in the tumor microenvironment, prostate cancer epithelial cells may respond to growth factors secreted by osteoblasts and also present in bone matrix. Growth-promoting effects on prostate cancer epithelial cells have been demonstrated with conditioned medium from cultured osteoblastsCitation54Citation56 and in three-dimensional culture systems.Citation57 Interleukin-6 produced by osteoblasts (and by osteomimetic prostate cancer cells) is a cytokine important in normal bone turnover and has been shown to induce prostate cancer cell proliferation and induction of androgen-regulated genes normally dependent on the AR.Citation55,Citation58 Prostate cancer cells cultured in decellularized bone matrix show induction of genes regulating migration and invasive potential, suggesting that the bone matrix is also a significant source of growth factors.Citation59 All of these events further promote a chain of events leading to prostate cancer bone metastases.Citation22

Pathways for therapy development

Several classes of novel therapies that disrupt signaling pathways within tumor microenvironments are currently under investigation (). In contrast to cytotoxic therapies that principally target the epithelial cell (regardless of its anatomic location), these agents disrupt the “crosstalk” between epithelial cells, stromal cells, and the ECM necessary for prostate cancer progression and metastases. Since skeletal metastases are the principal cause of morbidity and mortality from prostate cancer, the majority of clinical trials evaluating novel therapies enroll patients with advanced disease. Thus, the bone tumor microenvironment is most often being evaluated. There is emerging evidence, however, that high-risk localized tumors (ie, those tumors with a high chance of micrometastatic spread to lymph nodes) acquire the osteomimetic characteristics of skeletal metastases.Citation15,Citation49 To test the hypothesis that signaling pathways involved in the development of high-risk localized disease are shared with skeletal metastases, there are a growing number of neoadjuvant trials for high-risk patients.

Table 1 Therapeutic agents targeting the prostate cancer microenvironment

The first class of therapeutic agents to support the hypothesis that targeting the tumor microenvironment could benefit patients clinically is radiopharmaceuticals. Radiopharmaceuticals (eg, strontium-89, samarium-153, rhenium-186, and radium-223) concentrate at sites of bone-forming activity to deliver a potent dose of radiation. Radiopharmaceuticals have consistently been shown to palliate bone pain, and in some studies have demonstrated a survival benefit.Citation60 This benefit is attributed not only to a direct cytotoxic effect on cancer epithelial cells, but also the suppression of pathologic osteoblast and osteoclast activities (ie, suppression of the chain of events). Despite their success, radiopharmaceuticals lack target specificity, as evidenced by dose-limiting side effects such as bone marrow suppression. The novel small-molecule and immune-based therapeutics discussed below have the potential to provide greater efficacy, specificity, and safety.

Targeting signaling pathways mediated by cell adhesion proteins

The ECM is made up of many types of macromolecules, and plays an important role in maintaining normal cellular behavior and tissue architecture. Cell adhesion molecules and tight junction proteins on cell surfaces (eg, β1 integrins and E-cadherin) maintain cell–cell contacts and contact with the ECM.Citation23,Citation61 Gap junctions allow passive diffusion of small signaling molecules between adjacent cells.Citation61 In normal tissues, the basement membrane, a specialized form of ECM, separates the epithelium from stromal cells.Citation23 The ECM is essential for maintaining cell shape and behavior, and for maintaining the correct polarity of epithelial cells by establishing basal and apical surfaces.Citation61 Correct function of cell adhesion is necessary for organ homeostasis and, additionally, has been shown to be important in suppressing tumor formation.Citation61 In contrast, alterations in the ECM occur in tumorigenesis and can profoundly affect the malignant potential of epithelial cells.

The integrins have attracted much interest as potential therapeutic targets in cancer. Patterns of integrin expression are characteristically altered in prostate tumors, with marked changes in subunit composition.Citation44,Citation62 Integrins become distributed over the entire cell surface through disruption of their normal localization on the basal surface.Citation25,Citation44 Integrins signal by recruiting focal adhesion kinase and Src kinase. These, in turn, activate components that affect cell growth, adhesion, motility, and cell survival.Citation25,Citation63 Of these, the focal adhesion kinase–Src complex is important in promoting angiogenesis and protease-associated tumor metastasis.Citation63 Several integrin inhibitors, including the peptide cilengitide as well as integrin-blocking monoclonal antibodies CNTO 95 and MEDI-522, are in clinical development for prostate cancer.Citation62

Targeting signaling pathways mediated by myofibroblasts

Myofibroblasts present in cancer-activated stromal tissue are the source of many of the signals that direct tumor cell proliferation and survival.Citation25,Citation33 The importance of stromal cells in tumor progression has been demonstrated in coculture experiments.Citation39,Citation64 For example, the fibroblast growth factor signaling pathway (FGF/FGF receptor) mediates communication between prostate epithelium and stromal cells, including myofibroblasts.Citation25,Citation65 Signaling via FGF/FGF receptor leads to downstream activation of other prosurvival signal transduction pathways, including extracellular signal-regulated kinase, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase/Akt.Citation65 Signaling via phosphatidylinositol 3-kinase/Akt has been shown to have synergistic effects with AR signaling in the prostate epithelium.Citation65 Small-molecule kinase inhibitors such as SU5402 and dovitinib (TKI258), which inhibit FGF/FGF receptor signaling, may have potential as anticancer agents.Citation25

Targeting signaling pathways mediated by the AR

Several mechanisms are responsible for the castrate-resistant phenotype.Citation66,Citation67 One mechanism involves sensitization of the AR to lower levels of androgen present within tumors. This “androgen hypersensitivity” can occur from either a mutation at the ligand-binding site of the AR or from amplification of the gene encoding the AR. A second mechanism involves mutations in the AR that result in reduced ligand specificity.Citation66 These mutated “promiscuous” ARs can be inappropriately activated by estrogens, progestins, growth factor tyrosine kinases, and other oncogenic signaling molecules.Citation66,Citation68 A third mechanism involves ligand-independent activation of AR signaling. A variety of cytokines and growth factors present in bone and prostate microenvironments have the ability to directly activate AR in this manner, including insulin-like growth factor, keratinocyte growth factor, epidermal growth factor, and interleukin-6.Citation67,Citation68 Finally, a fourth mechanism involves truly AR-independent pathways. In these cases of castrate-resistant disease, there is often a complete loss of AR expression. Tumor progression is then sustained by entirely new growth-promoting pathways, including overexpression of B-cell lymphoma 2 and/or activation of Akt.Citation66,Citation67

Several agents have been developed to overcome these different mechanisms of castrate resistance. TAK-700, a novel cytochrome P450 CYP17 inhibitor, depletes intratumoral androgens to overcome AR hypersensitivity and AR promiscuity (upstream progestins are also inhibited). The investigational AR antagonist, MDV3100, blocks nuclear translocation and DNA binding of the AR and thus may prevent ligand-independent activation of the AR. Oblimersen sodium, an antisense oligonucleotide that targets B-cell lymphoma 2, initially exhibited promise in combination with docetaxel, as a therapy designed to overcome AR-independent survival pathways in mCRPC.Citation67 However, this therapeutic combination failed to meet the primary endpoints (prostate-specific antigen response, >30%; major toxic event rate, <45%) of a phase II study suggesting limited utility.Citation69

Targeting crosstalk between cancer epithelial and stromal cells

Endothelin A receptor ET-1 antagonists

Atrasentan is an antagonist of the endothelin A receptor ET-1 and has been studied in clinical trials for the treatment of CRPC for both metastatic disease and biochemical relapse with rising prostate-specific antigen only.Citation70,Citation71 Although atrasentan did not reduce the risk of disease progression in either disease state, it has demonstrated favorable effects on bone biomarkers (eg, reduction in bone-specific alkaline phosphatase), stimulating continued interest in blocking ET-1 as therapy for patients with advanced disease.Citation71 A second ET-1 antagonist, zibotentan (ZD4054), had shown a survival advantage in the treatment of patients with symptomatic mCRPC, but in a phase III trial that evaluated zibotentan added to the standard of care in 594 patients with mCRPC, there was no significant improvement in the primary endpoint of OS.Citation72 Denosumab, a monoclonal antibody against RANK ligand, has recently been shown to reduce skeletal-related events in patients with mCRPC.Citation73

Src kinase inhibitors

Src kinase is a nonreceptor kinase of considerable importance in prostate cancer progression. Src, an essential downstream signal transducer for many cell surface receptors, including epidermal growth factor receptor, platelet-derived growth factor receptor, VEGF receptor, c-Met, and integrins, is of specific importance in the regulation of bone homeostasis. In vitro and in vivo experiments with osteoblasts and osteoclasts with targeted disruptions of Src suggest that this kinase may function by positively regulating osteoclasts and negatively regulating osteoblasts.Citation74 Aberrant expression and/or activity of Src kinases occur in many advanced-stage tumors, including prostate cancer.Citation74,Citation75 Interestingly, elevated Src activity is correlated with decreased AR activity in prostate tumor specimens, further supporting a role for Src in the development of castration resistance and the potential benefit of utilizing Src inhibitors for therapy of advanced disease.Citation76

Tyrosine kinase inhibitors active against Src are being studied for the treatment of mCRPC. Three dual Src/Bcr-Abl (breakpoint cluster region-Abelson) inhibitors are currently under clinical development,Citation8 and dasatinib monotherapy has been shown in phase II studies to demonstrate a favorable effect on preventing progression of bone metastases and decreasing markers of bone metabolism.Citation71,Citation77 A randomized, phase III study comparing docetaxel with or without dasatinib in patients with mCRPC has now completed enrollment, and data are pending.Citation78 Two other dual Src/Bcr-Abl (breakpoint cluster region-Abelson) inhibitors, saracatinib (AZD0530) and bosutinib (SKI-606), are in early stage evaluation for the treatment of prostate cancer bone metastases.Citation8

TGF-β inhibitors

TGF-β has multiple functions which can be classified as either growth promoting or growth suppressive, depending on the tissue and cellular context.Citation79 Experiments with conditional inactivation of its receptor, TβRII, show that stromal cells (fibroblasts) require TGF-β in order to exert their normal effects in suppressing epithelial proliferation.Citation80 In benign prostate epithelium, TGF-β maintains cellular differentiation, inhibits cell proliferation, and induces cell cycle arrest and/or apoptosis.Citation81 Stimulation of fibroblasts by TGF-β induces genes specific for tissue remodeling, including myofibroblast-related factors, ECM remodeling factors, and growth factors.Citation82 In prostate epithelium, crosstalk between TGF-β and AR is necessary for cellular apoptosis in response to androgen withdrawal.Citation79,Citation83 Although TGF-β maintains physiologic homeostasis in normal tissues, its growth-promoting effects predominate in the malignant state.

TGF-β is frequently upregulated in cancer cells, and it has been proposed that this, in turn, leads to the differentiation of fibroblasts to myofibroblasts and other properties of reactive stroma.Citation24 Other tumorigenic effects of TGF-β include neovascularization, ECM degradation, and suppression of tumor-specific immune responses.Citation81 In established prostate tumors, stromal cell promotion of tumor growth has been shown to be dependent on the presence of TGF-β signaling.Citation82 An important role of TGF-β is also suggested by evidence that serum levels of TGF-β are usually elevated in patients with advanced prostate cancer.Citation81

Several approaches to inhibiting TGF-β signaling are presently being developed. These include monoclonal antibodies, antisense oligonucleotides, and small-molecule tyrosine kinase inhibitors. All have demonstrated high activity in preclinical models of prostate cancer, and are expected to enter human clinical trials in the near future.Citation81

Angiogenesis inhibitors

Angiogenesis is the process of new blood vessel formation. In normal tissues, angiogenesis occurs during growth, development, and wound repair. Angiogenesis is required by all solid tumors to sustain the expanding tumor mass.Citation84 The tumor and stroma promote angiogenesis primarily through ligand-receptor pathways mediated by soluble angiogenic factors. Examples of these include VEGF/VEGF receptor, FGF/FGF receptor, platelet-derived growth factor/platelet-derived growth factor receptor, and TGF-β/TGF-β receptor. Although recent studies with two potent angiogenesis inhibitors – the anti-VEGF monoclonal antibody bevacizumab and the multi-tyrosine kinase inhibitor sunitinib – failed to show any survival benefit in mCRPC, there is continued interest in developing these agents as therapy. For example, aflibercept (VEGF Trap) is a recombinant fusion protein of the extracellular domains of both VEGF receptor-1 and VEGF receptor-2 and the Fc portion of human immunoglobulin G1.Citation7 Single-agent activity has been demonstrated in phase II trials in several cancer types.Citation85Citation88 Clinical trials of combination chemotherapy with aflibercept are also underway. A phase III trial of aflibercept in combination with docetaxel and prednisone in men with mCRPCCitation89 has completed accrual and is pending final analysis.Citation7 A phase III combination trial with irinotecan/fluorouracil/folinic acid for second-line treatment of metastatic colorectal cancer has recently been reported.Citation90,Citation91 Other approaches to blocking tumor angiogenesis include cediranib (AZD-2171), a kinase inhibitor for all three VEGF receptors, undergoing clinical trials for a variety of cancer types. Interim results of a phase II study of cediranib combined with prednisone in mCRPC have demonstrated partial responses to therapy and evidence of tumor regression in many other patients.Citation92

The drug lenalidomide has multiple effects that may contribute to its antitumor properties. Lenalidomide demonstrates antiangiogenic properties and also induces immunity through enhancing T cell stimulation and inhibiting regulatory T cells. The immunomodulatory effects of lenalidomide are implicated in its efficacy against certain hematologic malignancies such as myelodysplastic syndrome, multiple myeloma, and chronic lymphocytic leukemia. Lenalidomide also has synergistic effects with docetaxel in preclinical models of prostate cancer.Citation93 Despite this, however, a phase III trial of lenalidomide plus docetaxel and prednisone in CRPC was discontinued in late 2011 due to lack of treatment benefit versus docetaxel and prednisone alone.Citation94 Lenalidomide is still receiving attention as a possible adjuvant therapy to cancer vaccines in the treatment of prostate cancer.Citation93

Hepatocyte growth factor (HGF)/c-Met signaling inhibitors

The c-Met receptor is present on most cell types. Activation by HGF, its only known ligand, transduces multiple activities in the cell, including motility, proliferation, survival, and morphogenesis.Citation95 Of particular relevance, the c-Met pathway activates a program of cell dissociation, cell motility, and protease production, which can permit tumor cell invasion and cell scattering that may be linked to metastasis.Citation96 Tumors of epithelial origin frequently overexpress c-Met, resulting in increased responsiveness to HGF produced by stromal cells.Citation97 Mutations in the gene encoding c-Met, resulting in delayed downregulation of signaling, have been found in human cancers.Citation96 Monoclonal antibodies against human HGF have been developed and shown to neutralize HGF in vivo and to have antitumor activity.Citation96,Citation98 A second agent, BMS-777607, which targets c-Met is currently in preclinical development. BMS-777607 is an orally available kinase inhibitor that selectively neutralizes HGF activity and abolishes its effect on cell scattering and motility.Citation99 Also in clinical trials is cabozantinib (XL-184), a kinase inhibitor with dual specificity for c-Met and VEGF receptor-2. Results of a phase II clinical trial in patients with mCRPC and progressive disease have recently been reported in which cabozantinib was shown to produce overall disease control in many of the enrolled patients.Citation100 Responses were characterized by reductions in pain and improvements in bone scans.Citation100

Agents that affect immunity

Host immune responses are capable of suppressing tumors. Similar to other tumor types, prostate tumors become infiltrated by T lymphocytes, consistent with the initial stages of a cell-mediated immune response.Citation101 However, this immune response is generally ineffective due to loss or downregulation of cell-surface major histocompatibility complex,Citation102 or the secretion of immune-suppressive factors including TGF-β and interleukin-10.Citation101 Tumor stroma interactions may potentially affect the immune response. There is evidence that myofibroblasts act as a physical barrier between tumor cells and immune cells.Citation34 In addition, TGF-β has been shown to reduce the proliferation and repress the cytotoxic activity of CD8 T lymphocytes, and to inhibit natural killer cell function.Citation103 Of particular relevance in prostate cancer, androgens have been shown to demonstrate an immunosuppressive effect, and an improvement in antitumor response occurs after androgen withdrawal therapy.Citation104 It has been suggested that immunotherapy for prostate cancer may be most effective in patients with early-stage disease and/or with low tumor burden. This is because a slow-growing tumor allows for sufficient time to build an optimal immune response; for example, by booster vaccination.Citation101 Several forms of immunotherapy are currently in development for prostate cancer.Citation101 Monoclonal antibodies, such as ipilimumab, block cytotoxic T cell lymphocyte-associated protein 4, resulting in nonspecific T cell activation and enhanced antitumor immunity. Ipilimumab is presently being tested in both androgen-dependent and castrate-resistant metastatic disease.Citation105,Citation106 Other vaccines “prime” immunity against prostate tumors by incorporating whole cell vaccines (based on prostate cancer cell lines) that can be genetically engineered to secrete immunostimulatory cytokines. GVAX®, perhaps the best studied of these, is a whole-cell vaccine consisting of allogeneic prostate cancer cell lines that have been transduced with granulocyte-macrophage colony-stimulating factor complementary DNA. While recently completed clinical trials failed to demonstrate survival benefit for GVAX in mCRPC, a second experimental vaccine based on similar cell lines but engineered to express interleukin-2 and interferon-β, is also being studied in patients with mCRPC.Citation101

Viral vectors based on vaccinia or fowlpox cause host cells to express modified prostate-specific antigen and immune costimulatory molecules, which then induce an antitumor immune response. PROSTVAC® contains a gene for prostate-specific antigen that has been modified to enhance its immunogenicity. In addition, it encodes three modified proteins involved in the T cell costimulation pathway.Citation101 Another approach to therapeutic vaccines, autologous dendritic cell vaccines (such as the currently licensed sipuleucel- T), are used to stimulate the patient’s dendritic cells with prostate-specific proteins in vitro. Reintroduced dendritic cells may then activate an effective T cell response capable of eradicating the tumor.

Conclusion

Experimental studies on the biological mechanisms of prostate cancer have revealed extensive signaling pathways that provide communication between cancer epithelial cells, stromal cells, and the ECM within tumor microenvironments. These signaling pathways are necessary for prostate cancer progression. Many novel therapeutic agents under development for prostate cancer are designed to counter these signaling pathways, including AR inhibitors, angiogenesis inhibitors, osteoclast inhibitors, stromal inhibitors, and vaccines. The promise of these therapies is improved efficacy, specificity, and safety. The recent approvals of abiraterone and sipuleucel-T illustrate the dividends of translational research and indicate real progress towards curing this disease.

Disclosure

Medical editorial assistance for the writing of this article was provided by Susan DePetris of Phase Five Communications Inc, funded by sanofi-aventis US LLC, A SANOFI Company. The author retained full editorial control over the content of the manuscript, and received no compensation from any party for their work.

References

  • American Cancer SocietyCancer Facts and Figures 2010Atlanta, GAAmerican Cancer Society2010
  • JemalASiegelRXuJWardECancer statistics, 2010CA Cancer J Clin201060527730020610543
  • FerlayJParkinDMSteliarova-FoucherEEstimates of cancer incidence and mortality in Europe in 2008Eur J Cancer201046476578120116997
  • RoehrbornCGBlackLKThe economic burden of prostate cancerBJU Int2011108680681321884356
  • SiegelRWardEBrawleyOJemalACancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deathsCA Cancer J Clin201161421223621685461
  • WalczakJRCarducciMAProstate cancer: a practical approach to current management of recurrent diseaseMayo Clin Proc200782224324917290734
  • ChiKNBjartellADearnaleyDCastration-resistant prostate cancer: from new pathophysiology to new treatment targetsEur Urol200956459460519560857
  • SturgeJCaleyMPWaxmanJBone metastasis in prostate cancer: emerging therapeutic strategiesNat Rev Clin Oncol20118635736821556025
  • HenshallSMQuinnDILeeCSAltered expression of androgen receptor in the malignant epithelium and adjacent stroma is associated with early relapse in prostate cancerCancer Res200161242342711212224
  • MassieCELynchARamos-MontoyaAThe androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesisEMBO J201130132719273321602788
  • National Comprehensive Cancer NetworkClinical practice guidelines in oncology: prostate cancer guidelines (version 2. 2011)2011 Available from: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp#siteAccessed November 20, 2010
  • SerugaBOcanaATannockIFDrug resistance in metastatic castration-resistant prostate cancerNat Rev Clin Oncol201181122320859283
  • MostaghelEAPageSTLinDWIntraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancerCancer Res200767105033504117510436
  • MontgomeryRBMostaghelEAVessellaRMaintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growthCancer Res200868114447445418519708
  • EfstathiouELogothetisCJA new therapy paradigm for prostate cancer founded on clinical observationsClin Cancer Res20101641100110720145177
  • AngJEOlmosDde BonoJSCYP17 blockade by abiraterone: further evidence for frequent continued hormone-dependence in castration-resistant prostate cancerBr J Cancer2009100567167519223900
  • de BonoJSLogothetisCJMolinaAAbiraterone and increased survival in metastatic prostate cancerN Engl J Med2011364211995200521612468
  • TannockIFde WitRBerryWRDocetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancerN Engl J Med2004351151502151215470213
  • de BonoJSOudardSOzgurogluMPrednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trialLancet201037697471147115420888992
  • KantoffPWHiganoCSShoreNDSipuleucel-T immunotherapy for castration-resistant prostate cancerN Engl J Med2010363541142220818862
  • SmallEJde BonoJSProstate cancer: evolution or revolution?J Clin Oncol201129273595359821859996
  • JossonSMatsuokaYChungLWZhauHEWangRTumor-stroma co-evolution in prostate cancer progression and metastasisSemin Cell Dev Biol2010211263219948237
  • JoyceJATherapeutic targeting of the tumor microenvironmentCancer Cell20057651352015950901
  • TuxhornJAAyalaGERowleyDRReactive stroma in prostate cancer progressionJ Urol200116662472248311696814
  • KarlouMTzelepiVEfstathiouETherapeutic targeting of the prostate cancer microenvironmentNat Rev Urol20107949450920818327
  • DudleyACShihSCCliffeARHidaKKlagsbrunMAttenuated p53 activation in tumour-associated stromal cells accompanies decreased sensitivity to etoposide and vincristineBr J Cancer200899111812518594537
  • HillRSongYCardiffRDVan DykeTSelective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesisCell200512361001101116360031
  • KiarisHChatzistamouITrimisGFrangou-PlemmenouMPafiti-KondiAKalofoutisAEvidence for nonautonomous effect of p53 tumor suppressor in carcinogenesisCancer Res20056551627163015753354
  • JohanssonJEHolmbergLJohanssonSBergstromRAdamiHOFifteen-year survival in prostate cancer. A prospective, population-based study in SwedenJAMA199727764674719020270
  • CunhaGRHaywardSWDahiyaRFosterBASmooth muscle-epithelial interactions in normal and neoplastic prostatic developmentActa Anat (Basel)1996155163728811117
  • ChungLWZhauHERoJYMorphologic and biochemical alterations in rat prostatic tumors induced by fetal urogenital sinus mesenchymeProstate19901721651742399192
  • RowleyDRWhat might a stromal response mean to prostate cancer progression?Cancer Metastasis Rev 1998–1999174411419
  • De WeverODemetterPMareelMBrackeMStromal myofibroblasts are drivers of invasive cancer growthInt J Cancer2008123102229223818777559
  • DesmouliereAGuyotCGabbianiGThe stroma reaction myofibroblast: a key player in the control of tumor cell behaviorInt J Dev Biol2004485–650951715349825
  • PowellDWMyofibroblasts: paracrine cells important in health and diseaseTrans Am Clin Climatol Assoc200011127129310881346
  • AlbiniASpornMBThe tumour microenvironment as a target for chemopreventionNat Rev Cancer20077213914717218951
  • TuxhornJAAyalaGESmithMJSmithVCDangTDRowleyDRReactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodelingClin Cancer Res2002892912292312231536
  • BarclayWWWoodruffRDHallMCCramerSDA system for studying epithelial-stromal interactions reveals distinct inductive abilities of stromal cells from benign prostatic hyperplasia and prostate cancerEndocrinology20051461131815471963
  • OlumiAFGrossfeldGDHaywardSWCarrollPRTlstyTDCunhaGRCarcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epitheliumCancer Res199959195002501110519415
  • HayashiNCunhaGRMesenchyme-induced changes in the neoplastic characteristics of the Dunning prostatic adenocarcinomaCancer Res19915118492449301893381
  • MiyakeHHaraIKurahashiTInoueTAEtoHFujisawaMQuantitative detection of micrometastases in pelvic lymph nodes in patients with clinically localized prostate cancer by real-time reverse transcriptase-PCRClin Cancer Res20071341192119717317829
  • BubendorfLSchopferAWagnerUMetastatic patterns of prostate cancer: an autopsy study of 1589 patientsHum Pathol200031557858310836297
  • ChengLBergstralhEJChevilleJCCancer volume of lymph node metastasis predicts progression in prostate cancerAm J Surg Pathol19982212149115009850175
  • Pontes-JuniorJReisSTDall’OglioMEvaluation of the expression of integrins and cell adhesion molecules through tissue microarray in lymph node metastases of prostate cancerJ Carcinog20098319240373
  • HayEDAn overview of epithelio-mesenchymal transformationActa Anat (Basel)199515418208714286
  • GuarinoMRubinoBBallabioGThe role of epithelial-mesenchymal transition in cancer pathologyPathology200739330531817558857
  • DattaKMudersMZhangHTindallDJMechanism of lymph node metastasis in prostate cancerFuture Oncol20106582383620465393
  • CaoYOpinion: emerging mechanisms of tumour lymphangiogenesis and lymphaticmetastasisNat Rev Cancer20055973574316079909
  • KoenemanKSYeungFChungLWOsteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environmentProstate199939424626110344214
  • LecroneVLiWDevollRELogothetisCFarach-CarsonMCCalcium signals in prostate cancer cells: specific activation by bone-matrix proteinsCell Calcium2000271354210726209
  • ThomasRTrueLDBassukJALangePHVessellaRLDifferential expression of osteonectin/SPARC during human prostate cancer progressionClin Cancer Res2000631140114910741745
  • CastellanoDSepulvedaJMGarcia-EscobarIRodriguez-AntolinASundlovACortes-FunesHThe role of RANK-ligand inhibition in cancer: the story of denosumabOncologist201116213614521285392
  • KellerETBrownJProstate cancer bone metastases promote both osteolytic and osteoblastic activityJ Cell Biochem200491471872914991763
  • LangSHMillerWRHabibFKStimulation of human prostate cancer cell lines by factors present in human osteoblast-like cells but not in bone marrowProstate19952752872937479396
  • BlaszczykNMasriBAMawjiNROsteoblast-derived factors induce androgen-independent proliferation and expression of prostate-specific antigen in human prostate cancer cellsClin Cancer Res20041051860186915014041
  • LiYSikesRAMalaebBSOsteoblasts can stimulate prostate cancer growth and transcriptionally down-regulate PSA expression in cell line modelsUrol Oncol201129680280820451417
  • SungSYHsiehCLLawACoevolution of prostate cancer and bone stroma in three-dimensional coculture: implications for cancer growth and metastasisCancer Res2008682399961000319047182
  • LuYZhangJDaiJOsteoblasts induce prostate cancer proliferation and PSA expression through interleukin-6-mediated activation of the androgen receptorClin Exp Metastasis200421539940815672864
  • ReichertJCQuentVMBurkeLJStansfieldSHClementsJAHutmacherDWMineralized human primary osteoblast matrices as a model system to analyse interactions of prostate cancer cells with the bone microenvironmentBiomaterials201031317928793620688384
  • ParkerCHeinrichDO’SullivanJMOverall survival benefit of radium-223 chloride (Alpharadin™) in the treatment of patients with symptomatic bone metastases in castration-resistant prostate cancer (CRPC): a phase III randomized trial (ALSYMPCA)Paper presented at: 2011 European Multidisciplinary Cancer CongressSeptember 24–27, 2011Stockholm, Sweden
  • BissellMJRadiskyDPutting tumours in contextNat Rev Cancer200111465411900251
  • GoelHLLiJKoganSLanguinoLRIntegrins in prostate cancer progressionEndocr Relat Cancer200815365766418524948
  • MitraSKSchlaepferDDIntegrin-regulated FAK-Src signaling in normal and cancer cellsCurr Opin Cell Biol200618551652316919435
  • ThalmannGNRheeHSikesRAHuman prostate fibroblasts induce growth and confer castration resistance and metastatic potential in LNCaP CellsEur Urol201058116217119747763
  • Abate-ShenCShenMMFGF signaling in prostate tumorigenesis – new insights into epithelial-stromal interactionsCancer Cell200712649549718068626
  • DuttSSGaoACMolecular mechanisms of castration-resistant prostate cancer progressionFuture Oncol2009591403141319903068
  • PientaKJBradleyDMechanisms underlying the development of androgen-independent prostate cancerClin Cancer Res20061261665167116551847
  • AntonarakisESArmstrongAJEvolving standards in the treatment of docetaxel-refractory castration-resistant prostate cancerProstate Cancer Prostatic Dis201114319220521577234
  • SternbergCNDumezHVan PoppelHDocetaxel plus oblimersen sodium (Bcl-2 antisense oligonucleotide): an EORTC multicenter, randomized phase II study in patients with castration-resistant prostate cancerAnn Oncol20092071264126919297314
  • CarducciMASaadFAbrahamssonPAA phase 3 randomized controlled trial of the efficacy and safety of atrasentan in men with metastatic hormone-refractory prostate cancerCancer200711091959196617886253
  • VishnuPTanWWUpdate on options for treatment of metastatic castration-resistant prostate cancerOnco Targets Ther20103395120616956
  • AstraZeneca halts phase III trial of ZIBOTENTAN in non-metastatic castrate resistant prostate cancer [press release]LondonAstraZeneca272011
  • FizaziKCarducciMSmithMDenosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind studyLancet2011377976881382221353695
  • FizaziKThe role of Src in prostate cancerAnn Oncol200718111765177317426060
  • SaadFSrc as a therapeutic target in men with prostate cancer and bone metastasesBJU Int2009103443444019154462
  • MendirattaPMostaghelEGuinneyJGenomic strategy for targeting therapy in castration-resistant prostate cancerJ Clin Oncol200927122022202919289629
  • AraujoJCMathewPArmstrongAJDasatinib combined with docetaxel for castration-resistant prostate cancer: results from a phase 1–2 studyCancer20121181637121976132
  • Bristol-Myers SquibbRandomized study comparing docetaxel plus dasatinib to docetaxel plus placebo in castration-resistant prostate cancer (READY)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2008 [updated April 30, 2012]. Available from: http://clinicaltrials.gov/show/NCT00744497. NLM Identifier: NCT00744497Accessed December 21, 2011.
  • DanielpourDFunctions and regulation of transforming growth factor-beta (TGF-beta) in the prostateEur J Cancer200541684685715808954
  • BhowmickNAChytilAPliethDTGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epitheliaScience2004303565984885114764882
  • JonesEPuHKyprianouNTargeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progressionExpert Opin Ther Targets200913222723419236240
  • VeronaEVElkahlounAGYangJBandyopadhyayAYehITSunLZTransforming growth factor-beta signaling in prostate stromal cells supports prostate carcinoma growth by up-regulating stromal genes related to tissue remodelingCancer Res200767125737574617575140
  • SongKWangHKrebsTLKimSJDanielpourDAndrogenic control of transforming growth factor-beta signaling in prostate epithelial cells through transcriptional suppression of transforming growth factor-beta receptor IICancer Res200868198173818218829577
  • MuellerMMFusenigNEFriends or foes – bipolar effects of the tumour stroma in cancerNat Rev Cancer200441183984915516957
  • MackayHJBuckanovichRJHirteHA phase II study single agent of aflibercept (VEGF Trap) in patients with recurrent or metastatic gynecologic carcinosarcomas and uterine leiomyosarcoma. A trial of the Princess Margaret Hospital, Chicago and California Cancer Phase II ConsortiaGynecol Oncol2012125113614022138373
  • ColomboNMangiliGMammolitiSA phase II study of aflibercept in patients with advanced epithelial ovarian cancer and symptomatic malignant ascitesGynecol Oncol20121251424722112608
  • de GrootJFLambornKRChangSMPhase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium studyJ Clin Oncol201129192689269521606416
  • TwardowskiPStadlerWMFrankelPPhase II study of aflibercept (VEGF-Trap) in patients with recurrent or metastatic urothelial cancer, a California Cancer Consortium TrialUrology201076492392620646741
  • Sanofi-Aventis; Regeneron PharmaceuticalsAflibercept in combination with docetaxel in metastatic androgen independent prostate cancer (VENICE)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2007 [updated April 30, 2012]. Available from: http://clinicaltrials.gov/show/NCT00519285. NLM Identifier: NCT00519285Accessed December 21, 2011
  • Van CutsemETaberneroJLakomyRIntravenous (IV) aflibercept versus placebo in combination with irinotecan/5-FU (FOLFIRI) for second-line treatment of metastatic colorectal cancer (mCRC): results of a multinational phase III trial (EFC10262-VELOUR) [abstract]Ann Oncol201122Suppl 5v18
  • Van CutsemEPrenenHGuillen-PonceCA phase I/II, open-label, randomised study of BIBF 1120 plus mFOLFOX6 compared to bevacizumab plus mFOLFOX6 in patients with metastatic colorectal cancerPaper presented at: 2011 European Multidisciplinary Cancer Congress2011 September 23–27Stockholm, Sweden.
  • AdelergDKarakunnelJJGulleyJLA phase II study of cediranib in post-docetaxel castration-resistant prostate cancerPaper presented at: American Society of Clinical Oncology 2010 Genitourinary Cancers SymposiumMarch 5–7, 2010San Francisco, CA.
  • DalgleishAGalustianCThe potential of immunomodulatory drugs in the treatment of solid tumorsFuture Oncol2010691479148420919830
  • Celgene to discontinue docetaxel and prednisone plus lenalidomide phase III trial on CRPC [press release]BoudryCelgene International Sarl11232011
  • ZarnegarRMichalopoulosGKThe many faces of hepatocyte growth factor: from hepatopoiesis to hematopoiesisJ Cell Biol19951295117711807775566
  • PeruzziBBottaroDPTargeting the c-Met signaling pathway in cancerClin Cancer Res200612123657366016778093
  • JiangWGHepatocyte growth factor and the hepatocyte growth factor receptor signalling complex as targets in cancer therapiesCurr Oncol2007142666917576468
  • CaoBSuYOskarssonMNeutralizing monoclonal antibodies to hepatocyte growth factor/scatter factor (HGF/SF) display antitumor activity in animal modelsProc Natl Acad Sci U S A200198137443744811416216
  • DaiYSiemannDWBMS-777607, a small-molecule met kinase inhibitor, suppresses hepatocyte growth factor-stimulated prostate cancer metastatic phenotype in vitroMol Cancer Ther2010961554156120515943
  • HussainMSmithMRSweeneyCCabozantinib (XL184) in metastatic castration-resistant prostate cancer (mCRPC): results from a phase II randomized discontinuation trial [abstract]J Clin Oncol201129 Suppl451622025164
  • Di LorenzoGBuonerbaCKantoffPWImmunotherapy for the treatment of prostate cancerNat Rev Clin Oncol20118955156121606971
  • BanderNHYaoDLiuHMHC class I and II expression in prostate carcinoma and modulation by interferon-alpha and -gammaProstate19973342332399397194
  • YangLPangYMosesHLTGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progressionTrends Immunol201031622022720538542
  • MercaderMBodnerBKMoserMTT cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancerProc Natl Acad Sci U S A20019825145651457011734652
  • MD Anderson Cancer Center, Bristol-Myers SquibbIpilimumab + androgen deprivation therapy in prostate cancerClinicalTrials gov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated April 11, 2012]. Available from: http://clinicaltrials.gov/show/NCT01377389. NLM Identifier: NCT01377389Accessed December 21, 2011
  • OHSU Knight Cancer InstituteNational Cancer Institute Ipilimumab in combination with androgen suppression therapy in treating patients with metastatic hormone-resistant prostate cancerClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2011 [updated March 22, 2012]. Available from: http://clinicaltrials.gov/show/NCT01498978. NLM Identifier: NCT01498978Accessed December 21, 2011