Abstract
Prostate cancer (PCa) is one of the most prevalent cancers and the second leading cause of cancer death among U.S. males. When diagnosed in an early disease stage, primary tumors of PCa may be treated with surgical resection or radiation, sometimes combined with androgen deprivation therapy, with favorable outcomes. Unfortunately, the treatment efficacy of each approach decreases significantly in later stages of PCa that involve metastasis to soft tissues and bone. Metastatic PCa is a heterogeneous disease containing host cells, mature cancer cells, and subpopulation of cancer stem cells (CSC). CSCs are highly tumorigenic due to their self-renewing and differentiating potential, clinically resulting in recurrence and resistance to standard therapies. Therefore, there is a large unmet clinical need to develop therapies, which target CSC activity. In this review, we summarize the main signaling pathways that are implicated in the current preclinical and clinical studies of recurrent metastatic PCa within the bone microenvironment targeting CSCs and discuss the trajectory of therapeutics moving forward.
Keywords: Prostate cancer, Metastasis, Cancer stem cells, Bone microenvironment, Signaling pathway, Therapy
Introduction
Prostate cancer (PCa) is one of the most commonly diagnosed cancers in American males (Denmeade and Isaacs, 2002). With the advent of prostate specific antigen (PSA) testing and increased screening, a significant increase in the diagnosis of early stage PCa has occurred (Grönberg, 2003). In organ confined, low grade PCa, surgical resection or radiation therapy have been highly successful in treating the disease with long term cancer free survival (Bedard and Chow, 2013). However, a larger or higher grade primary tumor increases the chance of recurrence, which is due to the radio-resistant cancer stem cell (CSC) subpopulations located within the tumor (Li et al., 2013). The radio-resistance is often associated with increased CSC self-renewal activity, increased DNA repair capacity, and enhanced reactive oxygen species (ROS) defenses (Ogawa et al., 2013).
There are several mechanisms whereby castrate resistant cells may develop including the reduction of androgen receptor (AR) expression, which occurs in the vast majority of the tumors (Tsujimura et al., 2002). These cells are capable of both self-renewal and possess the ability to differentiate to AR+ PCa cells (Collins et al., 2005; Moltzahn and Thalmann, 2013). In fact, androgen targeted therapies appear to enhance the PCa stem cells or progenitor cell populations, which ultimately contribute to castration resistance (Germann et al., 2012). While the mechanisms for development of mCRPC remain unclear, several intrinsic factors and signaling pathways within the tumors facilitate CSC expansion with an epithelial-mesenchymal transition (EMT) phenotype (Ojo et al., 2015). As a result, the link between mCRPC and CSC activity is critical to understand and target.
One of the major pathways utilized by resistant CSC populations is the engagement or activation of a non‐proliferative state (quiescence, dormancy, or G0 phase), which is highly conserved without multiple inputs and disrupt quiescence (Takeishi and Nakayama, 2016). As such, CSCs are able to bypass therapies that use the traditional anti-proliferative approaches, suggesting that new approaches are needed to identify and ultimately target CSCs directly (Jaworska et al., 2015).
The identification of the source and phenotype of CSCs in PCa has been investigated but remains in controversial. PCa CSCs can be distinguished from non-CSCs based upon the expression of CD44, CD133, and α2β1 integrin (Collins et al., 2005; Sharpe et al., 2013). Qin et al. proposed an integrative definition of cells, which highly expressed CD44, ALDH, and α2β1, and expressed little or no PSA (Qin et al., 2012). Another group proposed that mitotic quiescence rather than surface phenotype was more effective at defining CSCs (Patrawala et al., 2007; Wang et al., 2015a). Intracellularly, the expression of stemness-related transcriptional factors has also been considered to identify and potentially serve as therapeutic targets for CSCs. These include high expression of NANOG, MYC, OCT4, SOX2, and KLF4, which have shown to be expressed by CSCs in significant levels compared to non-CSCs of PCa cells (Jeter et al., 2011).
Many patients with metastatic PCa present with bone lesions (Logothetis and Lin, 2005). The source of pain and reduced quality of life are often associated with skeletal related event (SRE), including pathological fractures, spinal cord compression, and increasing pain (Coleman, 2016). Although SREs are partially managed using anti-osteoclast agents (Zoledronate and Denosumab), it is important to target the source of the bone dysfunctions by preventing or treating PCa (Maier et al., 2016; Zhang et al., 2017c). Signaling in the bone microenvironment is known to play an important role in initiation and maintenance of PCa metastasis and fate of CSCs (Casimiro et al., 2009; Holen, 2016).
Here we explore several pathways, which contribute to a CSC phenotype in the bone marrow: Notch, Wnt, Hedgehog, fibroblast growth factor (FGF), transforming growth factor β (TGFβ), stromal derived factor 1 (SDF-1; CXCL12), and growth arrest protein 6 (GAS6) (Figure 1). Further we summarize current preclinical and clinical studies, which target these pathways in relation to metastatic PCa and/or CSCs that contribute to PCa recurrence (Table 1), and discuss novel routes forward for the future therapeutics.
Figure 1. Core signaling pathways of cancer stem cells are associated with the recurrence of metastatic PCa in the bone microenvironment.

ADAM: a disintegrin and metalloproteinase; TACE: tumor necrosis factor-alpha converting enzyme; NICD: intracellular domain of the notch protein; HAT: acetyltransferase; SKIP: skeletal muscle and kidney enriched inositol phosphatase; CSL: CBF-1, suppressor of hairless, lag-2; MAML: mastermind-like; LRP: lipoprotein receptor-related protein; FZD: Frizzled; DSH: dishevelled; GSK3: glycogen synthase kinase 3; APC: adenomatous polyposis coli; TCF/LEF: T cell factor/lymphocyte enhancer binding factor; PTCH1: patched 1; SMO: smoothened; SUFU: suppressor of fused homolog; Gli1/2: glioma-associated oncogene 1/2; FGF: fibroblast growth factor; FGFR: fibroblast growth factor receptor; FOXO: forkhead box; TKs: tyrosine kinases; PLCγ: phospholipase C gamma; PKC: protein kinase C; MAPKK: mitogen-activated protein kinase kinase; MAPK: mitogen-activated protein kinase; PI3K: phosphoinositide 3-kinase; TGFβ: transforming growth factor beta; GTP: guanosine triphosphate; NFKB: nuclear factor kappa-light-chain-enhancer of activated B cells; GAS6: growth arrest specific 6; mTOR: mechanistic target of rapamycin.
Table 1.
The pre-clinical and clinical studies targeting PCa cells associated with signaling pathways in the bone microenvironment.
| Pathway | Drug/agent | Target PCa | Drug development stage | Description | Citation |
|---|---|---|---|---|---|
| Notch | PF-03084014 | mCRPC, CSC | Pre-clinical | γ-secretase inhibitor | Cui et al., 2015 |
| GSI | mCRPC, CSC | Pre-clinical | γ-secretase inhibitor | Stoyanova et al., 2016 | |
| DBZ | Metastatic | Pre-clinical | Notch inhibitor | Qui et al., 2018 | |
| RO4929097 | mCRPC | Clinical (phase II) NCT01200810 | γ-secretase/Notch inhibitor | NCI, 2010–2017 | |
| Wnt | LGK974 | Metastatic | Pre-clinical | Wnt inhibitor blocks porcupine-a membrane-bound O-acyltransferase | Ma et al., 2016 |
| ICG00I | Metastatic, CSC | Pre-clinical | β-catenin inhibitor | Zhang et al., 2018 | |
| Ipafricept (OMP-54F28) | Advanced, CSC | Clinical (Phase I) NCT01608867 | frizzled 8 receptor fused IgG1 Fc | Jimeno el al., 2017 | |
| Foxy-5 | Metastatic | Clinical (Phase I) NCT02020291 NCT02655952 | Formylated 6 amino acid peptide targets Wnt-5a | WntResearch AB, 2013–2016; WntResearch AB, 2016–2017 | |
| Hedghog | GANT61 | Metastatic | Pre-clinical | Gli inhibitor | Lauth et al., 2007; Rimkus et al, 2016 |
| Sonidegib (erismodegib, LDE225) | Metastatic, CSC | Pre-clinical | Hh inhibitor | Nanta et al., 2013 | |
| Sonidegib | Advanced | Clinical (Phase I) NCT021111870 | Hh inhibitor | Ross et al., 2017 | |
| Vismodegib | mCRPC | Clinical NCT02115828 | Hh inhibitor | Maughan et al., 2016 | |
| Itraconozale | Advanced | Clinical (Phase II) NCT018787331 | Antifungal agent | Lee et al., 2019 | |
| FGF | AZ8010 | Metastatic | Pre-clinical | FGFR inhibitor | Feng et al., 2012 |
| PD173074/CH5183284 | Metastatic | Pre-clinical | FGFR antagonist targeting FGFRI | Bluemn et al., 2017 | |
| Dovitinib (TK12S8) | Metastatic and osteoblasts | Pre-clinical | FGFR inhibitor | Wan et al., 2014;Yadav et al., 2017 | |
| Dovitinib | mCRPC | Clinical (Phase II) NCT01741116 | FGFR inhibitor | Choi et al., 2018 | |
| Nintedanib (BIBF1120) | Metastatic | Pre-clinical | FGFR inhibitor | da Silva et al., 2017, 2018 | |
| Nintedanib | mCRPC | Clinical (Phase I, II) NCT02856425 NCT00706628 | FGFR inhibitor | Bousquet el al., 2011; Droz el al., 2014; Gustave Roussy, Paris 2016–2018; Boehringer Ingelheim, 2008–2016 | |
| TGFβ | Tranilast | Metastatic | Pre-clinical | Anti-allergic agent targets TGFβl | Izumi et al., 2009 |
| IN-1130 | Metastatic | Pre-clinical | TGFβRI ALK-5 kinase inhibitor | Lee et al., 2008 | |
| SB-431542 | Metastatic | Pre-clinical | TGFβRI ALK-5 kinase inhibitor | Miles et al., 2012 | |
| LY2109761 | Metastatic | Pre-clinical | TGFβRI & RII kinase inhibitor | Wan et al., 2012 | |
| Galunisertib (LY2157299) | Metastatic | Pre-clinical | TGFβRI kinase inhibitor | Paller et al., 2018 | |
| Galunisertib | mCRPC | Clinical (Phase II) NCT02452008 | TGFβRI kinase inhibitor | Johns Hopkins, 2016–2021 | |
| CXCL12 | CTCE-0908 (CTE9908) or Plerixafor (AMD3100) | Metastatic | Pre-clinical | CXCR4 antagonist | Gravina et al., 2015 |
| Plerixafor | Metastatic. CSC | Pre-clinical | CXCR4 antagonist; DTC mobiliser by blocking the CXCR4 | Shiozawa et al., 2011; Dubrovska et al., 2012; Gravina et al., 2015; Wang et al., 2015; Conley-LaComb et al., 2016; Jung et al., 2018 | |
| Plerixafor | Metastatic | Clinical NCT02478125 | CXCR4 antagonist; DTC mobiliser by blocking the CXCR4 | Johns Hopkins. 2015–2017 | |
| GAS6 | Bemcentinib (R428, BOB-324) | Metastatic | Pre-clinical | Axl inhibitor blocks autophosphorylation of Axl | Bansal el al., 2015 |
| Amuvatinib (MP470) | Metastatic | Pre-clinical | Axl inhibitor blocks ATP-binding cassette Bl (ABCB1) | Lin et al., 2017 | |
| UNC1062 | Metastatic, CSC | Pre-clinical | Mer inhibitor blocks autophosphorylation of Mer | Shiozawa et al., 2016; Jung et al., 2016 | |
| Cabozantinib (XL-184, Exelixis) | mCRPC | Pre-clinical | Tyrosine kinase inhibitor (c-Met, VEGFRs, Alk, Ron, and Axl) | Graham et al., 2014; Dai et al., 2014; Stern and Alvares, 2014; Haider et al., 2015; Leibowitz-Amit et al., 2016 | |
| Cabozantinib | mCRPC | Clinical (Phase II, III) NCT01605227 NCT01522443 | Tyrosine kinase inhibitor (c-Met, VEGFRs, Alk, Ron, and Axl) | Exelixis, 2012–2018; Smith et al., 2013; Smith et al., 2014; Smith et al., 2016 | |
| Foretinib (GSK1363089, XL880) | Advanced | Clinical (Phase II) NCT00726323 | Tyrosine kimse inhibitor (c-Met, VEGFRs, Alk, Ron, and Axl) | GlaxoSmithKline, 2008–2017; Choueiri et al., 2013 | |
Bone Marrow Signaling Pathways Targeting Cancer Stem Cells
The bone marrow is comprised of diverse marrow cells including all of the hematopoietic lineage cells, hematopoietic stem cells (HSCs), mesenchymal stem cell (MSCs), bone marrow stromal cells (BMSCs), osteoblasts, endothelial cells, osteoclasts, and immune cells and extracellular matrix (ECM), which releases many different growth factors, cytokines, interleukins, adhesion molecules, as well as ECM proteins. BMSCs express many growth factors and adhesion molecules such as Hedgehog (Hh), FGFs, and Jagged 1. Endothelial cells produce vascular endothelial growth factor (VEGF) and CXCL12. Osteoblasts also produce a multitude of growth factors and cytokines such as Wnt, FGFs, TGFβ, CXCL12, GAS6, bone morphogenic proteins (BMP), osteopontin (OPN), interleukins, and receptor activator of nuclear factor kappa-B ligand (RANKL). Osteoblasts also express adhesion molecules (e.g., Jagged1, Annexin2) and integrins, which bind to different ECM proteins, such as collagen, laminin, vitronectin, and fibronectin. Many of factors from osteoblasts or calcified bone matrix activate osteoclast formation, which release the growth factors and proteolytic enzymes (Decker et al., 2017; Pedersen et al., 2012) (Figure 2).
Figure 2. Bone marrow signaling interacts with metastatic PCa CSCs.

BMSC: bone marrow stromal cell; HSC: hematopoietic stem cell; MSC: mesenchymal stem cell; ECM: extracellular matrix; VEGF: vascular endothelial growth factor; CXCL12: stromal derived factor 1 (SDF-1); GAS6: growth arrest protein 6; TGFβ: transforming growth factor beta; FGFs: fibroblast growth factors; BMPs: bone morphogenic proteins; IL: Interleukin, OPN: osteopontin; RANKL: receptor activator of nuclear factor kappa-B ligand.
Uniquely, PCa cells can convert to a CSC phenotype defined by dual expression of CD133 and CD44 (CD133+/CD44+) under the influence of the marrow (Shiozawa et al., 2016). These observations suggest that the metastatic bone marrow niche for tumor cells may contribute or even direct the establishment of PCa CSCs (Shiozawa et al., 2016). The fate of CSCs relies on marrow microenvironment and the signals from marrow control CSC activities including the protection against the environmental stress. Thus, development agents that target these signaling pathways will be the critical steps to eradicate CSCs in metastatic diseases.
Notch Pathway
Notch is a transmembrane receptor, which plays a critical role in embryogenesis, stem cell self-renewal, and differentiation in many tissues. The canonical Notch signaling pathway is activated through binding of Notch ligands (Jagged1/2 and Delta-like 1/3/4) to Notch receptors (1/2/3/4). The canonical Notch signaling pathway plays a critical role in stem cell activity (Gu et al., 2012). The non-canonical Notch signaling is thought to act in a ligand- or transcription-independent manner (Andersen et al., 2012).
In bone, BMSCs and osteoblasts express Jagged1, the ligand for Notch, and enhanced Jagged1 expression promotes Notch-expressing PCa cell bone metastasis (Delury et al., 2016). Several studies indicate that the Notch signaling plays a significant role in the development of an EMT phenotype and associated with CSC activities. Further, high levels of Notch1 expression are associated with an EMT phenotype frequently found in metastatic bone lesions compared with primary PCa tissues (Sethi et al., 2010). Down-regulation of Notch1 by microRNA-200 is partially responsible for the inhibition of sphere-forming ability and PCa cells (Kong et al., 2010). Importantly, when the ligand binds to its receptor, a disintegrin and metalloproteinases (ADAMs)/tumor necrosis factor-alpha converting enzyme (TACE) and gamma secretase cleave the Notch receptor, releasing the Notch intracellular domain (NICD) from the plasma membrane and facilitating its translocation into the nucleus (Takebe et al., 2015). A recent study also demonstrates that expression of NICD1 is highly elevated in high-risk PCa and castration-resistant PCa (Stoyanova et al., 2016). Moreover, the tumors driven by the combination of NICD1 with multiple (Ras/Raf/MAPK, myrAKT, or Myc) pathways have an increased capacity for self-renewal with enhanced metastatic potential (Stoyanova et al., 2016). Overexpression of Notch1 and NICD in PCa cells enhanced migration and invasion, and increased sphere formation of PCa cells (Zhang et al., 2017b). Expression of Notch2 also induces the stem-like properties and enhances docetaxel-resistance in PCa cells (Qiu et al., 2018).
In pre-clinical studies, a combination of a γ-secretase inhibitor (PF-03084014) and docetaxel reduced both docetaxel-sensitive and docetaxel-resistant CRPC tumor growth in soft tissues and bone better than either agent alone. Targeting this pathway with a γ-secretase inhibitor was associated with reduced survival, increased apoptosis, reduced microvessel density, reduced EMT, and reduced cancer stem-like cells (Cui et al., 2015). Further work on γ-secretase inhibitor (GSI) in vitro and murine model demonstrated inhibition of Notch1 signaling and γ-secretase, resulting in the delay of PCa cell growth and inhibition of colony formation (Stoyanova et al., 2016). Use of a γ-secretase inhibitor, Dibenzazepine (DBZ) with Docetaxel reduced tumor burden and prolonged survival following injection of PCa cells or patient-derived primary prostate carcinoma-1 (PPC1) cells in s.c. tumor xenograft models (Qiu et al., 2018). RO4929097, an inhibitor of γ-secretase in Notch signaling pathway, was tested in phase II clinical trial (clinicaltrials.gov: NCT01200810). This study evaluated efficacy of anti-androgen Bicalutamide with RO4929097 in patients with PCa recurrence after surgery or radiation; however, it was terminated early due to lack of sufficient drug supply.
Wnt Pathway
Wnt plays an important role in embryonic development and adult tissue homeostasis. Wnt signaling is mediated through at least three major pathways; canonical Wnt pathway (regulation of gene transcription), non-canonical planar cell polarity pathway (regulation of cytoskeleton), and non-canonical Wnt/calcium pathway (regulation of calcium). The canonical Wnt signaling pathway is known to play a critical role in self-renewal and maintenance of stem cells (Han et al., 2013).
In bone, Wnts promote the differentiation of pre-osteoblasts into mature osteoblasts through the β-catenin dependent canonical pathway (Kobayashi et al., 2008) and suppress bone resorption by altering RANKL/osteoprotegerin (OPG) ratios through the β-catenin dependent canonical pathway (Hall et al., 2006). Wnt signaling has been linked to PCa bone metastasis with induction of osteoblastic activity (Yardy and Brewster, 2005). In fact, β-catenin expression decreases during PCa progression and nuclear β-catenin decreases in PCa bone metastasis, which results in masking of osteogenic Wnts, thereby, switching the phenotype to osteolysis. However, during metastases progress, the expression of the Wnt antagonist Dickkopf-1 (DKK1) is decreased, which unmasks Wnt’s osteoblastic activities. Therefore, DKK1 is believed to serve as an important player switching from osteolytic to osteoblastic activities at the metastatic site during PCa bone metastasis (Hall et al., 2008; Hall et al., 2006). Critically, the Wnt/β-catenin signaling pathway is known to be highly activated in CSCs, and may play an important role in prostate stem cell self-renewal (Bisson and Prowse, 2009). Specifically, activation of Wnt3 signaling stimulates a significant induction of nuclear β-catenin, keratin 18, CD133, and CD44 expression in PCa cells, which regulates the sphere size and enhanced self-renewal capacity (Bisson and Prowse, 2009). Interestingly, a series of studies have shown that manipulation of the Wnt pathway is capable of regulating the CSC phenotype. For example, microRNA-320 suppresses the stem cell-like phenotype of PCa cells by downregulating Wnt/β-catenin signaling pathway (Hsieh et al., 2012). Additionally, AR79, an inhibitor of GSK-3β (a negative regulator of Wnt signaling), increased ALDH+/CD133+ stem-like population from tumor cells, which effectively promoted tumor growth in xenograft models (Jiang et al., 2013). Furthermore, high levels of PHD finger protein 21B (PHF21B) expression are associated with poor recurrence-free survival in PCa patients and PHF21B overexpression activated Wnt/β-catenin signaling pathway, promoting stem-like properties in PCa cells (Li et al., 2017). Recently, Wnt/β-catenin has been shown to enhance human telomerase reverse transcriptase (hTERT) activity in PCa cells and thereby regulate the self-renewal and differentiation activity of PCa cells (Zhang et al., 2017a). A novel Wnt co-activator, increases the canonical Wnt activity, which maintains the prostate CSC subpopulations in PCa (Pai et al., 2019). Together, all of these data support that Wnt signaling may be involved with PCa tumorigenesis with an induction of stem cell phenotype.
In preclinical models, the Wnt inhibitors 1) porcupine- a membrane-bound O-acyltransferase (LGK974) (Ma et al., 2016) and 2) β-catenin inhibitor (ICG001) (Zhang et al., 2018) have been shown to decrease CSC function and tumor growth. In fact, the inhibition of Wnt/β-catenin sensitizes PCa cells to Enzalutamide (Zhang et al., 2018). At a clinical level, Ipafricept (OMP-54F28), a recombinant fusion protein of frizzled 8 receptor fused to a human IgG1 Fc fragment that binds Wnt ligands, was evaluated in a phase I trial for treatment of solid tumors including PCa (clinicaltrials.gov: NCT01608867). Ipafricept was well tolerated by patients with solid tumors (Jimeno et al., 2017). The Foxy-5, a formylated 6 amino acid peptide fragment, is effective as a Wnt-5a inhibitor and as such was developed as an anti-metastatic agent. It was tested in two phase I clinical trials (clinicaltrials.gov: NCT02020291 and NCT02655952) to establish the recommended dose for a clinical phase II study, but in trial the results were inconclusive and no recommended phase II trial initiated.
Hedgehog Pathway
Hh is a conserved regulator for stem cell activities during animal development. Hh signaling is mediated through the transmembrane proteins Patched1 (PTCH1)-associated Smoothened (SMO), which activates glioma-associated oncogene (Gli) transcription factor leading to the activities related to proliferation and pluripotency (Hadjimichael et al., 2015). As such the Hh signaling pathway is essential for the maintenance of CSCs in human cancers (Chang et al., 2011; Han et al., 2013).
In bone, Hh is produced by bone marrow-derived mesenchymal stromal cells which supports cell survival in hematological disorders (Zou et al., 2015). Further, Hh signaling-activated Gli2 transcription induces BMP2 expression, which enhances osteoblast differentiation (Kesper et al., 2010; Zhao et al., 2006). Gli1 upregulates osteopontin (OPN), a factor linked to malignant behavior of cancer cells (Das et al., 2009). In advanced PCa lesions, overexpression of PTCH1 and SMO with P63+ basal/stem cells differentiate into AR+ and AR− progeny of metastatic tumor cells, which significantly increased the metastatic potential (Chang et al., 2011). Hh signaling is upregulated in prostate CSCs (Hurt et al., 2008; Klarmann et al., 2009).
In preclinical studies, the cell-permeable hexahydropyrimidine Gli inhibitor GANT61 strongly reduced PTCH1 expression, which decreased PCa growth and proliferation (Lauth et al., 2007; Rimkus et al., 2016). Sonidegib (Erismodegib, LDE225), an Hh inhibitor, that antagonizes SMO, downregulates pluripotency-maintaining factors NANOG, OCT4, SOX2, c-MYC and thereby inhibits CSC activity and ultimately tumor growth in vitro and a xenograft mouse model (Nanta et al., 2013). Critically, was tested in phase I trial (clinicaltrials.gov: NCT021111870) for men with localized high-risk PCa undergoing radical prostatectomy. Hh activity was detectable at baseline in men with localized high-risk PCa (as measured by Gli1 expression). Sonidegib penetrated into prostatic tissue and induced a >60-fold suppression of the Hh signaling (Ross et al., 2017). Vismodegib, an Hh inhibitor, has been evaluated in men with mCRPC (clinicaltrials.gov: NCT02115828). Vismodegib suppressed Hh signaling as evaluated by Gli1 mRNA expression in mCRPC tissues from the majority of patients. Despite this pharmaco-dynamic response, unfortunately this was little evidence of clinical activity (Maughan et al., 2016). Itraconozale is an antifungal agent tested in patients with biochemically relapsed PCa in a phase II trial (clinicaltrials.gov: NCT018787331). Results indicate that Itraconazole modulate serum PSA levels without affecting circulating serum testosterone levels (Lee et al., 2019).
FGF Pathway
FGF is involved in the regulation of many developmental processes including morphogenesis, differentiation, cell proliferation, and migration. FGFs are comprised of a gene family consisting of 22 structurally related proteins that are divided into 7 subfamilies based on sequence homology (Corn et al., 2013). FGF signaling is activated by ligand binding to four cognate high-affinity tyrosine kinases receptors (FGFR1-4). These interactions result in activation of multiple signal transduction pathways including the MAPK (Erk), PLCγ-PKC, and PI3K-AKT pathways (Kwabi-Addo et al., 2004).
In bone, multiple FGFs are produced by BMSCs and immune cells (Ishino et al., 2013; Itkin et al., 2013). FGF signaling in the bone microenvironment promotes castrate-resistant PCa growth (Lescarbeau et al., 2012; Li et al., 2008) and bone biopsies of mCRPC patients demonstrate greater frequency of FGF8 expression than patients with the primary tumors (West et al., 2001). FGF2, FGF8, and FGF9 are produced by PCa cells during bone metastases and are known to participate in osteogenesis and bone remodeling (Koeneman et al., 1999; Valta et al., 2008). A recent study has been reported a phenotypic shift occurs in metastatic PCa with the AR-null and neuroendocrine (NE)-null phenotype through elevated FGF and MAPK signaling pathways (Bluemn et al., 2017). At present more investigation is needed to elucidate the role of FGF signaling in CSC development in bone metastatic PCa.
Nevertheless, pre-clinical and clinical studies of FGF signaling pathway have been demonstrated in PCa. AZ8010, a selective inhibitor of FGFR1 and 4, inhibited invasion and tumor growth in vitro and in vivo tumor xenograft models (Feng et al., 2012). Combination of inhibitors of FGFR1 (PD173074 or CH-5183284) and/or MAPK (U0126) represses the growth of double-negative (AR-null and NE-null) metastatic PCa in vitro and in vivo tumor xenograft models (Bluemn et al., 2017). Dovitinib (TKI258), the FGFR antagonist targeting FGFR1-3, inhibits FGFR signaling in osteoblasts rather than in cancer cells, thereby improving bone quality and blocking cancer-bone interaction (Wan et al., 2014). Although Dovitinib shows efficacy in mouse-model of PCa bone metastasis, this Dovitinib induces NE phenotype from AR+ and AR-PCa cells, perhaps as a way of developing resistance (Yadav et al., 2017). A phase II study of Dovitinib in patients with mCRPC (clinicaltrials.gov: NCT01741116) demonstrated a modest antitumor effect (Choi et al., 2018). Dovitinib has manageable toxicities in men with mCRPC (Choi et al., 2018). A recent report shows that Nintedanib (BIBF1120), the tyrosine-kinase inhibitor of VEGF, FGF1-3, and PDGF signaling pathways, inhibits PCa growth by modulating both cell cycle and angiogenesis. Moreover, Nintedanib delays tumor progression in transgenic adenocarcinoma of the mouse prostate (TRAMP) (da Silva et al., 2017) as well as migration and invasion of PCa in nude mice (da Silva et al., 2018). Further, two studies show the efficacy and safety of two doses of Nintedanib in phase I and phase II trials. First study demonstrated in a phase I trial that Nintedanib (200mg twice daily) combined with Docetaxel (75mg/m2, every 3 weeks) was well tolerated to the patients, with promising results in chemo-naive hormone-refractory PCa (Bousquet et al., 2011). Second study demonstrated in a phase II trial that Nintedanib (250mg) showed only modest activity with manageable adverse events in patients with mCRPC with post-Docetaxel treatment (Droz et al., 2014). More recently, Nintedanib was tested in phase I and II clinical trials for the patients with hormone-resistant PCa (clinicaltrials.gov: NCT02856425, NCT00706628), although results have not been posted.
TGF Pathway
TGFβ is a secreted protein that performs many cellular functions including proliferation, differentiation, and migration (Massague, 2012). TGFβ has three isoforms, which bind to several different homo- or heterodimeric TGFβ receptors (Doré et al., 1998). TGFβ signaling has dual functions in terms of PCa; in the SMAD-dependent pathway TGFβ serves as a tumor suppressor in early stage PCa development and later TGFβ switches to non-SMAD pathways and serve as a tumor promoter in advanced PCa (Lebrun, 2012).
In bone, TGFβ is primarily produced by active osteoblasts and bone stromal cells and is a potent paracrine stimulator of PCa growth (Festuccia et al., 2000; Horner et al., 1998; Meng et al., 2018). In clinical studies, TGFβ plasma levels are markedly elevated in men with metastatic PCa to regional lymph nodes and bone and high preoperative TGFβ plasma levels correlate with disease progression after prostatectomy (Shariat et al., 2001). TGFβ signaling may play a significant role in establishing a CSC phenotype. Activation of TGFβ signaling induces significant enrichment of CD44+/CD133+/CD24− populations by downregulating poly r(C) binding protein (PCBP)-1 (Chen et al., 2015). Further, downregulation of TGFβ2 is associated with escape from dormancy in PCa xenograft models (Ruppender et al., 2015). Another member of the TGF superfamily BMP7 has been shown to play a critical role in dormancy and recurrence of prostate CSCs in bone via BMPR2 mediated p38 MAPK signaling (Kobayashi et al., 2011).
Pre-clinical studies have demonstrated that TGFβ signaling plays a significant role in PCa bone metastases. For example, Tranilast (N-3, 4-dimethoxycinnamoyl-anthranilic acid) is a therapeutic agent used in the treatment of allergic diseases. Tranilast suppresses TGFβ1 secretion from bone-derived stromal cells thereby inhibiting hormone refractory PCa cell growth (Izumi et al., 2009). Alternatively, IN-1130, a novel TGFβRI kinase (activin receptor-like kinase 5, ALK-5) inhibitor, decreases the PCa growth in mice in part by activating an host immune response (Lee et al., 2008), and SB-431542, another TGFRI ALK-5 inhibitor, also suppresses growth and motility of PCa cells (Miles et al., 2012). Inhibition of TGFβ signaling by a selective TGFβRI kinase inhibitor, LY2109761 was shown to control PCa growth in bone, while increasing the mass of normal bone (Wan et al., 2012). More recently, the combination of the TGFβR1 inhibitor Galunisertib (LY2157299) with Enzalutamide, significantly suppressed PCa growth by increasing apoptosis (Paller et al., 2018). At the clinical level, currently combination of Galunisertib and Enzalutamide in a phase II clinical trial (clinicaltrials.gov: NCT02452008) is ongoing for targeting mCRPC patients.
CXCL12 Pathway
CXCL12 is a chemokine, which is expressed in both embryonic and adult tissues including brain, heart, lung, liver, kidney, spleen, and bone marrow. CXCL12 plays a key role in tissue homeostasis, survival, trafficking of hematopoietic and immune cells, and cancer progression and metastases (Lataillade et al., 2004). CXCL12 binds primarily to CXC receptor 4 (CXCR4; CD184). CXCR4 is expressed in a variety of cell types including lymphocytes, hematopoietic stem cells, endothelial cells, epithelial cells, and cancer cells (Rossi and Zlotnik, 2000).
In bone, CXCL12 is highly expressed by pre-osteoblasts and endothelial cells and is down-regulated upon maturation and matrix synthesis (Jung et al., 2015). Importantly, this signaling pathway plays critical roles in the CSC activities that are responsible for the establishment and development of PCa bone metastases (Cojoc et al., 2013). Activation of CXCR4 by CXCL12 in the CD44+/CD133+ prostate CSCs induce self-renewal and tumorigenesis of PCa cells through activation of PI3K/AKT signaling and downregulation of the FOXO transcriptional factor, which ultimately impacts therapy resistance (Cojoc et al., 2013; Dubrovska et al., 2010; Dubrovska et al., 2009; Trautmann et al., 2014). High levels of CXCR4, CD44, and integrin α2/β1 expressing quiescent cells are significantly more tumorigenic than fast growing cells, indicating that CXCR4 is a CXC12 signaling mediator, which facilitates the regulation of CSC fate in bone metastases (Wang et al., 2015b). Interestingly, an intracellular splice variant of CXCL12, CXCL12γ induces the expression of a CSC (CD44+/CD133+) and neuroendocrine (high levels of CD44/CD133 expression) phenotypes of PCa cells through CXCR4-mediated PKCα and NFκB signaling. This activity results in the development of aggressive metastatic resistance of PCa (Jung et al., 2018).
Several pre-clinical studies of CXCR4 inhibitors have been shown to be significantly more effective at eradicating metastatic tumors when used in combination with conventional chemo-or radiation therapies that target PCa and/or PCa CSCs. For example, CXCR4 antagonist, CTCE-9908 (CTE9908) or Plerixafor (AMD3100) blocks CXCL12 binding to CXCR4 resulted in a significant improvement in the overall survival of mice and reduction of intra-osseous PCa tumor growth. In conjunction, decreased osteolysis, serum mouse tartrate-resistant acid phosphatase (mTRAP), and type I collagen fragments were observed (Gravina et al., 2015). Furthermore, AMD3100 alters the homing of CXCR4 expressing-quiescent tumor cells to the bone and reduces binding of tumor cells to the bone surface, although no significant reductions in the total number of tumor cells were detected in marrow (Wang et al., 2015b). AMD3100 also decreases the initial establishment of bone tumors without affecting the expansion of existing bone tumors in tumor xenograft models in a metastatic setting (Conley-LaComb et al., 2016). Not surprisingly, given the role in homing, AMD3100 is able to mobilize disseminated tumor cells (DTCs) out of the bone marrow niche in tumor xenograft models (Shiozawa et al., 2011). Critically, AMD3100 is able to target the induction of prostate stem-like population in PCa cells in vitro and in vivo tumor xenograft models (Dubrovska et al., 2012). AMD3100 inhibits the sphere formation in PCa cells and sensitizes PCa cells to Docetaxel chemotherapy (Jung et al., 2018). However, AMD3100 had minimal impact altering CXCL12γ-mediated prostatosphere formation, given the primary function of CXCL12γ is to signal intracellularly (Jung et al., 2018). Co-treatment of AMD3100 with granulocyte colony-stimulating factor (G-CSF) increases the mobilization of hematopoietic stem cells (HSCs), which is important as a source of HSCs for transplantation. AMD3100 is currently being used for the patients with lymphoma and multiple myeloma (Cashen et al., 2008; DiPersio et al., 2009). Recently, a clinical trial was conducted to evaluate whether CXCR4 inhibition in PCa patients by AMD3100 was able to mobilize DTCs out of the bone marrow (clinicaltrials.gov: NCT02478125), although results have not been posted.
GAS6 Pathway
GAS6 is a vitamin K dependent grow factor, which is expressed in many human tissues and regulates proliferation, survival, and migration (Goruppi et al., 1999). GAS6 binds to its receptors, TRYO3, AXL, and MER (TAM), which are known to play important roles in regulating DTC dormancy and CSCs in a number of settings (Taichman et al., 2013).
In bone, GAS6 is expressed by osteoblasts in marrow and is known to regulate bone metastasis and induction of DTC dormancy (Cackowski et al., 2017; Shiozawa et al., 2010; Taichman et al., 2013). The studies from our group have recently linked GAS6 to a CSC phenotype through MER receptor signaling (Jung et al., 2016; Shiozawa et al., 2016). In the first of these studies, we showed that DTCs recovered from murine marrow were significantly enriched for a CSC phenotype. Critically, the conversion of DTCs to CSCs (CD133+/CD44+) was regulated by niche-derived GAS6 through the MER and mTOR signaling pathways in vitro and in vivo tumor xenograft model. GAS6 triggered mTOR signaling in PCa followed by increases of both mTORC1 and mTORC2, and these were diminished by the mTORC1 inhibitor Rapamycin and the dual mTORC1/2 inhibitor pp242. Inhibition of MER signaling by a MER inhibitor decreased mTORC2 signaling activated by GAS6. Thus, the study demonstrated that the marrow niche plays a significant role in maintaining tumor-initiating PCa and suggests a functional relationship between CSCs and dormancy (Shiozawa et al., 2016). In a second of these studies, we demonstrated that elevated endogenous GAS6 expression can be found preferentially in putative CSCs (CD133+/CD44+) compared to non-CSCs (CD133-/CD44-) isolated from the coculture of PCa cells and osteoblasts in vitro. Similar observations were made in DTCs isolated from the bone marrow at 24 hours post injection in tumor xenograft models. The elevated endogenous GAS6 expression activates the phosphorylation of MER receptor signaling and subsequent induction of a CSC phenotype. The study supports the concept that endogenous GAS6 and MER receptor signaling contributes to the establishment of PCa CSCs in the bone marrow microenvironment (Jung et al., 2016).
Inhibitors of GAS6 and TAM signaling pathways are being tested in PCa tumors in combination with chemotherapeutic drugs in pre-clinical studies (Corno et al., 2016; Varkaris et al., 2011). For example, Bemcentinib known as R428 or BGB-324 is a novel small molecule relatively specific AXL inhibitor that blocks auto-phosphorylation of AXL. The activation of this agent blocks cancer progression, invasion, metastasis, and drug resistance in models of metastatic PCa. R428 sensitizes AXL expressing PCa cells (DU145) to Metformin treatment (Bansal et al., 2015). Thus, a combination of Docetaxel chemotherapy with R428 is significantly more effective to suppress proliferation, migration, invasion, and tumor growth, and resistance in PCa cells (PC3 and DU145) in vitro and in vivo tumor xenograft model. Further, Amuvatinib (MP470), a different AXL inhibitor, significantly blocks expression of the ATP-binding cassette B1 (ABCB1) in the resistant PCa cells (Lin et al., 2017). UNC1062, a MER inhibitor, prevents sphere-forming ability of PCa cells, while it did not affect cell viability (Jung et al., 2016; Shiozawa et al., 2016). Cabozantinib (XL-184, Exelixis) is an orally bioavailable inhibitor of pro-oncogenic tyrosine kinases including c-MET, vascular endothelial growth factor receptors (VEGFRs), ALK, RON, and AXL. Not unexpectedly given the broad range of targets, Cabozantinib has potent effects on both tumor and tumor-induced bone matrix remodeling in the intratibial VCaP xenograft models (Graham et al., 2014). Interestingly Cabozantinib inhibited tumor growth in tumor xenograft models in a metastatic setting and produced changes in the bone microenvironment, including a biphasic effect on an increase of osteoblast activity and inhibition of osteoclast production, resulting in bone remodeling (Dai et al., 2014). Cabozantinib has been shown to decrease RANKL expression in osteoblastic cells and produces inhibition of osteoclastogenesis and parathyroid hormone-related protein (PTHrP)-stimulated bone resorption (Stern and Alvares, 2014). Short-term treatment with Cabozantinib to animals without tumor induced rapid modification of the bone microenvironment including significant elongation of the epiphyseal growth plate and hypertrophic chondrocyte zone with increased osteoblast numbers and reduced osteoclasts as well as altering other marrow elements including megakaryocytes (Haider et al., 2015). In the plasma of Cabozantinib-treated mCRPC patients, significant increase levels of VEGFA, FLT3L, c-MET, AXL, GAS6A, bone-specific alkaline phosphatase, interleukin-8, hypoxia markers CA9 and Clusterin and decrease levels of VEGFR2, TRAP5b, Angiopoietin-2, TIMP-2 and TIE-2 were observed (Leibowitz-Amit et al., 2016).
At the clinical level, in Phase II and III clinical studies (clinicaltrials.gov: NCT01605227, NCT01522443), Cabozantinib showed clinical activity in men with mCRPC, including improvements in bone scans, pain, measurable soft tissue disease, circulating tumor cells, and decreases in biomarkers for bone turnover (Smith et al., 2013; Smith et al., 2016; Smith et al., 2014). Despite these encouraging results and early stage clinical activity, Cabozantinib failed to increase survival of heavily pre-treated mCRPC patients in a phase III trial (Smith et al., 2016). These observations highlight the need to further understand regulation of CSCs and castration resistance in order to best bring effective treatments to patients. For example, based on laboratory studies, Cabozantinib may be best used effectively with other agents. This was recently studied in a phase I trial in combination with second generation anti-androgen Abiraterone (Choudhury et al., 2018). Foretinib (GSK1363089 or XL880) is a multi-targeted tyrosine kinase inhibitor whose targets include c-MET, RON, AXL, VEGFR2, and PDGFR. Foretinib has similar pharmacologic effects to Cabozantinib. Foretinib has been tested in several solid tumors in phase II studies including the few patients with PCa (clinicaltrials.gov: NCT00726323). Foretinib showed modest antitumor activity with manageable toxicity profiles in patients (Choueiri et al., 2013).
Conclusion and Future Direction
A poor prognosis and reduced quality of life has been observed in patients with elevated numbers of PCa stem cells, which have been identified in bone. As metastatic PCa develops resistance to many of the current therapies, development of new therapies with novel approaches are needed. One approach is to target the bone microenvironment by focusing on signaling pathways which also govern PCa metastasis and CSC activities as those discussed in this review. Many studies have already been activated and several groups have progressed to preclinical and clinical stages of therapy development as summarized previously. This approach opens several new therapeutic possibilities in PCa and other tumor types. For example, breast cancer, which shares lineage-similarity with PCa, have been shown to be sensitive to selective pathway inhibitors identified by rapid high-throughput screening methods (Gupta et al., 2009). Furthermore, nano-therapy delivering siRNA (Zhou et al., 2013) or by targeting epigenetic events and/or genetic manipulation of these pathways have also shown early promise in breast cancer therapeutics (Eccles et al., 2013). These are a few of the many possible avenues not currently explored in the PCa field, but which, if applied in the context of targeting CSCs may be of great value in the future.
Acknowledgements
Dr. Younghun Jung gave valuable critique on the signaling pathway of cancer stem cells. This work is directly supported by the National Cancer Institute (R.S. Taichman (CA093900 and CA163124)), the Department of Defense (R.S. Taichman (W81XW-15-1-0413 and W81XWH-14-1-0403)) and the Prostate Cancer Foundation Challenge Award R.S. Taichman (16CHAL05). R.T. receives support as the Major McKinley Ash Collegiate Professor. F.C. Cackowski receives support from Prostate Cancer Foundation Challenge Award (16CHAL05) and additionally from a Career Enhancement Award, Sub-Award (F048931), of NIH/NCI Prostate Cancer Specialized Program in Research Excellence (SPORE) to Arul Chinnaiyan at the University of Michigan (F036250). F.C. Cackowski also receives Prostate Cancer Foundation Young Investigator Award (18YOUN04), and University of Michigan Department of Internal Medicine start-up funds.
Footnotes
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
References
- Andersen P, Uosaki H, Shenje LT, and Kwon C. 2012. Non-canonical Notch signaling: emerging role and mechanism. Trends in cell biology 22:257–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bansal N, Mishra PJ, Stein M, DiPaola RS, and Bertino JR. 2015. Axl receptor tyrosine kinase is up-regulated in metformin resistant prostate cancer cells. Oncotarget 6:15321–15331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bedard G, and Chow E. 2013. The failures and challenges of bone metastases research in radiation oncology. Journal of bone oncology 2:84–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bisson I, and Prowse DM. 2009. WNT signaling regulates self-renewal and differentiation of prostate cancer cells with stem cell characteristics. Cell Res 19:683–697. [DOI] [PubMed] [Google Scholar]
- Bluemn EG, Coleman IM, Lucas JM, Coleman RT, Hernandez-Lopez S, Tharakan R, Bianchi-Frias D, Dumpit RF, Kaipainen A, and Corella AN. 2017. Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling. Cancer cell 32:474–489. e476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bousquet G, Alexandre J, Le Tourneau C, Goldwasser F, Faivre S, de Mont-Serrat H, Kaiser R, Misset JL, and Raymond E. 2011. Phase I study of BIBF 1120 with docetaxel and prednisone in metastatic chemo-naive hormone-refractory prostate cancer patients. Br J Cancer 105:1640–1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cackowski FC, Eber MR, Rhee J, Decker AM, Yumoto K, Berry JE, Lee E, Shiozawa Y, Jung Y, and Aguirre‐Ghiso JA. 2017. Mer tyrosine kinase regulates disseminated prostate cancer cellular dormancy. Journal of cellular biochemistry 118:891–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cashen A, Lopez S, Gao F, Calandra G, MacFarland R, Badel K, and DiPersio J. 2008. A phase II study of plerixafor (AMD3100) plus G-CSF for autologous hematopoietic progenitor cell mobilization in patients with Hodgkin lymphoma. Biology of Blood and Marrow Transplantation 14:1253–1261. [DOI] [PubMed] [Google Scholar]
- Casimiro S, Guise TA, and Chirgwin J. 2009. The critical role of the bone microenvironment in cancer metastases. Molecular and cellular endocrinology 310:71–81. [DOI] [PubMed] [Google Scholar]
- Chang HH, Chen BY, Wu CY, Tsao ZJ, Chen YY, Chang CP, Yang CR, and Lin DP. 2011. Hedgehog overexpression leads to the formation of prostate cancer stem cells with metastatic property irrespective of androgen receptor expression in the mouse model. J Biomed Sci 18:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q, Cai Z.-k., Chen Y.-b., Gu M, Zheng D.-c., Zhou J, and Wang Z. 2015. Poly r (C) binding protein-1 is central to maintenance of cancer stem cells in prostate cancer cells. Cellular Physiology and Biochemistry 35:1052–1061. [DOI] [PubMed] [Google Scholar]
- Choi YJ, Kim HS, Park SH, Kim BS, Kim KH, Lee HJ, Song HS, Shin DY, Lee HY, Kim HG, Lee KH, Lee JL, and Park KH. 2018. Phase II Study of Dovitinib in Patients with Castration-Resistant Prostate Cancer (KCSG-GU11–05). Cancer research and treatment : official journal of Korean Cancer Association 50:1252–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choudhury AD, Gray KP, Supko JG, Harshman LC, Taplin ME, Pace AF, Farina M, Zukotynski KA, Bernard B, Kantoff PW, Pomerantz M, and Sweeney C. 2018. A dose finding clinical trial of cabozantinib (XL184) administered in combination with abiraterone acetate in metastatic castration-resistant prostate cancer. Prostate [DOI] [PMC free article] [PubMed]
- Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, Ottesen LH, Laubscher KH, Sherman L, McDermott DF, Haas NB, Flaherty KT, Ross R, Eisenberg P, Meltzer PS, Merino MJ, Bottaro DP, Linehan WM, and Srinivasan R. 2013. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 31:181–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cojoc M, Peitzsch C, Trautmann F, Polishchuk L, Telegeev GD, and Dubrovska A. 2013. Emerging targets in cancer management: role of the CXCL12/CXCR4 axis. Onco Targets Ther 6:1347–1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coleman R 2016. Bone targeted treatments in cancer - The story so far. J Bone Oncol 5:90–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collins AT, Berry PA, Hyde C, Stower MJ, and Maitland NJ. 2005. Prospective identification of tumorigenic prostate cancer stem cells. Cancer research 65:10946–10951. [DOI] [PubMed] [Google Scholar]
- Conley-LaComb MK, Semaan L, Singareddy R, Li Y, Heath EI, Kim S, Cher ML, and Chinni SR. 2016. Pharmacological targeting of CXCL12/CXCR4 signaling in prostate cancer bone metastasis. Mol Cancer 15:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corn PG, Wang F, McKeehan WL, and Navone N. 2013. Targeting fibroblast growth factor pathways in prostate cancer. Clinical cancer research 19:5856–5866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corno C, Gatti L, Lanzi C, Zaffaroni N, Colombo D, and Perego P. 2016. Role of the receptor tyrosine kinase Axl and its targeting in cancer cells. Current medicinal chemistry 23:1496–1512. [DOI] [PubMed] [Google Scholar]
- Cui D, Dai J, Keller JM, Mizokami A, Xia S, and Keller ET. 2015. Notch Pathway Inhibition Using PF-03084014, a gamma-Secretase Inhibitor (GSI), Enhances the Antitumor Effect of Docetaxel in Prostate Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 21:4619–4629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Silva RF, Dhar D, Raina K, Kumar D, Kant R, Cagnon VHA, Agarwal C, and Agarwal R. 2018. Nintedanib inhibits growth of human prostate carcinoma cells by modulating both cell cycle and angiogenesis regulators. Sci Rep 8:9540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Silva RF, Nogueira-Pangrazi E, Kido LA, Montico F, Arana S, Kumar D, Raina K, Agarwal R, and Cagnon VHA. 2017. Nintedanib antiangiogenic inhibitor effectiveness in delaying adenocarcinoma progression in Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP). J Biomed Sci 24:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai J, Zhang H, Karatsinides A, Keller JM, Kozloff KM, Aftab DT, Schimmoller F, and Keller ET. 2014. Cabozantinib inhibits prostate cancer growth and prevents tumor-induced bone lesions. Clinical Cancer Research 20:617–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Das S, Harris LG, Metge BJ, Liu S, Riker AI, Samant RS, and Shevde LA. 2009. The hedgehog pathway transcription factor GLI1 promotes malignant behavior of cancer cells by up-regulating osteopontin. Journal of Biological Chemistry 284:22888–22897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Decker AM, Cackowski FC, Jung Y, and Taichman RS. 2017. Biochemical Changes in the Niche Following Tumor Cell Invasion. J Cell Biochem 118:1956–1964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delury C, Hart C, Brown M, Clarke N, and Parkin E. 2016. Stroma-induced Jagged1 expression drives PC3 prostate cancer cell migration; disparate effects of RIP-generated proteolytic fragments on cell behaviour and Notch signaling. Biochemical and biophysical research communications 472:255–261. [DOI] [PubMed] [Google Scholar]
- Denmeade SR, and Isaacs JT. 2002. A history of prostate cancer treatment. Nature reviews. Cancer 2:389–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiPersio JF, Micallef IN, Stiff PJ, Bolwell BJ, Maziarz RT, Jacobsen E, Nademanee A, McCarty J, Bridger G, and Calandra G. 2009. Phase III prospective randomized double-blind placebo-controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-Hodgkin’s lymphoma. Journal of Clinical Oncology 27:4767–4773. [DOI] [PubMed] [Google Scholar]
- Doré JJ, Edens M, Garamszegi N, and Leof EB. 1998. Heteromeric and homomeric transforming growth factor-β receptors show distinct signaling and endocytic responses in epithelial cells. Journal of Biological Chemistry 273:31770–31777. [DOI] [PubMed] [Google Scholar]
- Droz J-P, Medioni J, Chevreau C, De Mont-Serrat H, Merger M, Stopfer P, Kaiser R, and Oudard S. 2014. Randomized phase II study of nintedanib in metastatic castration-resistant prostate cancer postdocetaxel. Anti-cancer drugs 25:1081–1088. [DOI] [PubMed] [Google Scholar]
- Dubrovska A, Elliott J, Salamone RJ, Kim S, Aimone LJ, Walker JR, Watson J, Sauveur-Michel M, Garcia-Echeverria C, and Cho CY. 2010. Combination therapy targeting both tumor-initiating and differentiated cell populations in prostate carcinoma. Clinical Cancer Research 16:5692–5702. [DOI] [PubMed] [Google Scholar]
- Dubrovska A, Elliott J, Salamone RJ, Telegeev GD, Stakhovsky AE, Schepotin IB, Yan F, Wang Y, Bouchez LC, Kularatne SA, Watson J, Trussell C, Reddy VA, Cho CY, and Schultz PG. 2012. CXCR4 expression in prostate cancer progenitor cells 9. PloS one 7:e31226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubrovska A, Kim S, Salamone RJ, Walker JR, Maira SM, Garcia-Echeverria C, Schultz PG, and Reddy VA. 2009. The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proceedings of the National Academy of Sciences of the United States of America 106:268–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eccles SA, Aboagye EO, Ali S, Anderson AS, Armes J, Berditchevski F, Blaydes JP, Brennan K, Brown NJ, and Bryant HE. 2013. Critical research gaps and translational priorities for the successful prevention and treatment of breast cancer. Breast Cancer Research 15:R92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng S, Shao L, Yu W, Gavine P, and Ittmann M. 2012. Targeting fibroblast growth factor receptor signaling inhibits prostate cancer progression. Clinical cancer research : an official journal of the American Association for Cancer Research 18:3880–3888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Festuccia C, Angelucci A, Gravina GL, Villanova I, Teti A, Abini A, and Bologna M. 2000. Osteoblast‐derived TGF‐β1 modulates matrix degrading protease expression and activity in prostate cancer cells. International journal of cancer 85:407–415. [PubMed] [Google Scholar]
- Germann M, Wetterwald A, Guzmán‐Ramirez N, van der Pluijm G, Culig Z, Cecchini MG, Williams ED, and Thalmann GN. 2012. Stem‐like cells with luminal progenitor phenotype survive castration in human prostate cancer. Stem cells 30:1076–1086. [DOI] [PubMed] [Google Scholar]
- Goruppi S, Ruaro E, Varnum B, and Schneider C. 1999. Gas6-mediated survival in NIH3T3 cells activates stress signalling cascade and is independent of Ras. Oncogene 18:4224. [DOI] [PubMed] [Google Scholar]
- Graham TJ, Box G, Tunariu N, Crespo M, Spinks TJ, Miranda S, Attard G, de Bono J, Eccles SA, and Davies FE. 2014. Preclinical evaluation of imaging biomarkers for prostate cancer bone metastasis and response to cabozantinib. Journal of the National Cancer Institute 106:dju033. [DOI] [PubMed] [Google Scholar]
- Gravina GL, Mancini A, Muzi P, Ventura L, Biordi L, Ricevuto E, Pompili S, Mattei C, Di Cesare E, and Jannini EA. 2015. CXCR4 pharmacogical inhibition reduces bone and soft tissue metastatic burden by affecting tumor growth and tumorigenic potential in prostate cancer preclinical models. The Prostate 75:1227–1246. [DOI] [PubMed] [Google Scholar]
- Grönberg H 2003. Prostate cancer epidemiology. The Lancet 361:859–864. [DOI] [PubMed] [Google Scholar]
- Gu J-W, Rizzo P, Pannuti A, Golde T, Osborne B, and Miele L. 2012. Notch signals in the endothelium and cancer” stem-like” cells: opportunities for cancer therapy. Vascular cell 4:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, and Lander ES. 2009. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 138:645–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, Papamatheakis J, and Kretsovali A. 2015. Common stemness regulators of embryonic and cancer stem cells. World J Stem Cells 7:1150–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haider M-T, Hunter KD, Robinson SP, Graham TJ, Corey E, Dear TN, Hughes R, Brown NJ, and Holen I. 2015. Rapid modification of the bone microenvironment following short-term treatment with Cabozantinib in vivo. Bone 81:581–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall CL, Daignault SD, Shah RB, Pienta KJ, and Keller ET. 2008. Dickkopf‐1 expression increases early in prostate cancer development and decreases during progression from primary tumor to metastasis. The Prostate 68:1396–1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall CL, Kang S, MacDougald OA, and Keller ET. 2006. Role of Wnts in prostate cancer bone metastases. Journal of cellular biochemistry 97:661–672. [DOI] [PubMed] [Google Scholar]
- Han L, Shi S, Gong T, Zhang Z, and Sun X. 2013. Cancer stem cells: therapeutic implications and perspectives in cancer therapy. Acta Pharmaceutica Sinica B 3:65–75. [Google Scholar]
- Holen I 2016. The bone microenvironment - Multiple players involved in cancer progression. J Bone Oncol 5:87–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horner A, Kemp P, Summers C, Bord S, Bishop N, Kelsall A, Coleman N, and Compston J. 1998. Expression and distribution of transforming growth factor-β isoforms and their signaling receptors in growing human bone. Bone 23:95–102. [DOI] [PubMed] [Google Scholar]
- Hsieh I-S, Chang K-C, Tsai Y-T, Ke J-Y, Lu P-J, Lee K-H, Yeh S-D, Hong T-M, and Chen Y-L. 2012. MicroRNA-320 suppresses the stem cell-like characteristics of prostate cancer cells by downregulating the Wnt/beta-catenin signaling pathway. Carcinogenesis 34:530–538. [DOI] [PubMed] [Google Scholar]
- Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, and Farrar WL. 2008. CD44+ CD24(−) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer 98:756–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishino R, Minami K, Tanaka S, Nagai M, Matsui K, Hasegawa N, Roeder RG, Asano S, and Ito M. 2013. FGF7 supports hematopoietic stem and progenitor cells and niche-dependent myeloblastoma cells via autocrine action on bone marrow stromal cells in vitro. Biochemical and biophysical research communications 440:125–131. [DOI] [PubMed] [Google Scholar]
- Itkin T, Kaufmann KB, Gur-Cohen S, Ludin A, and Lapidot T. 2013. Fibroblast growth factor signaling promotes physiological bone remodeling and stem cell self-renewal. Current opinion in hematology 20:237–244. [DOI] [PubMed] [Google Scholar]
- Izumi K, Mizokami A, Li YQ, Narimoto K, Sugimoto K, Kadono Y, Kitagawa Y, Konaka H, Koh E, and Keller ET. 2009. Tranilast inhibits hormone refractory prostate cancer cell proliferation and suppresses transforming growth factor β1‐associated osteoblastic changes. The prostate 69:1222–1234. [DOI] [PubMed] [Google Scholar]
- Jaworska D, Król W, and Szliszka E. 2015. Prostate cancer stem cells: research advances. International journal of molecular sciences 16:27433–27449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeter CR, Liu B, Liu X, Chen X, Liu C, Calhoun-Davis T, Repass J, Zaehres H, Shen JJ, and Tang DG. 2011. NANOG promotes cancer stem cell characteristics and prostate cancer resistance to androgen deprivation. Oncogene 30:3833–3845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Y, Dai J, Zhang H, Sottnik JL, Keller JM, Escott KJ, Sanganee HJ, Yao Z, McCauley LK, and Keller ET. 2013. Activation of the Wnt pathway through AR79, a GSK3beta inhibitor, promotes prostate cancer growth in soft tissue and bone. Molecular cancer research : MCR 11:1597–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jimeno A, Gordon M, Chugh R, Messersmith W, Mendelson D, Dupont J, Stagg R, Kapoun AM, Xu L, Uttamsingh S, Brachmann RK, and Smith DC. 2017. A First-in-Human Phase I Study of the Anticancer Stem Cell Agent Ipafricept (OMP-54F28), a Decoy Receptor for Wnt Ligands, in Patients with Advanced Solid Tumors. Clinical cancer research : an official journal of the American Association for Cancer Research 23:7490–7497. [DOI] [PubMed] [Google Scholar]
- Jung Y, Cackowski FC, Yumoto K, Decker AM, Wang J, Kim JK, Lee E, Wang Y, Chung JS, and Gursky AM. 2018. CXCL12g promotes metastatic castration-resistant prostate cancer by inducing cancer stem cell and neuroendocrine phenotypes. Cancer research 78:2026–2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jung Y, Decker AM, Wang J, Lee E, Kana LA, Yumoto K, Cackowski FC, Rhee J, Carmeliet P, Buttitta L, Morgan TM, and Taichman RS. 2016. Endogenous GAS6 and Mer receptor signaling regulate prostate cancer stem cells in bone marrow. Oncotarget 7:25698–25711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jung Y, Wang J, Lee E, McGee S, Berry JE, Yumoto K, Dai J, Keller ET, Shiozawa Y, and Taichman RS. 2015. Annexin 2-CXCL12 interactions regulate metastatic cell targeting and growth in the bone marrow. Mol Cancer Res 13:197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kesper DA, Didt‐Koziel L, and Vortkamp A. 2010. Gli2 activator function in preosteoblasts is sufficient to mediate ihh‐dependent osteoblast differentiation, whereas the repressor function of Gli2 is dispensable for endochondral ossification. Developmental Dynamics 239:1818–1826. [DOI] [PubMed] [Google Scholar]
- Klarmann GJ, Hurt EM, Mathews LA, Zhang X, Duhagon MA, Mistree T, Thomas SB, and Farrar WL. 2009. Invasive prostate cancer cells are tumor initiating cells that have a stem cell-like genomic signature. Clinical & experimental metastasis 26:433–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kobayashi A, Okuda H, Xing F, Pandey PR, Watabe M, Hirota S, Pai SK, Liu W, Fukuda K, Chambers C, Wilber A, and Watabe K. 2011. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. The Journal of experimental medicine 208:2641–2655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kobayashi Y, Maeda K, and Takahashi N. 2008. Roles of Wnt signaling in bone formation and resorption. Japanese Dental Science Review 44:76–82. [Google Scholar]
- Koeneman KS, Yeung F, and Chung LW. 1999. Osteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. The Prostate 39:246–261. [DOI] [PubMed] [Google Scholar]
- Kong D, Banerjee S, Ahmad A, Li Y, Wang Z, Sethi S, and Sarkar FH. 2010. Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PloS one 5:e12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwabi-Addo B, Ozen M, and Ittmann M. 2004. The role of fibroblast growth factors and their receptors in prostate cancer. Endocrine-related cancer 11:709–724. [DOI] [PubMed] [Google Scholar]
- Lataillade JJ, Domenech J, and Le Bousse-Kerdiles MC. 2004. Stromal cell-derived factor-1 (SDF-1)\CXCR4 couple plays multiple roles on haematopoietic progenitors at the border between the old cytokine and new chemokine worlds: survival, cell cycling and trafficking. European cytokine network 15:177–188. [PubMed] [Google Scholar]
- Lauth M, Bergström Å, Shimokawa T, and Toftgård R. 2007. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proceedings of the National Academy of Sciences 104:8455–8460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebrun JJ 2012. The Dual Role of TGFbeta in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN Mol Biol 2012:381428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee GT, Hong JH, Mueller TJ, Watson JA, Kwak C, Sheen YY, Kim D-K, Kim SJ, and Kim IY. 2008. Effect of IN-1130, a small molecule inhibitor of transforming growth factor-β type I receptor/activin receptor-like kinase-5, on prostate cancer cells. The Journal of urology 180:2660–2667. [DOI] [PubMed] [Google Scholar]
- Lee M, Hong H, Kim W, Zhang L, Friedlander TW, Fong L, Lin AM, Small EJ, Wei XX, and Rodvelt TJ. 2019. Itraconazole as a Noncastrating Treatment for Biochemically Recurrent Prostate Cancer: A Phase 2 Study. Clinical genitourinary cancer 17:e92–e96. [DOI] [PubMed] [Google Scholar]
- Leibowitz-Amit R, Pintilie M, Khoja L, Azad AA, Berger R, Laird AD, Aftab DT, Chi KN, and Joshua AM. 2016. Changes in plasma biomarkers following treatment with cabozantinib in metastatic castration-resistant prostate cancer: a post hoc analysis of an extension cohort of a phase II trial. Journal of translational medicine 14:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lescarbeau RM, Seib FP, Prewitz M, Werner C, and Kaplan DL. 2012. In vitro model of metastasis to bone marrow mediates prostate cancer castration resistant growth through paracrine and extracellular matrix factors. PloS one 7:e40372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Ye L, Guo W, Wang M, Huang S, and Peng X. 2017. PHF21B overexpression promotes cancer stem cell-like traits in prostate cancer cells by activating the Wnt/β-catenin signaling pathway. Journal of Experimental & Clinical Cancer Research 36:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Cozzi P, Graham P, Bucci J, and Kearsley J. 2013. Cancer Stem Cells “The Root of Radioresistance” in Prostate Cancer Radiotherapy. World Journal of Cancer Research 1:85–88. [Google Scholar]
- Li ZG, Mathew P, Yang J, Starbuck MW, Zurita AJ, Liu J, Sikes C, Multani AS, Efstathiou E, Lopez A, Wang J, Fanning TV, Prieto VG, Kundra V, Vazquez ES, Troncoso P, Raymond AK, Logothetis CJ, Lin SH, Maity S, and Navone NM. 2008. Androgen receptor-negative human prostate cancer cells induce osteogenesis in mice through FGF9-mediated mechanisms. J Clin Invest 118:2697–2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin JZ, Wang ZJ, De W, Zheng M, Xu WZ, Wu HF, Armstrong A, and Zhu JG. 2017. Targeting AXL overcomes resistance to docetaxel therapy in advanced prostate cancer. Oncotarget 8:41064–41077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logothetis CJ, and Lin SH. 2005. Osteoblasts in prostate cancer metastasis to bone. Nature reviews. Cancer 5:21–28. [DOI] [PubMed] [Google Scholar]
- Ma F, Ye H, He HH, Gerrin SJ, Chen S, Tanenbaum BA, Cai C, Sowalsky AG, He L, and Wang H. 2016. SOX9 drives WNT pathway activation in prostate cancer. The Journal of clinical investigation 126:1745–1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maier GS, Eberhardt C, and Kurth AA. 2016. Ibandronate: The loading dose concept in the treatment of metastatic bone pain. Journal of bone oncology 5:1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massague J 2012. TGFbeta signalling in context. Nature reviews. Molecular cell biology 13:616–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maughan BL, Suzman DL, Luber B, Wang H, Glavaris S, Hughes R, Sullivan R, Harb R, Boudadi K, and Paller C. 2016. Pharmacodynamic study of the oral hedgehog pathway inhibitor, vismodegib, in patients with metastatic castration-resistant prostate cancer. Cancer chemotherapy and pharmacology 78:1297–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng X, Vander Ark A, Daft P, Woodford E, Wang J, Madaj Z, and Li X. 2018. Loss of TGF-β signaling in osteoblasts increases basic-FGF and promotes prostate cancer bone metastasis. Cancer letters 418:109–118. [DOI] [PubMed] [Google Scholar]
- Miles FL, Tung NS, Aguiar AA, Kurtoglu S, and Sikes RA. 2012. Increased TGF‐β1‐ mediated suppression of growth and motility in castrate‐resistant prostate cancer cells is consistent with Smad2/3 signaling. The Prostate 72:1339–1350. [DOI] [PubMed] [Google Scholar]
- Moltzahn F, and Thalmann GN. 2013. Cancer stem cells in prostate cancer. Translational andrology and urology 2:242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nanta R, Kumar D, Meeker D, Rodova M, Van Veldhuizen P, Shankar S, and Srivastava R. 2013. NVP-LDE-225 (Erismodegib) inhibits epithelial–mesenchymal transition and human prostate cancer stem cell growth in NOD/SCID IL2Rγ null mice by regulating Bmi-1 and microRNA-128. Oncogenesis 2:e42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogawa K, Yoshioka Y, Isohashi F, Seo Y, Yoshida K, and Yamazaki H. 2013. Radiotherapy targeting cancer stem cells: current views and future perspectives. Anticancer research 33:747–754. [PubMed] [Google Scholar]
- Ojo D, Lin X, Wong N, Gu Y, and Tang D. 2015. Prostate cancer stem-like cells contribute to the development of castration-resistant prostate cancer. Cancers 7:2290–2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pai VC, Hsu CC, Chan TS, Liao WY, Chuu CP, Chen WY, Li CR, Lin CY, Huang SP, and Chen LT. 2019. Correction: ASPM promotes prostate cancer stemness and progression by augmenting Wnt-Dvl-3-β-catenin signaling. Oncogene 38:1354–1354. [DOI] [PubMed] [Google Scholar]
- Paller C, Pu H, Begemann DE, Wade CA, Hensley PJ, and Kyprianou N. 2018. TGF‐β receptor I inhibitor enhances response to enzalutamide in a pre‐clinical model of advanced prostate cancer. The Prostate [DOI] [PMC free article] [PubMed]
- Patrawala L, Calhoun-Davis T, Schneider-Broussard R, and Tang DG. 2007. Hierarchical organization of prostate cancer cells in xenograft tumors: the CD44+ α2β1+ cell population is enriched in tumor-initiating cells. Cancer research 67:6796–6805. [DOI] [PubMed] [Google Scholar]
- Pedersen EA, Shiozawa Y, Mishra A, and Taichman RS. 2012. Structure and function of the solid tumor niche. Frontiers in bioscience (Scholar edition) 4:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin J, Liu X, Laffin B, Chen X, Choy G, Jeter CR, Calhoun-Davis T, Li H, Palapattu GS, Pang S, Lin K, Huang J, Ivanov I, Li W, Suraneni MV, and Tang DG. 2012. The PSA(-/lo) prostate cancer cell population harbors self-renewing long-term tumor-propagating cells that resist castration. Cell Stem Cell 10:556–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu S, Deng L, Bao Y, Jin K, Tu X, Li J, Liao X, Liu Z, Yang L, and Wei Q. 2018. Reversal of docetaxel resistance in prostate cancer by Notch signaling inhibition. Anti-cancer drugs 29:871–879. [DOI] [PubMed] [Google Scholar]
- Rimkus T, Carpenter R, Qasem S, Chan M, and Lo H-W. 2016. Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors. Cancers 8:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross AE, Hughes RM, Glavaris S, Ghabili K, He P, Anders NM, Harb R, Tosoian JJ, Marchionni L, Schaeffer EM, Partin AW, Allaf ME, Bivalacqua TJ, Chapman C, O’Neal T, DeMarzo AM, Hurley PJ, Rudek MA, and Antonarakis ES. 2017. Pharmacodynamic and pharmacokinetic neoadjuvant study of hedgehog pathway inhibitor Sonidegib (LDE-225) in men with high-risk localized prostate cancer undergoing prostatectomy. Oncotarget 8:104182–104192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossi D, and Zlotnik A. 2000. The biology of chemokines and their receptors. Annual review of immunology 18:217–242. [DOI] [PubMed] [Google Scholar]
- Ruppender N, Larson S, Lakely B, Kollath L, Brown L, Coleman I, Coleman R, Nguyen H, Nelson PS, and Corey E. 2015. Cellular adhesion promotes prostate cancer cells escape from dormancy. PLoS One 10:e0130565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sethi S, Macoska J, Chen W, and Sarkar FH. 2010. Molecular signature of epithelial-mesenchymal transition (EMT) in human prostate cancer bone metastasis. Am J Transl Res 3:90–99. [PMC free article] [PubMed] [Google Scholar]
- Shariat SF, Shalev M, Menesses-Diaz A, Kim IY, Kattan MW, Wheeler TM, and Slawin KM. 2001. Preoperative plasma levels of transforming growth factor beta(1) (TGF-beta(1)) strongly predict progression in patients undergoing radical prostatectomy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 19:2856–2864. [DOI] [PubMed] [Google Scholar]
- Sharpe B, Beresford M, Bowen R, Mitchard J, and Chalmers AD. 2013. Searching for prostate cancer stem cells: markers and methods. Stem Cell Reviews and Reports 9:721–730. [DOI] [PubMed] [Google Scholar]
- Shiozawa Y, Berry JE, Eber MR, Jung Y, Yumoto K, Cackowski FC, Yoon HJ, Parsana P, Mehra R, Wang J, McGee S, Lee E, Nagrath S, Pienta KJ, and Taichman RS. 2016. The marrow niche controls the cancer stem cell phenotype of disseminated prostate cancer. Oncotarget 7:41217–41232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, and Ziegler AM. 2011. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. The Journal of clinical investigation 121:1298–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shiozawa Y, Pedersen EA, Patel LR, Ziegler AM, Havens AM, Jung Y, Wang J, Zalucha S, Loberg RD, Pienta KJ, and Taichman RS. 2010. GAS6/AXL axis regulates prostate cancer invasion, proliferation, and survival in the bone marrow niche. Neoplasia 12:116–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith DC, Smith MR, Sweeney C, Elfiky AA, Logothetis C, Corn PG, Vogelzang NJ, Small EJ, Harzstark AL, Gordon MS, Vaishampayan UN, Haas NB, Spira AI, Lara PN Jr., Lin CC, Srinivas S, Sella A, Schoffski P, Scheffold C, Weitzman AL, and Hussain M. 2013. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 31:412–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, Krainer M, Bergman A, Hoelzer W, De Wit R, Bogemann M, Saad F, Cruciani G, Thiery-Vuillemin A, Feyerabend S, Miller K, Houede N, Hussain S, Lam E, Polikoff J, Stenzl A, Mainwaring P, Ramies D, Hessel C, Weitzman A, and Fizazi K. 2016. Phase III Study of Cabozantinib in Previously Treated Metastatic Castration-Resistant Prostate Cancer: COMET-1. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34:3005–3013. [DOI] [PubMed] [Google Scholar]
- Smith MR, Sweeney CJ, Corn PG, Rathkopf DE, Smith DC, Hussain M, George DJ, Higano CS, Harzstark AL, Sartor AO, Vogelzang NJ, Gordon MS, de Bono JS, Haas NB, Logothetis CJ, Elfiky A, Scheffold C, Laird AD, Schimmoller F, Basch EM, and Scher HI. 2014. Cabozantinib in chemotherapy-pretreated metastatic castration-resistant prostate cancer: results of a phase II nonrandomized expansion study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 32:3391–3399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stern PH, and Alvares K. 2014. Antitumor Agent Cabozantinib Decreases RANKL Expression in Osteoblastic Cells and Inhibits Osteoclastogenesis and PTHrP‐Stimulated Bone Resorption. Journal of cellular biochemistry 115:2033–2038. [DOI] [PubMed] [Google Scholar]
- Stoyanova T, Riedinger M, Lin S, Faltermeier CM, Smith BA, Zhang KX, Going CC, Goldstein AS, Lee JK, and Drake JM. 2016. Activation of Notch1 synergizes with multiple pathways in promoting castration-resistant prostate cancer. Proceedings of the National Academy of Sciences 113:E6457–E6466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taichman RS, Patel LR, Bedenis R, Wang J, Weidner S, Schumann T, Yumoto K, Berry JE, Shiozawa Y, and Pienta KJ. 2013. GAS6 receptor status is associated with dormancy and bone metastatic tumor formation. PloS one 8:e61873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX, and Ivy SP. 2015. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nature reviews. Clinical oncology 12:445–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takeishi S, and Nakayama KI. 2016. To wake up cancer stem cells, or to let them sleep, that is the question. Cancer science 107:875–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trautmann F, Cojoc M, Kurth I, Melin N, Bouchez LC, Dubrovska A, and Peitzsch C. 2014. CXCR4 as biomarker for radioresistant cancer stem cells. International journal of radiation biology 90:687–699. [DOI] [PubMed] [Google Scholar]
- Tsujimura A, Koikawa Y, Salm S, Takao T, Coetzee S, Moscatelli D, Shapiro E, Lepor H, Sun T-T, and Wilson EL. 2002. Proximal location of mouse prostate epithelial stem cells: a model of prostatic homeostasis. The Journal of cell biology 157:1257–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valta MP, Tuomela J, Bjartell A, Valve E, Väänänen HK, and Härkönen P. 2008. FGF‐8 is involved in bone metastasis of prostate cancer. International journal of cancer 123:22–31. [DOI] [PubMed] [Google Scholar]
- Varkaris A, Corn PG, Gaur S, Dayyani F, Logothetis CJ, and Gallick GE. 2011. The role of HGF/c-Met signaling in prostate cancer progression and c-Met inhibitors in clinical trials. Expert opinion on investigational drugs 20:1677–1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wan X, Corn PG, Yang J, Palanisamy N, Starbuck MW, Efstathiou E, Tapia EMLN, Zurita AJ, Aparicio A, and Ravoori MK. 2014. Prostate cancer cell–stromal cell crosstalk via FGFR1 mediates antitumor activity of dovitinib in bone metastases. Science translational medicine 6:252ra122–252ra122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wan X, Li Z-G, Yingling JM, Yang J, Starbuck MW, Ravoori MK, Kundra V, Vazquez E, and Navone NM. 2012. Effect of transforming growth factor beta (TGF-β) receptor I kinase inhibitor on prostate cancer bone growth. Bone 50:695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang N, Docherty F, Brown HK, Reeves K, Fowles A, Lawson M, Ottewell PD, Holen I, Croucher PI, and Eaton CL. 2015a. Mitotic quiescence, but not unique “stemness,” marks the phenotype of bone metastasis-initiating cells in prostate cancer. FASEB J 29:3141–3150. [DOI] [PubMed] [Google Scholar]
- Wang N, Docherty F, Brown HK, Reeves K, Fowles A, Lawson M, Ottewell PD, Holen I, Croucher PI, and Eaton CL. 2015b. Mitotic quiescence, but not unique “stemness,” marks the phenotype of bone metastasis-initiating cells in prostate cancer. The FASEB Journal 29:3141–3150. [DOI] [PubMed] [Google Scholar]
- West AF, O’Donnell M, Charlton RG, Neal DE, and Leung HY. 2001. Correlation of vascular endothelial growth factor expression with fibroblast growth factor-8 expression and clinico-pathologic parameters in human prostate cancer. Br J Cancer 85:576–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yadav SS, Li J, Stockert JA, Herzog B, O’Connor J, Garzon-Manco L, Parsons R, Tewari AK, and Yadav KK. 2017. Induction of neuroendocrine differentiation in prostate cancer cells by dovitinib (TKI-258) and its therapeutic implications. Translational oncology 10:357–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yardy GW, and Brewster SF. 2005. Wnt signalling and prostate cancer. Prostate Cancer Prostatic Dis 8:119–126. [DOI] [PubMed] [Google Scholar]
- Zhang K, Guo Y, Wang X, Zhao H, Ji Z, Cheng C, Li L, Fang Y, Xu D, and Zhu HH. 2017a. WNT/β-catenin directs self-renewal symmetric cell division of hTERThigh prostate cancer stem cells. Cancer research 77:2534–2547. [DOI] [PubMed] [Google Scholar]
- Zhang L, Sha J, Yang G, Huang X, Bo J, and Huang Y. 2017b. Activation of Notch pathway is linked with epithelial-mesenchymal transition in prostate cancer cells. Cell Cycle 16:999–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Cheng L, Li J, Farah E, Atallah NM, Pascuzzi PE, Gupta S, and Liu X. 2018. Inhibition of the Wnt/beta-Catenin Pathway Overcomes Resistance to Enzalutamide in Castration-Resistant Prostate Cancer. Cancer Res 78:3147–3162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z, Pu F, and Shao Z. 2017c. The skeletal-related events of denosumab versus zoledronic acid in patients with bone metastases: A meta-analysis of randomized controlled trials. Journal of bone oncology 9:21–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao M, Qiao M, Harris SE, Chen D, Oyajobi BO, and Mundy GR. 2006. The zinc finger transcription factor Gli2 mediates bone morphogenetic protein 2 expression in osteoblasts in response to hedgehog signaling. Molecular and cellular biology 26:6197–6208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou G, Yin L-C, Wang D-W, Li L, Pei S, Gentle IR, Li F, and Cheng H-M. 2013. Fibrous hybrid of graphene and sulfur nanocrystals for high-performance lithium–sulfur batteries. Acs Nano 7:5367–5375. [DOI] [PubMed] [Google Scholar]
- Zou J, Hong Y, Tong Y, Wei J, Qin Y, Shao S, Wang C, and Zhou K. 2015. Sonic hedgehog produced by bone marrow-derived mesenchymal stromal cells supports cell survival in myelodysplastic syndrome. Stem Cells Int 2015:957502. [DOI] [PMC free article] [PubMed] [Google Scholar]
