pathogenesis of osteoblastic bone metastases from prostate cancer

13
Pathogenesis of Osteoblastic Bone Metastases From Prostate Cancer Toni Ibrahim, MD, PhD; Emanuela Flamini, BSc; Laura Mercatali, BSc; Emanuele Sacanna, BSc; Patrizia Serra, BSc; and Dino Amadori, MD Prostate cancer is the second leading cause of cancer-related death in men. A typical feature of this disease is its ability to metastasize to bone. It is mainly osteosclerotic, and is caused by a relative excess of osteoblast activity, leading to an abnormal bone formation. Bone metastases are the result of a complex series of steps that are not yet fully understood and depend on dynamic crosstalk between metastatic cancer cells, cellular components of the bone marrow microenvironment, and bone matrix (osteoblasts and osteoclasts). Prostate cancer cells from primary tissue undergo an epithelial-mesenchymal transition to disseminate and acquire a bone-like phenotype to metastasize in bone tissue. This review discusses the biological processes and the molecules involved in the progression of bone metastases. Here we focus on the routes of osteoblast differentiation and activation, the crosstalk between bone cells and tumor cells, and the molecules involved in these processes that are expressed by both osteoblasts and tumor cells. Furthermore, this review deals with the recently elucidated role of osteoclasts in prostate cancer bone metasta- ses. Certainly, to better understand the underlying mechanisms of bone metastasis and so improve targeted bone therapies, further studies are warranted to shed light on the probable role of the premetastatic niche and the involve- ment of cancer stem cells. Cancer 2010;116:1406–18. V C 2010 American Cancer Society . KEYWORDS: osteoblasts, osteoclasts, prostate cancer, bone, metastasization. Solid tumors, such as breast and prostate cancer, have an affinity to metastasize to bone, causing osteolysis and abnor- mal bone formation. Bone metastasis starts with the tropism of cancer cells to the bone through specific migratory and invasive processes. Once in the bone marrow, metastatic cells are able to survive and grow. Here, they actively interact with bone marrow stem cells and hematopoietic progenitors in the so-called metastatic niche, where they acquire a bone- like phenotype. This leads to the formation of bone lesions (lytic or osteoblastic), obtained through reciprocal paracrine amplification and cell-to-cell communication with bone cells. The complex molecular pathogenesis mechanisms of bone metastasis offer several potential targets for prevention and therapy. Prostate cancer is the second leading cause of cancer-related death in men, and a typical feature of this disease is its ability to metastasize to bone. Indeed, it has been estimated that >80% of men who die from prostate cancer develop bone metastases. 1,2 Although most bone metastases from prostate cancer have been classified as osteoblastic, based on the radiographic appearance of lesions, it is clear that bone resorption and bone formation are dysregulated. 3-5 The Metastatic Process: From Primary Tumor to Growth in Bone Tissue The first step in metastasization is the acquisition of motility and invasiveness; capabilities that are not compatible with nor- mal tissue. Cancer cells must therefore shed many of their epithelial characteristics, detach from epithelial sheets, and undergo a drastic alteration, a process referred to as the epithelial-mesenchymal transition. 6 Acquisition of this invasive phe- notype is reminiscent of that which occurs during early embryonic development. 7 In malignancy, genetic alterations and the tumor environment can both induce epithelial-mesenchymal transition (EMT) in tumor cells. The important steps that facil- itate metastasis appear to be reversible, 7 and cannot be explained solely by irreversible genetic alterations, indicating the exis- tence of a dynamic component to human tumor progression. In cancer, although the PI3K/Akt pathway is the primary inducer of epithelial-mesenchymal transition, the Wnt/B-catenin, Notch, Ras, integrin-linked kinase, and integrin signaling DOI: 10.1002/cncr.24896, Received: May 14, 2009; Revised: July 2, 2009; Accepted: July 17, 2009, Published online January 27, 2010 in Wiley InterScience (www.interscience.wiley.com) Corresponding author: Dino Amadori, MD, Osteo-oncology Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, via Piero Maroncelli 40, 47014 Meldola (FC), Italy; Fax: (011) 39 0543 739249; [email protected] Osteo-oncology Center, Scientific Institute of Romagnolo for the Study and Cure of Tumors, Meldola, Italy We thank Dr. Ian Seymour for help with editing the article. 1406 Cancer March 15, 2010 Review Article

Upload: toni-ibrahim

Post on 11-Jun-2016

213 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Pathogenesis of osteoblastic bone metastases from prostate cancer

Pathogenesis of Osteoblastic Bone MetastasesFrom Prostate CancerToni Ibrahim, MD, PhD; Emanuela Flamini, BSc; Laura Mercatali, BSc; Emanuele Sacanna, BSc; Patrizia Serra, BSc;

and Dino Amadori, MD

Prostate cancer is the second leading cause of cancer-related death in men. A typical feature of this disease is its

ability to metastasize to bone. It is mainly osteosclerotic, and is caused by a relative excess of osteoblast activity,

leading to an abnormal bone formation. Bone metastases are the result of a complex series of steps that are not yet

fully understood and depend on dynamic crosstalk between metastatic cancer cells, cellular components of the bone

marrow microenvironment, and bone matrix (osteoblasts and osteoclasts). Prostate cancer cells from primary tissue

undergo an epithelial-mesenchymal transition to disseminate and acquire a bone-like phenotype to metastasize in

bone tissue. This review discusses the biological processes and the molecules involved in the progression of bone

metastases. Here we focus on the routes of osteoblast differentiation and activation, the crosstalk between bone cells

and tumor cells, and the molecules involved in these processes that are expressed by both osteoblasts and tumor

cells. Furthermore, this review deals with the recently elucidated role of osteoclasts in prostate cancer bone metasta-

ses. Certainly, to better understand the underlying mechanisms of bone metastasis and so improve targeted bone

therapies, further studies are warranted to shed light on the probable role of the premetastatic niche and the involve-

ment of cancer stem cells. Cancer 2010;116:1406–18. VC 2010 American Cancer Society.

KEYWORDS: osteoblasts, osteoclasts, prostate cancer, bone, metastasization.

Solid tumors, such as breast and prostate cancer, have an affinity to metastasize to bone, causing osteolysis and abnor-mal bone formation. Bone metastasis starts with the tropism of cancer cells to the bone through specific migratory andinvasive processes. Once in the bone marrow, metastatic cells are able to survive and grow. Here, they actively interactwith bone marrow stem cells and hematopoietic progenitors in the so-called metastatic niche, where they acquire a bone-like phenotype. This leads to the formation of bone lesions (lytic or osteoblastic), obtained through reciprocal paracrineamplification and cell-to-cell communication with bone cells. The complex molecular pathogenesis mechanisms of bonemetastasis offer several potential targets for prevention and therapy.

Prostate cancer is the second leading cause of cancer-related death in men, and a typical feature of this disease is itsability to metastasize to bone. Indeed, it has been estimated that >80% of men who die from prostate cancer developbone metastases.1,2 Although most bone metastases from prostate cancer have been classified as osteoblastic, based on theradiographic appearance of lesions, it is clear that bone resorption and bone formation are dysregulated.3-5

The Metastatic Process: From Primary Tumor to Growth in Bone Tissue

The first step in metastasization is the acquisition of motility and invasiveness; capabilities that are not compatible with nor-mal tissue. Cancer cells must therefore shed many of their epithelial characteristics, detach from epithelial sheets, andundergo a drastic alteration, a process referred to as the epithelial-mesenchymal transition.6 Acquisition of this invasive phe-notype is reminiscent of that which occurs during early embryonic development.7 In malignancy, genetic alterations and thetumor environment can both induce epithelial-mesenchymal transition (EMT) in tumor cells. The important steps that facil-itate metastasis appear to be reversible,7 and cannot be explained solely by irreversible genetic alterations, indicating the exis-tence of a dynamic component to human tumor progression. In cancer, although the PI3K/Akt pathway is the primaryinducer of epithelial-mesenchymal transition, the Wnt/B-catenin, Notch, Ras, integrin-linked kinase, and integrin signaling

DOI: 10.1002/cncr.24896, Received: May 14, 2009; Revised: July 2, 2009; Accepted: July 17, 2009, Published online January 27, 2010 in Wiley InterScience

(www.interscience.wiley.com)

Corresponding author: Dino Amadori, MD, Osteo-oncology Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, via Piero Maroncelli 40,

47014 Meldola (FC), Italy; Fax: (011) 39 0543 739249; [email protected]

Osteo-oncology Center, Scientific Institute of Romagnolo for the Study and Cure of Tumors, Meldola, Italy

We thank Dr. Ian Seymour for help with editing the article.

1406 Cancer March 15, 2010

Review Article

Page 2: Pathogenesis of osteoblastic bone metastases from prostate cancer

pathways are also involved.8-11 The main characteristics ofepithelial-mesenchymal transition in prostate cancer cells,and the complex molecular interactions that facilitate thisprocess, are summarized in Figure 1.

After epithelial-mesenchymal transition, prostatecancer cells must go through a multistep process to meta-stasize to bone, which involves dislodgement from a pri-mary site, survival in the circulation, binding to theresident cells in bone, and survival and proliferation in thebone and bone marrow.12-18 The dissemination of pros-tate cancer cells may take place early in disease progressionwith tumor cells preferentially engaged in the bone mar-row, and a subset of cells surviving and evolving into clini-cally apparent disease. These cells then enter a period ofdormancy in which they either stop proliferating, or pro-liferate at a reduced rate before showing evidence of me-tastasis; a process that can sometimes exceed 10 years.19-23

However, in some situations, there is at least 1 further andcrucial event that takes place, the trigger that reactivatestumor cell dormancy. However, the mechanisms thatfacilitate this process remain unknown.

Circulating prostate cancer cells preferentiallyadhere to bone marrow endothelial cells and then migrate

through the endothelial layer.24 This process involves avariety of adhesion molecules (eg, selectins, integrins, andcadherins) present on the surfaces of endothelial and met-astatic prostate cancer cells. Cell adhesion and migrationare mediated in part by many integrin-extracellular matrixinteractions, especially those involving integrin avb3,whose expression is enhanced by stromal cell-derived fac-tor-1 (SDF-1).25,26 Several lines of evidence suggest thatSDF-1 contributes to the pathogenesis of prostate cancermetastases. Inhibition of chemokines reduces the in vitroproliferation of PC-3 cells. Expression of the SDF-1 re-ceptor, CXCR4, has been detected in prostate cancer cells,and SDF-1 has also been reported to increase their migra-tion capacity.1,27 This evidence implies that several boneparacrine factors regulate the expression of adhesion mole-cules in disseminated prostate cancer cells, and supportsthe interaction between cancer cells and resident bonecells, thereby contributing to the tropism of circulatingprostate cancer cells toward bone. Cadherin 11 has beenimplicated in this process because of the finding that it isoverexpressed in prostate cancer bone metastases but notin normal tissue or nonmetastatic cancer. Furthermore, ina mouse model, injection of PC3 cells did not lead to any

Figure 1. Genetic modifications of prostate cancer cells from primary tumors to bone tissue are shown. UPA indicates urokinase-type plasminogen activator; PTHrP, parathyroid hormone-related protein; IGFBF, insulin-like growth factor-binding protein; TGF-b,transforming growth factor-b; IGF, insulinlike growth factor; BMP, bone morphogenetic protein; ET-1, endothelin 1; PGDF, platelet-derived growth factor; FGF, fibroblast growth factor; VEGF, vascular endothelial growth factor; RANKL, receptor activator of thenuclear factor-jB ligand.

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1407

Page 3: Pathogenesis of osteoblastic bone metastases from prostate cancer

bone metastasis if cadherin 11 was silenced.28 Monocytechemotactic protein-1 (MCP-1) is chemotactic for pros-tate cancer cells, and the expression of its receptor, CCR2,correlates with pathologic stage.29-31 Lu et al reported thatthe activation of MCP-1/CCR2 axis promotes prostatecancer growth in bone.32 Once metastatic prostate cancercells arrive in the bone, they are stimulated by growth fac-tors present in the noncellular fraction of bone marrow,improving their interactions with the resident bone cells.

Metastasis suppressor genes encode a class of pro-teins that block the metastatic process without affectingprimary tumor progression. These genes are rarelymutated, but are instead thought to be controlled by epi-genetic events such as gene methylation. The first metasta-sis suppressor gene characterized, Nm23,33 whenoverexpressed in metastatic cell lines, has been shown todecrease metastatic competency in vivo, and motility andinvasion ability in vitro.33-36 There is mounting evidencethat many metastasis suppressor genes, including KAI1,CD44, and MAPK4, are able to block metastasis in pros-tate cancer cell lines, and several clinical studies have alsosupported their involvement.37-39 KAI1, the first prostatecancer metastasis suppressor gene identified, is down-regulated in both metastatic and high-grade prostate can-cer.37,40 Although the mechanism of suppression is stillunclear, it is believed that KAI1 interacts with severalmembrane proteins implicated in metastasis progression,such as E-cadherin, beta1 integrins, and epidermal growthfactor receptor. CD44 is a widely expressed cell adhesionprotein, and its down-regulation has been correlated toprostate cancer grade and metastatic stage.38,41,42 Similarresults have also been reported forMAPK4.39

Osteoblast Function and Their Regulation

The same regulatory pathways essential for bone develop-ment and remodeling are probably also involved in pros-tate cancer bone metastases. Osteoblast growth anddifferentiation are regulated by complex signaling path-ways mediated by growth factors such as bone morphoge-netic proteins (BMPs), insulinlike growth factor (IGF)-Iand IGF-II, transforming growth factor-b1 (TGFb1) andTGFb2, fibroblast growth factor (FGF), platelet-derivedgrowth factor (PDGF), and Wnt.43-48 Moreover, otherfactors, such as vascular endothelial growth factor(VEGF), are thought to have an indirect action on osteo-blast function through their modification of the bonemicroenvironment.

The transcription factor RUNX2 (also called corebinding factor 1) stimulates BMP2 and FGF, and is essen-

tial for osteoblast differentiation.49 In mice, inactivationof RUNX2 results in a skeleton made entirely of cartilage,indicating that RUNX2 is necessary for bone formation.49

Another transcription factor, Osterix, is believed to con-trol osteogenesis, based on the evidence thatOsx null micedo not undergo any bone development.50 Furthermore,b-catenin, a factor that mediates the Wnt signaling path-way, has also been implicated in osteogenesis regulation.A Wnt coreceptor, which regulates bone formationthrough b-catenin, interacts with Wnt by forming a com-plex with the Wnt receptor, Frizzled.51 This complex acti-vates the canonical Wnt signaling pathway, in turnstabilizing free cytoplasmic b-catenin, which is then trans-located to the nucleus. Here, it heterodimerizes with tran-scription factors of the lymphoid enhancer-bindingfactor/T-cell factor (LEF/TCF) family to regulateunknown genes that control bone formation.52,53

Interactions between prostate cancerand bone cells

Histopathologic analysis of prostate cancer bonemetastases typically shows a large number of osteoblastsadjacent to prostate cancer cells. In contrast, osteoblastsare almost always absent in normal bone or in bone metas-tases from other cancers (such as breast, lung, and kidney),which largely contain osteoclasts.51 The increase in pros-tate cancer bone-forming activity gives rise to a wovenbone, characterized by an osteosclerotic appearance dis-tinct from the typical lamellar structure seen in normalbone. These lesions are associated with an increase inbone mass at the lesion site, and often have an elevatedosteoid surface area, osteoid volume, and mineral apposi-tion rate.54-56

Supporting the observation that prostate cancerbone metastasis is associated with increased osteoblast ac-tivity, serum levels of osteoblast proliferation markers,such as bone-specific alkaline phosphatase and type 1 pro-tocollagen C-propeptide, have been observed to be higherin metastatic prostate cancer patients.57 The increasednumber of osteoblasts often present in the woven bonesurrounding the metastatic lesions suggests a correspond-ing increase in osteoblast proliferation and differentiation.This may contribute to prostate cancer cell survival andinvasiveness by providing abundant extracellular matri-ces.58 In vivo experiments by Gleave et al59 confirmedosteoblast involvement in prostate cancer bone metastasis,showing that factors secreted by bone fibroblasts, but notother cell types, accelerate prostate cancer growth. Invitro, Fizazi et al60 established the stimulation of prostate

Review Article

1408 Cancer March 15, 2010

Page 4: Pathogenesis of osteoblastic bone metastases from prostate cancer

cancer cells by the culture medium of osteoblast-like celllines. In these studies, primary mouse osteoblasts, cocul-tured with MDA prostate cancer 2b cells, led to cancercell proliferation, hinting at the possibility of paracrineinteractions between osteoblasts and the prostate cancercell line.60 In addition, osteoblast-conditioned mediumwas found to stimulate prostate cancer cells into produc-ing matrix metalloproteinase-9 (MMP-9) and urokinase-type plasminogen activator (uPA), while at the same timeincreasing the rate of prostate cancer proliferation.61 Fur-thermore, newly formed bone may secrete chemoattrac-tants that favor migration and enhance the invasioncapability of prostate cancer cells. Osteopontin, osteonec-tin, and bone sialoprotein can also modulate the proper-ties of prostate cancer cells.62 Osteonectin increases themigration and invasiveness of PC3 and DU145 cells,63

and bone sialoprotein facilitates the attachment of cancercells to the bone and enhances their metastatic potential.64

These observations indicate that bone-derived factors areable to initiate a series of cellular events that collectivelyhelp in cancer cell proliferation. Indirect evidence has alsoshown that this effect is specific to prostate cancer cells,because conditioned medium from osteoblasts does notenhance the growth of other cancer cell types.65 Clinicaltrials with specific bone-homing radiopharmaceuticalssuch as strontium-89, samarium, and rhenium, with dep-osition at sites of increased osteoblast activity and bone-matrix synthesis, have provided significant palliative bene-fits.66,67 Taken together, these observations indicate thatosteoblast inhibition has a direct effect on the pathogene-sis of osteoblastic lesions.

Receptor Activator of the Nuclear Factor-jB/RANK Ligand/Osteoprotegerin Axis

Osteoblasts also control osteoclast activity through theexpression of cytokines such as the receptor activator ofthe nuclear factor-jB (RANK) ligand (RANKL), a key ac-tivator of osteoclast differentiation, and osteoprotegerin, asoluble decoy receptor that inhibits RANKL.68,69 Anincrease in the level of serum osteoprotegerin (OPG) hasalso been established in advanced prostate cancerpatients.57,70 These findings show that osteoblasts, bymeans of their ability to control prostate cancer cell andosteoclast proliferation, operate as the ‘‘master switch’’ forprostate cancer progression in bone. Previous breast andprostate cancer studies in rodent models have proved thatRANKL inhibition decreases bone lesion developmentand tumor growth in bone.71-73 Furthermore, RANK is

expressed in prostate cancer cells and promotes invasionin a RANKL-dependent manner.74

Acquisition of a Bone-Like Phenotype

The acquisition of a bone cell-like phenotype allows pros-tate cancer cells to home, survive, and proliferate in bone.It has yet to be confirmed whether cancer cells alreadypossess osteomimetic phenotypes when they detach fromprimitive sites, or whether some of these characteristics areinstead acquired in the bone marrow. Prostate cancer cellsexpress several factors involved in normal bone develop-ment and remodeling (Fig. 1 and Table 1). These factorshave been implicated in osteoblastic lesions, either byaffecting osteoblast function directly, or by influencingbone formation indirectly through modification of thebone matrix or microenvironment.

BMPs

BMPs belong to the TGFb superfamily, and are the mostpowerful inductive bone factors enriching the bone ma-trix.75 Primary and metastatic prostate cancer have differ-ent phenotypic patterns of BMP expression, and adoptdiscrete downstream signaling pathways.76 BMPs demon-strate a clear and close relationship with the developmentand progression of primary prostate cancer, and also con-tribute to the onset and development of bone metastases.As a result, it has been postulated that BMPs play an im-portant role in the etiology of the osteoblastic bone metas-tasis phenotype. Indeed, BMP-secreting prostate cancercell cultures have been shown to promote in vitro

Table 1. Molecules Produced by Prostate Cancer CellsInvolved in Bone Metastases

Effects on Osteoblasts

Molecules Differentiation Proliferation Migration

ET1 � �

BMPs �

IGF � �

FGF � �

MDA-BF-1 � �

PDGF �

PSA � �

uPA � �

VEGF �

TGF-b �

Wnt �

PTHrP �

ET1 indicates endothelin 1; BMP, bone morphogenetic protein; IGF, insulin-

like growth factor; FGF, fibroblast growth factor; PDGF, platelet-derived

growth factor; PSA, prostate-specific antigen; uPA, urokinase-type plasmin-

ogen activator; VEGF, vascular endothelial growth factor; TGF-b, transform-

ing growth factor-b; PTHrP, parathyroid-hormone-related protein.

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1409

Page 5: Pathogenesis of osteoblastic bone metastases from prostate cancer

mineralization (potency from high to low: BMP6 >

BMP7 > BMP4).77 Furthermore, several BMPs areknown angiogenic factors that indirectly facilitate the de-velopment of bone metastases via the angiogenic route.On 1 hand, BMPs produced by prostate cancer cells areable to activate osteoblasts, leading to lesions that are pre-dominantly osteoblastic in nature. On the other hand,BMPs synthesized by osteoblasts, or released from thebone matrix, subsequently enhance the growth andaggressiveness of prostate cancer cells, which in turnincreases the production of BMPs by tumor cells.

Growth Factors

The IGF system, comprising 3 receptors, 3 ligands, and 6IGF-binding proteins (IGFBPs), is involved in the mito-genic, transformational, and antiapoptotic activities ofseveral cell types. In the prostate, this system plays an im-portant role in both normal and tumoral cells. IGF-I issecreted by the prostatic epithelium, probably in responseto growth hormones, and even in hypertrophy, high levelsof IGF-I have been reported.78 Prostate cancer cell lineshave been shown to express a range of different IGF com-ponents.79 IGF-I and II are abundant in the bone envi-ronment, and are known to stimulate osteoblasts,increasing bone matrix apposition, while at the same timedecreasing collagen degradation.80 Several studies haveshown a possible relationship between IGFs and bone me-tastasis from prostate cancer.81,82 IGF-I and II increasethe proliferative and chemotaxis activities of prostate can-cer cells in vitro.81 Moreover, the IGF-I pathway is up-regulated in prostate cancer cells metastasized to bone.82

Several in vivo studies have added further weight to thisimportant link. Serum IGF-I levels are correlated with therisk of developing prostate cancer,83 plasma IGFBP-III islower in patients with bone metastases, and IGFBP-II iselevated in prostate cancer patients.84 In addition, highIGF-I and low IGFBP-III levels are associated with therisk of developing advanced prostate cancer.85 Further-more, polymorphisms of IGF-I and cytochrome P450enzyme 19 have been reported to accurately predict bonemetastasis in prostate cancer patients.86 However, Rubinet al87 recently showed that IGF-I is neither necessary norsufficient for the osteoblastic response to prostate cancermetastases. For this reason, it is still not clear how the IGFsystem participates in the formation of osteoblastic pros-tate cancer bone metastasis, and additional studies arerequired to delineate the role of the IGF system in thisprocess.

Parathyroid Hormone-Related Proteinand Endothelin 1

The osteolytic factor parathyroid hormone (PTH)-relatedprotein (PTHrP) is a homolog of PTH that has a directaction on PTH receptors, increasing bone resorption andrenal tubular calcium reabsorption.88 In bone metastases,the release of PTHrP by cancer cells, together with otherfactors, contributes significantly to metastatic spread.89,90

PTHrP is abundant in prostate cancer bone metastases,and in these tumors osteoblastic lesions tend to predomi-nate.91 One possible explanation for this paradox is thatprostate cancer-derived PTHrP mediates the interactionsbetween the bone marrow microenvironment and pros-tate cancer, which further facilitates the establishment ofskeletal metastases and osteoblastic alterations. Liao et al92

have provided evidence supporting this opinion, report-ing that PTHrP increases osteoblastogenesis-stimulatingosteoblast progenitor cell proliferation and induces earlyosteoblast differentiation.

Another theory is that NH2-terminal fragments ofPTHrP share a strong sequence homology with endothe-lin (ET) 1, thus stimulating new bone formation throughactivation of the ETA receptor.93 As an osteoblast mito-genic factor, ET1 promotes osteoblast growth at meta-static sites.94 Nelson et al95 showed that plasma ET1concentrations are significantly higher in metastatic pros-tate cancer patients, suggesting that ET1 may be secretedby prostate cancer cells. A recent study has also suggestedthat ET1 increases osteoblast proliferation and new boneformation through activation of the Wnt signaling path-way and suppression of the Wnt pathway inhibitor,DKK1.96

uPA

Prostate cancer cells produce other factors, including uPAand prostate-specific antigen (PSA) that, although notdirectly involved in normal skeletal development, affectosteoblast function by modifying the bone microenviron-ment.97 The serine protease uPA is involved in degrada-tion of the extracellular matrix, which facilitates tumorcell invasion by increasing osteoblastic prostate cancerbone metastases. The pathogenetic model of osteoblasticprostate cancer metastases can be summarized as follows.The neoplastic cell produces high molecular weight uPA(HMW-uPA) that binds uPA receptor (uPAR) on the tu-mor cell surface and exerts its proteolytic action, leadingto tumoral invasion. HMW-uPA is then split into lowmolecular weight uPA and amino terminal fragment,which binds uPAR on the osteoblast cell surface, helping

Review Article

1410 Cancer March 15, 2010

Page 6: Pathogenesis of osteoblastic bone metastases from prostate cancer

to trigger the cascade of events leading to osteoblast prolif-eration and activation. Furthermore, uPA can cleave andactive TGFb, which is produced in a latent form by osteo-blasts. TGFb regulates osteoblast and osteoclast differen-tiation as well as the growth of tumor cells themselves. Inaddition, uPA stimulates osteoblast proliferation, prob-ably by hydrolyzing IGFBPs, thereby increasing free IGFlevels.98-100 PSA belongs to the kallikrein serine proteasefamily, is secreted by prostate cancer cells, and is usedroutinely as a marker of prostate cancer progression. PSAnot only cleaves PTHrP to release osteoblastic PTHrPfragments, but also activates osteoblast growth factorssuch as TGFb.100 Consequently, PTHrP degradation byPSA decreases bone resorption,101 allowing the osteo-blast response to predominate. As with uPA, PSA canalso cleave IGFBP3, thereby freeing up IGF-I to bind toits receptor and stimulate osteoblast proliferation.102

Other Molecules

The growth factor MDA-BF-1 has recently been identi-fied in the supernatant of bone marrow specimens frompatients with prostate cancer bone metastases.103 Thissecreted form of the ErbB3 growth factor receptor isexpressed in prostate cancer cells that metastasize to bone,but not in the primary tumors of patients with localizeddisease, nor in prostate cancer cells that metastasize toother sites. Its function is mediated by an osteoblast-expressed receptor,104 and through specific interactionsbetween prostate cancer cells and bone. Scientific litera-ture is rich in papers describing new proteins that seemto be involved in prostate cancer bone metastases. One ofthese is the tyrosine kinase receptor c-Kit, and its ligandstem cell factor (SCF).105 In an experimental model,prostate cancer bone metastasis was found to stronglyexpress c-Kit, but prostate cancer cell lines were c-Kitnegative. In addition, immunohistochemical analysisshowed a higher expression of this protein in bone metas-tasis compared with primary tumors, and SCF was oftenoverexpressed.105

Prostate cancer cells produce a range of additionalosteoblast-regulatory growth factors, including PDGF,FGF, and VEGF. The dimeric polypeptide growth factorPDGF has 2 subunits, A and B, that form AA, BB, andAB isoforms. The BB isoform is known to be a potentosteotropic factor, contributing to the formation of osteo-blastic lesions through the promotion of osteoblast migra-tion and proliferation.106,107 Although FGF1 (acidic) andFGF2 (basic) are both able to increase osteoblast prolifera-tion, only FGF2 suppresses osteoclast formation.108 It is

clear that further investigations are necessary to fullyunderstand the complex FGF interplay in bone metasta-sis. VEGF appears to have both a direct and an indirecteffect on bone growth by activating osteoblasts and pro-moting angiogenesis, respectively.77,109 The Wnt familyligand has been reported to be up-regulated in the tumorcells of advanced metastatic prostate cancer patients.110

Furthermore, Wnt production can act in a paracrine wayto induce osteoblastic bone metastases. As mentioned pre-viously, the Wnt signaling pathway can be blocked by theWnt antagonist, DKK1. This molecule is usuallyexpressed early in the development of skeletal metastases,resulting in the masking of osteogenic Wnts, which favorsmetastatic site osteolysis. As metastasis progresses, adecrease in DKK1 expression unmasks Wnt-mediatedosteoblastic activity, leading to osteosclerosis.111,112

The Role of Osteoclasts

Although prostate cancer bone metastasis is osteoblastic innature, as revealed by radiographic imaging, the presenceof osteolytic-osteogenic bone lesions in osteoblastic casesmay account for the increase in observed fractures.113 Keysteps in the successful establishment of prostate cancerbone metastases appear to be osteoclast formation andbone resorption, followed by the release of several growthfactors from the bone matrix.114 However, the mecha-nisms by which prostate cancer cells promote this processremain unclear. It has been suggested that RANKL, inter-leukin (IL) 6, IL-8, and C-C chemokine ligand 2 mediateosteoclast formation from human mononuclear bonemarrow cells.31,32,99 Furthermore, prostate cancer cellshave been shown to secrete factors that promote humanbone marrow mononuclear cell osteoclastogenesis.31

Other authors have observed that IL-6 affects fusion butnot resorption, and that osteoclastogenesis in prostatecancer bone metastasis is induced by a RANKL-independ-ent mechanism. A possible hypothesis put forward is thatprostate cancer cells, by producing PTHrP, can facilitateboth osteoclastogenesis and osteoblastogenesis.115

Another supposition is focused on C-C chemokine ligand2, which may serve 2 distinct roles in the developmentand promotion of prostate cancer: a direct effect on epi-thelial prostate cancer cells, and an indirect effect onosteoclasts and endothelial cells at the metastatic site,thereby supporting tumor growth. The presence of C-Cchemokine ligand 2 in the bone microenvironment maycontribute to prostate cancer growth by initiating osteo-clastogenesis and angiogenesis, creating a favorable nichefor metastases.116

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1411

Page 7: Pathogenesis of osteoblastic bone metastases from prostate cancer

The Vicious Cycle of OsteoblasticBone Metastasis

The complex interactions between tumor cells, bone cells,and the bone matrix constitute a vicious cycle of osteo-blast-mediated bone metastasis (Fig. 2). In the earlystages, prostate cancer cells produce osteogenic factorssuch as PDGF, ET1 and BMPs, which activate osteo-blasts. Once differentiated from their progenitor cells,osteoblasts deposit a new matrix for bone formation.However, this unmineralized matrix provides a more fer-tile soil for tumor cells, enriched with growth factors andnoncollagen proteins. Moreover, newly formed bone mayprovide additional factors to attract prostate cancer cells,allowing them to survive and proliferate in the bone envi-ronment, which in turn further activates osteoblasts.However, the osteoblastic nature of bone metastases mayrepresent a double-edged sword regarding the develop-ment of lesions; the initial increase in bone volume may

limit the space available to cancer cells, and therefore helpto confine the tumor. This may explain why osteoblasticprostate cancer metastases seem to progress more slowlythan osteolytic metastases from other tumors. In addition,especially at the beginning of the process, tumor-derivedfactors and RANKL-secreting osteoblasts can both acti-vate osteoclasts, leading to some level of bone resorption,which subsequently creates more space for dominantosteoblastic lesions. Cytokines released from the bone ma-trix during bone resorption can enhance this vicious cycleby facilitating the continued proliferation of prostate can-cer cells and osteoblasts. Regrettably, as soon as the meta-static disease enters the osteoclastic stage, diseaseprogression is rapid, and survival times are short.54,117,118

Clinical Applications

A deeper understanding of the molecular mechanisms ofprostate cancer bone metastases will hopefully identify

Figure 2. The vicious cycle of osteoblastic bone metastasis is shown. (A) Prostate cancer cells secrete osteogenic growth factors,activating osteoblasts to deposit new bone matrix. (B) Osteoblasts secrete a range of additional factors such as insulin-likegrowth factor (IGF), fibroblast growth factor (FGF), and transforming growth factor-b (TGFb). (C) These factors attract prostatecancer cells, further enhancing their proliferation and growth. ET1 indicates endothelin 1; PDGF, platelet-derived growth factor;BMP, bone morphogenetic protein; RANK, receptor activator of the nuclear factor-jB; RANKL, RANK ligand; OPG,osteoprotegerin.

Review Article

1412 Cancer March 15, 2010

Page 8: Pathogenesis of osteoblastic bone metastases from prostate cancer

new biological targets for innovative drugs, besides zole-dronic acid, which has become the standard treatment forthis disease, to be used in combination with conventionaltherapies, as well as new predictive or prognostic markers.One of the most extensively studied pathways in recentyears, the RANK/RANKL/OPG axis, has been advocatedas a potential therapeutic target because it governs bonehomeostasis, both under normal physiologic conditions,and during bone metastasis progression. In preclinical andclinical trials, inhibition of RANKL with OPG reducesbone turnover markers,119 suggesting that RANKL, inaddition to having an effect on bone lysis, also has a directaction on tumor cells.120,121 The anti-RANKL monoclo-nal antibody, denosumab, has recently been tested againstzoledronic acid in a randomized prostate cancer bone me-tastases phase 2 study,122 and was found to decrease uri-nary bone turnover markers in zoledronic acid resistantpatients. Phase 3 bone metastases studies are ongoing withskeletal-related events (SREs) as the primary objective,and results should be available shortly.

Atrasentan, an ETA receptor antagonist, is beingtested in clinical trials for the treatment of prostate cancer,and it has been shown to prevent osteoblastic bone metas-tases in mouse models, as well as reduce disease progres-sion in advanced phase 2 prostate cancer trials.123,124

However, a recent phase 3 study of atrasentan versus pla-cebo reported no significant differences in disease progres-sion,125 and docetaxel/atrasentan prostate cancer studiesare still ongoing. The monoclonal antibody vitaxin bindsintegrin avb3, and androgen-independent prostate cancerclinical studies are currently in progress.126 This moleculehas demonstrated both in vivo and in vitro antitumoractivities, affecting bone resorption by impairing osteo-clast attachment, whereas osteoclast formation and multi-nucleation processes remain unaffected.

Other targets of bone metastases include Akt, cyclo-oxygenase-2, and MMP-9. Diaz et al127 recently investi-gated the Akt inhibitor Palomid 529 in vitro, and foundthat it had a synergistic effect with radiotherapy, enhanc-ing the antiproliferative effect of radiation in prostate can-cer cells, while decreasing p-Akt, VEGF, MMP-9, MMP-2, and Id-1 levels. Another drug target of MMP-9, 3,30-diindolylmethane,128,129 is an inhibitor of the DNAbinding activity of nuclear factor-jB, which mediatesthe expression of many genes such as VEGF, IL8, anduPA. Antibodies against IGF-I receptor, and IGF-I re-ceptor-specific tyrosine kinase inhibitors, have beendeveloped as anticancer agents, and some have alreadybeen tested in clinical trials against solid tumors, show-

ing promising results.130,131 These are just a few of thevast array of new drugs currently under preclinical andclinical experimentation.

Regarding the monitoring of metastatic prostatecancer, the most commonly cited markers seem to be theproducts of collagen degradation.132-135 This categoryincludes N-telopeptide (NTx) or C-telopeptide type Icollagen (CTX), amino-terminal procollagen propep-tides (PINP), and cross-linked C-terminal telopeptides(ICTP). Other common markers of osteoblastic bone me-tastases include total and bone-specific alkaline phospha-tase, serum tartrate-resistant acid phosphatase, andPSA.132,133 In a recent study of 100 patients, Klepziget al134 observed that procollagen propeptides couldreliably predict prostate cancer bone metastases. Anotherimportant study by Lein et al133 has focused further atten-tion toward procollagen propeptides, NTx, and PSA,establishing bone markers as useful tools for the predic-tion and diagnosis of SREs in patients with prostate can-cer bone metastases undergoing zoledronic acid therapy.NTx has recently undergone validation in 3 large retro-spective phase 3 trials.135 This marker was found todecline to normal physiologic concentrations within 3months, and, compared with high NTx levels, was associ-ated with a reduced risk of skeletal complications anddeath. In contrast, PSA has been reported to be a poormarker, both for identifying patient populations at highrisk of metastatic disease and for monitoring skeletal pro-gression during treatment.132 A further study by Jung etal57 tested 10 serum markers in 117 prostate cancerpatients, citing OPG and bone sialoprotein as excellentbone metastases markers in addition to being independentprognostic factors for prostate cancer-related death.

Conclusions

Cancer cells can only metastasize in organs where themicroenvironment accommodates their growth. In otherwords, the complex interactions between cancer cells andthe bone/bone marrow microenvironment are fundamen-tal to the establishment of bone metastases. Prostate can-cer cells from primary tissue undergo epithelial-mesenchymal transition to disseminate and acquire abone-like phenotype to metastasize in bone tissue. Afterthe arrival of prostate cancer cells in bone, their crosstalkwith the bone microenvironment facilitates bone metasta-ses, and defines their osteoblastic pattern. Finally, recentdiscoveries on the mechanisms of bone metastasization,for example epithelial-mesenchymal transition and osteo-mimicry, will certainly help to improve bone target

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1413

Page 9: Pathogenesis of osteoblastic bone metastases from prostate cancer

therapies. Without doubt, there is an urgent need for fur-ther work, particularly on the potential role of the preme-tastatic bone niche, and on the involvement of cancerstem cells in bone metastases.

CONFLICT OF INTEREST DISCLOSURESThe authors made no disclosures.

REFERENCES1. Keller ET, Zhang J, Cooper CR, et al. Prostate carcinoma

skeletal metastases: cross-talk between tumor and bone.Cancer Metastasis Rev. 2001;20:333-349.

2. Bubendorf L, Schopfer A, Wagner U, et al. Metastatic pat-terns of prostate cancer: an autopsy study of 1,589patients. Hum Pathol. 2000;31:578-583.

3. Roodman GD. Mechanisms of bone metastasis. N Engl JMed. 2004;350:1655-1664.

4. Mundy GR. Metastasis to bone: causes, consequences andtherapeutic opportunities. Nat Rev Cancer. 2002;2:584-593.

5. Roudier MP, Vesselle H, True LD, et al. Bone histologyat autopsy and matched bone scintigraphy findings inpatients with hormone refractory prostate cancer: the effectof bisphosphonate therapy on bone scintigraphy results.Clin Exp Metastasis. 2003;20:171-180.

6. Thiery JP. Epithelial-mesenchymal transitions in tumourprogression. Nat Rev Cancer. 2002;2:442-454.

7. Stoker M, Perryman M. An epithelial scatter factor releasedby embryo fibroblasts. J Cell Sci. 1985;77:209-223.

8. Edme N, Downward J, Thiery JP, Boyer B. Ras inducesNBT-II epithelial cell scattering through the coordinateactivities of Rac and MAPK pathways. J Cell Sci.2002;115:2591-2601.

9. Janda E, Lehmann K, Killisch I, et al. Ras and TGF[beta]cooperatively regulate epithelial cell plasticity and metasta-sis: dissection of Ras signaling pathways. J Cell Biol.2002;156:299-313.

10. Nicolas FJ, Lehmann K, Warne PH, Hill CS, DownwardJ. Epithelial to mesenchymal transition in Madin-Darbycanine kidney cells is accompanied by down-regulation ofSmad3 expression, leading to resistance to transforminggrowth factor-beta-induced growth arrest. J Biol Chem.2003;278:3251-3256.

11. Psichari E, Balmain A, Plows D, Zoumpourlis V, PintzasA. High activity of serum response factor in the mesenchy-mal transition of epithelial tumor cells is regulated byRhoA signaling. J Biol Chem. 2002;277:29490-29495.

12. Davis TL, Cress AE, Dalkin BL, Nagle RB. Unique expres-sion pattern of the alpha6beta4 integrin and laminin-5 inhuman prostate carcinoma. Prostate. 2001;46: 240-248.

13. Hao J, Jackson L, Calaluce R, McDaniel K, Dalkin BL,Nagle RB. Investigation into the mechanism of the loss oflaminin 5 (alpha3beta3gamma2) expression in prostatecancer. Am J Pathol. 2001;158:1129-1135.

14. Udayakumar TS, Chen ML, Bair EL, et al. Membranetype-1-matrix metalloproteinase expressed by prostate car-cinoma cells cleaves human laminin-5 beta3 chain andinduces cell migration. Cancer Res. 2003;63:2292-2299.

15. Cooper CR, McLean L, Mucci NR, Poncza P, Pienta KJ.Prostate cancer cell adhesion to quiescent endothelial cellsis not mediated by beta-1 integrin subunit. Anticancer Res.2000;20:4159-4162.

16. Chay CH, Cooper CR, Gendernalik JD, et al. A func-tional thrombin receptor (PAR1) is expressed on bone-derived prostate cancer cell lines. Urology. 2002;60:760-765.

17. Honn KV, Tang DG. Adhesion molecules and tumor cellinteraction with endothelium and subendothelial matrix.Cancer Metastasis Rev. 1992;11:353-375.

18. Elgavish A, Prince C, Chang PL, Lloyd K, Lindsey R,Reed R. Osteopontin stimulates a subpopulation of quies-cent human prostate epithelial cells with high proliferativepotential to divide in vitro. Prostate. 1998;35:83-94.

19. Morrissey C, Vessella RL. The role of tumor microenvir-onment in prostate cancer bone metastasis. J Cell Biochem.2007;101:873-886.

20. Ellis WJ, Pfitzenmaier J, Colli J, Arfman E, Lange PH,Vessella RL. Detection and isolation of prostate cancercells from peripheral blood and bone marrow. Urology.2003;61:277-281.

21. Klein CA, Blankenstein TJ, Schmidt-Kittler O, et al.Genetic heterogeneity of single disseminated tumourcells in minimal residual cancer. Lancet. 2002;360:683-689.

22. Kraus J, Pantel K, Pinkel D, Albertson DG, Speicher MR.High-resolution genomic profiling of occult micrometa-static tumor cells. Genes Chromosomes Cancer. 2003;36: 159-166.

23. Melchior SW, Corey E, Ellis WJ, et al. Early tumor celldissemination in patients with clinically localized carci-noma of the prostate. Clin Cancer Res. 1997;3:249-256.

24. Sikes RA, Nicholson BE, Koeneman KS, et al. Cellularinteractions in the tropism of prostate cancer to bone. IntJ Cancer. 2004;110:497-503.

25. Cooper CR, Chay CH, Pienta KJ. The role of alpha(v)-beta(3) in prostate cancer progression. Neoplasia. 2002;4:191-194.

26. Hall CL, Dai J, van Golen KL, Keller ET, Long MW.Type I collagen receptor (alpha 2 beta 1) signaling pro-motes the growth of human prostate cancer cells withinthe bone. Cancer Res. 2006;66:8648-8654.

27. Sun YX, Wang J, Shelburne CE, et al. Expression ofCXCR4 and CXCL12 (SDF-1) in human prostate cancers(PCa) in vivo. J Cell Biochem. 2003;89:462-473.

28. Chu K, Cheng CJ, Ye X, et al. Cadherin-11 promotes themetastasis of prostate cancer cells to bone. Mol Cancer Res.2008;6:1259-1267.

29. Lu Y, Cai Z, Galson DL, et al. Monocyte chemotacticprotein-1 (MCP-1) acts as a paracrine and autocrine factorfor prostate cancer growth and invasion. Prostate. 2006;66:1311-1318.

30. Loberg RD, Ying C, Craig M, et al. Targeting CCL2 withsystemic delivery of neutralizing antibodies induces pros-tate cancer tumor regression in vivo. Cancer Res. 2007;67:9417-9424.

31. Lu Y, Cai Z, Xiao G, et al. CCR2 expression correlateswith prostate cancer progression. J Cell Biochem. 2007;101:676-685.

32. Lu Y, Chen Q, Corey E, et al. Activation of MCP-1/CCR2 axis promotes prostate cancer growth in bone. ClinExp Metastasis. 2009;26:161-169.

Review Article

1414 Cancer March 15, 2010

Page 10: Pathogenesis of osteoblastic bone metastases from prostate cancer

33. Leone A, Flatow U, King CR, et al. Reduced tumor inci-dence, metastatic potential, and cytokine responsivenessof nm23-transfected melanoma cells. Cell. 1991;65:25-35.

34. Kantor JD, McCormick B, Steeg PS, Zetter BR. Inhibi-tion of cell motility after nm23 transfection of human andmurine tumor cells. Cancer Res. 1993;53:1971-1973.

35. Leone A, Flatow U, VanHoutte K, Steeg PS. Transfectionof human nm23-H1 into the human MDA-MB-435breast carcinoma cell line: effects on tumor metastaticpotential, colonization and enzymatic activity. Oncogene.1993;8:2325-2333.

36. Hsu S, Huang F, Wang L, Banerjee S, Winawer S, Fried-man E. The role of nm23 in transforming growth factorbeta 1-mediated adherence and growth arrest. Cell GrowthDiffer. 1994;5:909-917.

37. Dong JT, Lamb PW, Rinker-Schaeffer CW, et al. KAI1, ametastasis suppressor gene for prostate cancer on humanchromosome 11p11.2. Science. 1995;268:884-886.

38. Gao AC, Lou W, Dong JT, Isaacs JT. CD44 is a metasta-sis suppressor gene for prostatic cancer located on humanchromosome 11p13. Cancer Res. 1997;57:846-849.

39. Kim HL, Vander Griend DJ, Yang X, et al. Mitogen-acti-vated protein kinase kinase 4 metastasis suppressor geneexpression is inversely related to histological pattern inadvancing human prostatic cancers. Cancer Res. 2001;61:2833-2837.

40. Ueda T, Ichikawa T, Tamaru J, et al. Expression of theKAI1 protein in benign prostatic hyperplasia and prostatecancer. Am J Pathol. 1996;149:1435-1440.

41. Noordzij MA, van Steenbrugge GJ, Schroder FH, Van derKwast TH. Decreased expression of CD44 in metastaticprostate cancer. Int J Cancer. 1999;84:478-483.

42. Nagabhushan M, Pretlow TG, Guo YJ, Amini SB, PretlowTP, Sy MS. Altered expression of CD44 in human pros-tate cancer during progression. Am J Clin Pathol. 1996;106:647-651.

43. Canalis E. Effect of insulinlike growth factor I on DNAand protein synthesis in cultured rat calvaria. J Clin Invest.1980;66:709-719.

44. Hauschka PV, Mavrakos AE, Iafrati MD, Doleman SE,Klagsbrun M. Growth factors in bone matrix. Isolation ofmultiple types by affinity chromatography on heparin-Sepharose. J Biol Chem. 1986;261:12665-12674.

45. Rodan SB, Wesolowski G, Thomas K, Rodan GA.Growth stimulation of rat calvaria osteoblastic cells byacidic fibroblast growth factor. Endocrinology. 1987;121:1917-1923.

46. Globus RK, Patterson-Buckendahl P, Gospodarowicz D.Regulation of bovine bone cell proliferation by fibroblastgrowth factor and transforming growth factor beta. Endo-crinology. 1988;123:98-105.

47. Wang EA, Rosen V, D’Alessandro JS, et al. Recombinanthuman bone morphogenetic protein induces bone forma-tion. Proc Natl Acad Sci U S A. 1990;87:2220-2224.

48. Spencer EM, Liu CC, Si EC, Howard GA. In vivo actionsof insulin-like growth factor-I (IGF-I) on bone formationand resorption in rats. Bone. 1991;12:21-26.

49. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G.Osf2/Cbfa1: a transcriptional activator of osteoblast differ-entiation. Cell. 1997;89:747-754.

50. Nakashima K, Zhou X, Kunkel G, et al. The novel zincfinger-containing transcription factor osterix is required for

osteoblast differentiation and bone formation. Cell. 2002;108:17-29.

51. Logothetis CJ, Lin SH. Osteoblasts in prostate cancer me-tastasis to bone. Nat Rev Cancer. 2005;5:21-28.

52. Behrens J, von Kries JP, Kuhl M, et al. Functional interac-tion of beta-catenin with the transcription factor LEF-1.Nature. 1996;382:638-642.

53. van de Wetering M, Cavallo R, Dooijes D, et al. Arma-dillo coactivates transcription driven by the product of theDrosophila segment polarity gene dTCF. Cell. 1997;88:789-799.

54. Cook GB, Watson FR. Events in the natural history ofprostate cancer: using salvage curves, mean age distribu-tions and contingency coefficients. J Urol. 1968;99:87-96.

55. Charhon SA, Chapuy MC, Delvin EE, Valentin-Opran A,Edouard CM, Meunier PJ. Histomorphometric analysis ofsclerotic bone metastases from prostatic carcinoma specialreference to osteomalacia. Cancer. 1983;51:918-924.

56. Clarke NW, McClure J, George NJ. Osteoblast functionand osteomalacia in metastatic prostate cancer. Eur Urol.1993;24:286-290.

57. Jung K, Lein M, Stephan C, et al. Comparison of 10 se-rum bone turnover markers in prostate carcinoma patientswith bone metastatic spread: diagnostic and prognosticimplications. Int J Cancer. 2004;111:783-791.

58. Jacobs SC. Spread of prostatic cancer to bone. Urology.1983;21:337-344.

59. Gleave M, Hsieh JT, Gao CA, von Eschenbach AC,Chung LW. Acceleration of human prostate cancer growthin vivo by factors produced by prostate and bone fibro-blasts. Cancer Res. 1991;51:3753-3761.

60. Fizazi K, Yang J, Peleg S, et al. Prostate cancer cells-osteo-blast interaction shifts expression of growth/survival-relatedgenes in prostate cancer and reduces expression of osteo-protegerin in osteoblasts. Clin Cancer Res. 2003;9:2587-2597.

61. Festuccia C, Giunciuglio D, Guerra F, et al. Osteoblastsmodulate secretion of urokinase-type plasminogen activator(uPA) and matrix metalloproteinase-9 (MMP-9) in humanprostate cancer cells promoting migration and matrigelinvasion. Oncol Res. 1999;11:17-31.

62. Rosol TJ. Pathogenesis of bone metastases: role of tumor-related proteins. J Bone Miner Res. 2000;15:844-850.

63. Jacob K, Webber M, Benayahu D, Kleinman HK. Osteo-nectin promotes prostate cancer cell migration and inva-sion: a possible mechanism for metastasis to bone. CancerRes. 1999;59:4453-4457.

64. Zhang JH, Tang J, Wang J, et al. Over-expression of bonesialoprotein enhances bone metastasis of human breastcancer cells in a mouse model. Int J Oncol. 2003;23:1043-1048.

65. Lang SH, Miller WR, Habib FK. Stimulation of humanprostate cancer cell lines by factors present in humanosteoblast-like cells but not in bone marrow. Prostate.1995;27:287-293.

66. Quilty PM, Kirk D, Bolger JJ, et al. A comparison of thepalliative effects of strontium-89 and external beam radio-therapy in metastatic prostate cancer. Radiother Oncol.1994;31:33-40.

67. Han SH, de Klerk JM, Tan S, et al. The PLACORHENstudy: a double-blind, placebo-controlled, randomized ra-dionuclide study with (186) Re-etidronate in hormone-re-sistant prostate cancer patients with painful bone

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1415

Page 11: Pathogenesis of osteoblastic bone metastases from prostate cancer

metastases. Placebo Controlled Rhenium Study. J NuclMed. 2002;43:1150-1156.

68. Udagawa N, Takahashi N, Yasuda H, et al. Osteoprote-gerin produced by osteoblasts is an important regulator inosteoclast development and function. Endocrinology. 2000;141:3478-3484.

69. Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentia-tion and activation. Nature. 2003;423:337-342.

70. Brown JM, Vessella RL, Kostenuik PJ, Dunstan CR,Lange PH, Corey E. Serum osteoprotegerin levels areincreased in patients with advanced prostate cancer. ClinCancer Res. 2001;7:2977-2983.

71. Morony S, Capparelli C, Kostenuik PJ, et al. Osteoprote-gerin prevents bone destruction in athymic and syngeneicmodels of experimental tumor metastasis to bone. Cancer.2000;88:3107.

72. Morony S, Capparelli C, Sarosi I, Lacey DL, DunstanCR, Kostenuik PJ. Osteoprotegerin inhibits osteolysis anddecreases skeletal tumor burden in syngeneic and nudemouse models of experimental bone metastasis. Cancer Res.2001;61:4432-4436.

73. Zhang J, Dai J, Qi Y, et al. Osteoprotegerin inhibits pros-tate cancer-induced osteoclastogenesis and prevents pros-tate tumor growth in the bone. J Clin Invest. 2001;107:1235-1244.

74. Armstrong AP, Miller RE, Jones JC, Zhang J, Keller ET,Dougall WC. RANKL acts directly on RANK-expressingprostate tumor cells and mediates migration and expressionof tumor metastasis genes. Prostate. 2008;68:92-104.

75. Urist MR. Bone: formation by autoinduction. Science.1965;150:893-899.

76. Ide H, Yoshida T, Matsumoto N, et al. Growth regulationof human prostate cancer cells by bone morphogeneticprotein-2. Cancer Res. 1997;57:5022-5027.

77. Dai J, Kitagawa Y, Zhang J, et al. Vascular endothelialgrowth factor contributes to the prostate cancer-inducedosteoblast differentiation mediated by bone morphogeneticprotein. Cancer Res. 2004;64:994-999.

78. Ruan W, Powell-Braxton L, Kopchick JJ, Kleinberg DL.Evidence that insulin-like growth factor I and growth hor-mone are required for prostate gland development. Endo-crinology. 1999;140:1984-1989.

79. Cohen P, Peehl DM, Lamson G, Rosenfeld RG. Insulin-like growth factors (IGFs), IGF receptors, and IGF-bind-ing proteins in primary cultures of prostate epithelial cells.J Clin Endocrinol Metab. 1991;73:401-407.

80. Koch H, Jadlowiec JA, Campbell PG. Insulin-like growthfactor-I induces early osteoblast gene expression in humanmesenchymal stem cells. Stem Cells Dev. 2005;14: 621-631.

81. Ritchie CK, Andrews LR, Thomas KG, Tindall DJ, Fitz-patrick LA. The effects of growth factors associated withosteoblasts on prostate carcinoma proliferation and chemo-taxis: implications for the development of metastatic dis-ease. Endocrinology. 1997;138:1145-1150.

82. Rubin J, Chung LW, Fan X, et al. Prostate carcinoma cellsthat have resided in bone have an upregulated IGF-I axis.Prostate. 2004;58:41-49.

83. Chan JM, Stampfer MJ, Giovannucci E, et al. Plasma in-sulin-like growth factor-I and prostate cancer risk: a pro-spective study. Science. 1998;279:563-566.

84. Shariat SF, Lamb DJ, Kattan MW, et al. Association ofpreoperative plasma levels of insulin-like growth factor I

and insulin-like growth factor binding proteins-2 and �3with prostate cancer invasion, progression, and metastasis.J Clin Oncol. 2002;20:833-841.

85. Chan JM, Stampfer MJ, Ma J, et al. Insulin-like growthfactor-I (IGF-I) and IGF binding protein-3 as predictorsof advanced-stage prostate cancer. J Natl Cancer Inst.2002;94:1099-1106.

86. Tsuchiya N, Wang L, Suzuki H, et al. Impact of IGF-Iand CYP19 gene polymorphisms on the survival ofpatients with metastatic prostate cancer. J Clin Oncol.2006;24:1982-1989.

87. Rubin J, Fan X, Rahnert J, et al. IGF-I secretion byprostate carcinoma cells does not alter tumor-bone cellinteractions in vitro or in vivo. Prostate. 2006;66:789-800.

88. Yates AJ, Gutierrez GE, Smolens P, et al. Effects of a syn-thetic peptide of a parathyroid hormone-related protein oncalcium homeostasis, renal tubular calcium reabsorption,and bone metabolism in vivo and in vitro in rodents.J Clin Invest. 1988;81:932-938.

89. Guise TA, Yin JJ, Taylor SD, et al. Evidence for a causalrole of parathyroid hormone-related protein in the patho-genesis of human breast cancer-mediated osteolysis. J ClinInvest. 1996;98:1544-1549.

90. Shen X, Falzon M. PTH-related protein modulates PC-3prostate cancer cell adhesion and integrin subunit profile.Mol Cell Endocrinol. 2003;199:165-177.

91. Liao J, McCauley LK. Skeletal metastasis: established andemerging roles of parathyroid hormone related protein(PTHrP). Cancer Metastasis Rev. 2006;25:559-571.

92. Liao J, Li X, Koh AJ, et al. Tumor expressed PTHrP facil-itates prostate cancer-induced osteoblastic lesions. Int JCancer. 2008;123:2267-2278.

93. Schluter KD, Katzer C, Piper HM. A N-terminal PTHrPpeptide fragment void of a PTH/PTHrP-receptor bindingdomain activates cardiac ET(A) receptors. Br J Pharmacol.2001;132:427-432.

94. Takuwa Y, Masaki T, Yamashita K. The effects of the endo-thelin family peptides on cultured osteoblastic cells from ratcalvariae. Biochem Biophys Res Commun. 1990;170: 998-1005.

95. Nelson JB, Nguyen SH, Wu-Wong JR, et al. New boneformation in an osteoblastic tumor model is increased byendothelin-1 overexpression and decreased by endothelin Areceptor blockade. Urology. 1999;53:1063-1069.

96. Clines GA, Mohammad KS, Bao Y, et al. Dickkopf hom-olog 1 mediates endothelin-1-stimulated new bone forma-tion. Mol Endocrinol. 2007;21:486-498.

97. Achbarou A, Kaiser S, Tremblay G, et al. Urokinase over-production results in increased skeletal metastasis by pros-tate cancer cells in vivo. Cancer Res. 1994;54:2372-2377.

98. Rabbani SA, Desjardins J, Bell AW, et al. An amino-ter-minal fragment of urokinase isolated from a prostate can-cer cell line (PC-3) is mitogenic for osteoblast-like cells.Biochem Biophys Res Commun. 1990;173:1058-1064.

99. Sommerfeldt DW, Rubin CT. Biology of bone and how itorchestrates the form and function of the skeleton. EurSpine J. 2001;10(suppl 2):S86-S95.

100. Killian CS, Corral DA, Kawinski E, Constantine RI. Mi-togenic response of osteoblast cells to prostate-specific anti-gen suggests an activation of latent TGF-beta and aproteolytic modulation of cell adhesion receptors. BiochemBiophys Res Commun. 1993;192:940-947.

Review Article

1416 Cancer March 15, 2010

Page 12: Pathogenesis of osteoblastic bone metastases from prostate cancer

101. Iwamura M, Hellman J, Cockett AT, Lilja H, GershagenS. Alteration of the hormonal bioactivity of parathyroidhormone-related protein (PTHrP) as a result of limitedproteolysis by prostate-specific antigen. Urology. 1996;48:317-325.

102. Cohen P, Peehl DM, Graves HC, Rosenfeld RG. Biologi-cal effects of prostate specific antigen as an insulin-likegrowth factor binding protein-3 protease. J Endocrinol.1994;142:407-415.

103. Vakar-Lopez F, Cheng CJ, Kim J, et al. Up-regulation ofMDA-BF-1, a secreted isoform of ErbB3, in metastaticprostate cancer cells and activated osteoblasts in bone mar-row. J Pathol. 2004;203:688-695.

104. Liang AK, Liu J, Mao SA, Siu VS, Lee YC, Lin SH.Expression of recombinant MDA-BF-1 with a kinase rec-ognition site and a 7-histidine tag for receptor bindingand purification. Protein Expr Purif. 2005;44:58-64.

105. Wiesner C, Nabha SM, Dos Santos EB, et al. C-kit andits ligand stem cell factor: potential contribution to pros-tate cancer bone metastasis. Neoplasia. 2008;10:996-1003.

106. Yi B, Williams PJ, Niewolna M, Wang Y, Yoneda T. Tu-mor-derived platelet-derived growth factor-BB plays a criti-cal role in osteosclerotic bone metastasis in an animalmodel of human breast cancer. Cancer Res. 2002;62:917-923.

107. Mehrotra M, Krane SM, Walters K, Pilbeam C. Differen-tial regulation of platelet-derived growth factor stimulatedmigration and proliferation in osteoblastic cells. J Cell Bio-chem. 2004;93:741-752.

108. Dunstan CR, Boyce R, Boyce BF, et al. Systemic admin-istration of acidic fibroblast growth factor (FGF-1) pre-vents bone loss and increases new bone formation inovariectomized rats. J Bone Miner Res. 1999;14:953-959.

109. Street J, Bao M, deGuzman L, et al. Vascular endothelialgrowth factor stimulates bone repair by promoting angio-genesis and bone turnover. Proc Natl Acad Sci U S A.2002;99:9656-9661.

110. Chen G, Shukeir N, Potti A, et al. Up-regulation of Wnt-1 and beta-catenin production in patients with advancedmetastatic prostate carcinoma: potential pathogenetic andprognostic implications. Cancer. 2004;101:1345-1356.

111. Tian E, Zhan F, Walker R, et al. The role of the Wnt-sig-naling antagonist DKK1 in the development of osteolyticlesions in multiple myeloma. N Engl J Med. 2003;349:2483-2494.

112. Hall CL, Kang S, MacDougald OA, Keller ET. Role ofWnts in prostate cancer bone metastases. J Cell Biochem.2006;97:661-672.

113. Roudier MP, Morrissey C, True LD, Higano CS, VessellaRL, Ott SM. Histopathological assessment of prostate can-cer bone osteoblastic metastases. J Urol. 2008;180:1154-1160.

114. Taichman RS, Loberg RD, Mehra R, Pienta KJ. Theevolving biology and treatment of prostate cancer. J ClinInvest. 2007;117:2351-2361.

115. Mizutani K, Sud S, Pienta KJ. Prostate cancer promotesCD11b positive cells to differentiate into osteoclasts. J CellBiochem. 2009;106:563-569.

116. Li X, Loberg R, Liao J, et al. A destructive cascade medi-ated by CCL2 facilitates prostate cancer growth in bone.Cancer Res. 2009;69:1685-1692.

117. Reddi AH, Roodman D, Freeman C, Mohla S. Mecha-nisms of tumor metastasis to the bone: challenges andopportunities. J Bone Miner Res. 2003;18:190-194.

118. Ye L, Kynaston HG, Jiang WG. Bone metastasis in pros-tate cancer: molecular and cellular mechanisms [review].Int J Mol Med. 2007;20:103-111.

119. Body JJ, Greipp P, Coleman RE, et al. A phase I study ofAMGN-0007, a recombinant osteoprotegerin construct, inpatients with multiple myeloma or breast carcinomarelated bone metastases. Cancer. 2003;97:887-892.

120. Jones DH, Nakashima T, Sanchez OH, et al. Regulationof cancer cell migration and bone metastasis by RANKL.Nature. 2006;440:692-696.

121. Mori K, Le Goff B, Charrier C, Battaglia S, Heymann D,Redini F. DU145 human prostate cancer cells expressfunctional receptor activator of NFkappaB: new insights inthe prostate cancer bone metastasis process. Bone. 2007;40:981-990.

122. Fizazi K, Bosserman L, Gao G, Skacel T, Markus R.Denosumab treatment of prostate cancer with bone metas-tases and increased urine N-telopeptide levels after therapywith intravenous bisphosphonates: results of a randomizedphase II trial. J Urol. 2009;182:509-515; discussion 515-516.

123. Nelson JB, Hedican SP, George DJ, et al. Identification ofendothelin-1 in the pathophysiology of metastatic adeno-carcinoma of the prostate. Nat Med. 1995;1:944-949.

124. Carducci MA, Padley RJ, Breul J, et al. Effect of endothe-lin-A receptor blockade with atrasentan on tumor progres-sion in men with hormone-refractory prostate cancer: arandomized, phase II, placebo-controlled trial. J ClinOncol. 2003;21:679-689.

125. Carducci MA, Saad F, Abrahamsson PA, et al. A phase 3randomized controlled trial of the efficacy and safety ofatrasentan in men with metastatic hormone-refractoryprostate cancer. Cancer. 2007;110:1959-1966.

126. Gramoun A, Shorey S, Bashutski JD, et al. Effects ofVitaxin, a novel therapeutic in trial for metastatic bonetumors, on osteoclast functions in vitro. J Cell Biochem.2007;102:341-352.

127. Diaz R, Nguewa PA, Diaz-Gonzalez JA, et al. The novelAkt inhibitor Palomid 529 (P529) enhances the effect ofradiotherapy in prostate cancer. Br J Cancer. 2009;100:932-940.

128. Kong D, Li Y, Wang Z, Banerjee S, Sarkar FH. Inhibitionof angiogenesis and invasion by 3,30-diindolylmethane ismediated by the nuclear factor-kappaB downstream targetgenes MMP-9 and uPA that regulated bioavailability ofvascular endothelial growth factor in prostate cancer. Can-cer Res. 2007;67:3310-3319.

129. Ahmad A, Kong D, Sarkar SH, Wang Z, Banerjee S, Sar-kar FH. Inactivation of uPA and its receptor uPAR by3,30-diindolylmethane (DIM) leads to the inhibition ofprostate cancer cell growth and migration. J Cell Biochem.2009;107:516-527.

130. Sachdev D, Li SL, Hartell JS, Fujita-Yamaguchi Y, MillerJS, Yee D. A chimeric humanized single-chain antibodyagainst the type I insulin-like growth factor (IGF) receptorrenders breast cancer cells refractory to the mitogeniceffects of IGF-I. Cancer Res. 2003;63:627-635.

131. Haluska P, Carboni JM, TenEyck C, et al. HER receptorsignaling confers resistance to the insulin-like growth

Bone Metastases and Prostate Cancer/Ibrahim et al

Cancer March 15, 2010 1417

Page 13: Pathogenesis of osteoblastic bone metastases from prostate cancer

factor-I receptor inhibitor, BMS-536924. Mol CancerTher. 2008;7:2589-2598.

132. Leeming DJ, Hegele A, Byrjalsen I, et al. Biochemicalmarkers for monitoring response to therapy: evidence forhigher bone specificity by a novel marker compared withroutine markers. Cancer Epidemiol Biomarkers Prev.2008;17:1269-1276.

133. Lein M, Miller K, Wirth M, et al. Bone turnover markersas predictive tools for skeletal complications in men with

metastatic prostate cancer treated with zoledronic acid.Prostate. 2009;69:624-632.

134. Klepzig M, Jonas D, Oremek GM. Procollagen type 1amino-terminal propeptide: a marker for bone metastases inprostate carcinoma. Anticancer Res. 2009;29:671-673.

135. Lipton A, Cook R, Saad F, et al. Normalization of bonemarkers is associated with improved survival in patients withbone metastases from solid tumors and elevated bone resorp-tion receiving zoledronic acid. Cancer. 2008;113:193-201.

Review Article

1418 Cancer March 15, 2010