The hematogenous metastatic spread of prostate cancer is preferentially to bone and can result in significant patient morbidity. Although these metastatic lesions are typically osteoblastic, bone resorption is believed to have a prerequisite role in their development. Src kinase has been identified to contribute to prostate cancer tumor growth and metastasis. In addition, Src is also essential in bone metabolism, especially in bone resorption. We hypothesized that inhibiting Src activity with the specific Src family kinase inhibitor saracatinib (AZD0530) would inhibit tumor cell growth and osteoclast differentiation in the tumor-bone interface, thus providing a new approach for advanced prostate cancer. We found that saracatinib inhibited PC-3 cell growth and invasion in a dose-dependent manner. Phosphorylation of Src, focal adhesion kinase, and P38 kinases was inhibited by saracatinib at the submicromolar range. Saracatinib also inhibited the expression and secretion of invasion-related molecules interlukin-8, urokinase-type plasminogen activator, and matrix metalloprotease-9. Receptor activator of NF-κB ligand (RANKL)–induced osteoclastogenesis and signaling were inhibited by saracatinib in both macrophages and PC-3 cells. In in vivo studies, control mice developed more severe osteolytic lesions compared with the treatment group. Immunohistochemical and biochemical assays of bone metabolites confirmed that saracatinib preserved bone architecture in the presence of prostate cancer tumor cells. In summary, we have shown the inhibition of PC3 cell growth and invasion by saracatinib. Src inhibition also blocked the RANKL stimulatory pathway in osteoclasts and PC3 cells. The inhibition of Src thus targets multiple sites involved in prostate cancer bone metastasis, which may offer a therapeutic advantage in treating advanced prostate cancer. Mol Cancer Ther; 9(6); 1629–37. ©2010 AACR.

Prostate cancer skeletal involvement is the most common metastatic site in advanced-stage disease, observed in up to 80% of patients (1). Unlike other solid tumor metastases that cause bone loss, prostate cancer bone metastases result in an overall increase in bone mass or osteoblastogenesis. It is not entirely clear why prostate tumor cells localize to the bone environment. The enriched blood source and complex growth factors in the bone marrow provide a good soil for circulating prostate tumor cells to seed and multiply. Growth factors released from tumor cells support increase of both bone resorption and formation (2). Increased osteoclastogenesis has been detected in prostate cancer bone metastases (3). The initial osteoclast-mediated osteolytic process is critical to the subsequent osteoblastic lesions, and a metastatic phenotype consists of a combination of both bone resorption and formation (46). A vicious cycle in the tumor cell–bone interface drives the metastasis, whereby growth factors secreted by tumor cells stimulate osteoclast formation and matrix turnover. This osteoclastic resorption causes the local release of more growth factors that in turn activate tumor growth. Src, a non–receptor tyrosine kinase, is implicated in the cross-talk in this tumor-bone interface through its central role in bone metastasis and tumor progression.

The importance of Src kinase in osteoclasts was first discovered in Src knockout mice in which defective osteoclastic resorption gave rise to the phenotype of osteopetrosis and toothlessness (7, 8). In bone formation and remodeling, Src is activated in osteoclasts upon attachment of cells to bone matrix through integrin receptors, and activated Src recruits several signaling proteins for motility and cytoskeletal rearrangement (9, 10). Src is also important in the receptor activator of NF-κB ligand (RANKL)/RANK/osteoprotegerin signaling pathway, which contributes to osteoclast differentiation. Membrane-bound or soluble RANKL binds to its receptor RANK to initiate the signaling cascade leading to osteoclast activation. Upon ligand binding, RANK recruits the tumor necrosis factor receptor–associated factors to its cytoplasmic domain (1113). This crucial step leads to activation of NF-κB (14), mitogen-activated protein kinase (MAPK; refs. 1517), and Src (10, 1820) signaling pathways in osteoclasts. In addition to osteoclast activation, Src recruits phosphatidylinositol-3 kinase and activates the Akt/mammalian target of rapamycin pathway to promote osteoclast survival (21, 22).

Our previous studies showed that Src kinase plays an important role in promoting androgen-independent prostate tumor growth (2326). Upregulation of Src family kinase (SFK) activities has been reported in 28% of androgen-independent prostate cancer patients (27). Moreover, this enhanced SFK activity correlated with prostate cancer bone metastasis and a significant decrease in survival. The specific SFK inhibitor saracatinib (AZD0530) inhibited the activities of Src and focal adhesion kinase (FAK) kinases and their downstream substrates. Phosphorylation of Src and FAK was inhibited by saracatinib in the nanomolar range. As a result, tumor metastasis was profoundly inhibited in an animal study using castration-resistant LNCaP expressing neuropeptide gastrin-releasing peptide (LNCaP-GRP cells; ref. 26).

We proposed to use saracatinib not only as an adjunct to hormone ablation therapy and to target patients with upregulation in SFK activity but also for patients with bone metastasis. In this study, we use PC-3 cells for their ability to steadily induce osteolysis when implanted in the mouse tibia. In addition to downregulation of cell proliferation and migration as reported before, saracatinib inhibited the expression of interleukin-8 (IL-8), urokinase plasminogen activator (uPA), and matrix metalloprotease-9 (MMP-9), and PC-3 invasion, which altogether suggests that saracatinib may inhibit prostate tumor cell invasion into the bone matrix. Induced differentiation of macrophage cells into osteoclasts by RANKL was inhibited by saracatinib, as was the activation of Src, P38, and IκB-α in PC-3 cells—events generally mediated through RANKL stimulation. Our animal study further shows the ability of saracatinib to retard osteolytic lesions, a potential therapeutic advantage in treating patients with prostate cancer.

Cell lines

The PC-3 prostate cancer cell line was obtained from the American Type Culture Collection and cultured in RPMI 1640 supplemented with 5% fetal bovine serum (FBS). RAW264.7 cells, a murine osteoclast precursor, was from the American Type Culture Collection and kept in DMEM with 10% FBS.

Proliferation assay

PC-3 cells were seeded (2,000 per well) in triplicates in 96-well plates. Varied concentrations of saracatinib (0–5 μmol/L, AstraZeneca) were added, and cell growth was monitored by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT, Sigma) assay at 1, 3, and 5 days posttreatment.

Invasion assay

Invasion assays were done according to procedures described by Lochter and colleagues (28) and modified as follows. PC-3 cells (1 × 105) were suspended in growth medium containing dimethyl sulfoxide or various concentrations of saracatinib (8 nmol/L to 5 μmol/L in 5-fold increments) into the cell culture inserts containing 8 μmol/L pores for 24-well plates. The inserts were coated with Matrigel (2–3 mg/mL protein) and allowed to solidify before cell plating. The lower chambers were filled with 300 μL of growth medium containing control or drugs of corresponding concentrations. After 72 hours in culture, cells were fixed with 5% glutaraldehyde in PBS and stained with 0.5% toluidine blue in 2% Na2CO3. Only cells that penetrated the membrane were counted in five microscopic fields per filter.

Osteoclast differentiation from RAW264.7 cells

Osteoclast formation assay was done by plating 5 × 104 RAW264.7 cells per well in 24-well plates in DMEM with 10% FBS for 4 days. Soluble RANKL (100 ng/mL, Santa Cruz Biotechnology) was added to stimulate osteoclast differentiation, and saracatinib (1 μmol/L) was added for inhibition. At the end of incubation, the cells were fixed and stained with a tartrate-resistant acid phosphatase (TRAP) staining kit (Acid Phosphatase Kit 387-A, Sigma), and TRAP-positive staining osteoclasts (multinuclear cells with > 3 nuclei) were counted in five different microscopic fields.

Reverse transcriptase-PCR and quantitative reverse transcriptase-PCR

Total RNA was isolated from PC-3 cells treated with or without 1 μmol/L of saracatinib at different time points (0, 6, and 24 hours). Two micrograms of RNA were reverse transcribed into cDNA followed by PCR using specific primers. Quantitative PCR was done in iCycler with SYBR Greener QPCR iCYLer (Invitrogen), and the output was normalized against the controlled amplification products with glyceraldehyde-3-phosphate dehydrogenase.

Gel zymography

MMP-9 activity was analyzed with gelatin zymography as described below. Conditioned media harvested from PC-3 treated with or without various concentrations of saracatinib for 48 hours were concentrated through centrifugation using Microcon concentrators (Amicon). Concentrated conditioned medium was normalized to total cellular protein (equivalent to 100 μg per loading), then subjected to electrophoresis in gels containing 0.2% gelatin without denaturing. After electrophoresis, the gels were washed in 2% Triton X-100 for 30 minutes and incubated in 50 mmol/L Tris-HCl (pH 7.4), 200 mmol/L NaCl, and 10 mmol/L CaCl2 at 37°C overnight. Gels were then stained with Coomassie brilliant blue R-250 and destained with methanol/HOAc/H2O (4:1:5) until clear bands appeared.

Western blot analyses

PC-3 cells were cultured to 70% confluency and treated with various concentrations of saracatinib for 1 hour. Cells were then lysed in ice-cold buffer (Tris 25 mmol/L, MgCl2 10 mmol/L, β-glycerolphosphate 25 mmol/L, Na3VO4 5 mmol/L, and protease inhibitor cocktail) and ruptured through three freeze-thaw cycles. Cell lysates were separated from debris by centrifugation, and protein concentrations were determined by the bicinchoninic acid assay (Pierce). Thirty micrograms of protein were separated on SDS-PAGE and electrotransferred onto polyvinylidene difluoride membrane. Immunoblotting was done by incubating membranes with the primary antibodies overnight, followed by 1 hour of secondary antibody incubation. Signals were detected by SuperSignal West Pico CL (Pierce) coupled with X-ray film exposure. For inhibition of the RANKL/RANK pathway, PC-3 cells were serum-starved for 48 hours before stimulation by RANKL (100 ng/mL) in 2% FBS medium for 0, 2, 5, 15, 30, and 60 minutes. SFK inhibitor saracatinib (1 μmol/L) was added to the last three time points together with RANKL. At the end of treatments, cells were collected as described, and lysates were subjected to gel electrophoresis. Antibodies against p-Src (Tyr416), total Src (36D10), Akt, p-Akt (Ser473), P38, p-P38 (Thr180/Tyr182, 28B10), extracellular signal-regulated kinase 1/2 (Erk1/2; p44/42MAPK), IκBα (44D4), and p-IκBα (Ser32, 14D4) were purchased from Cell Signaling; anti–p-FAK (Y397) was from BD Biosciences; and anti–p-FAK (Y861) was from Affinity BioReagents.

In vivo inhibition study

PC-3 cells (2 × 105) mixed with equal amounts of Matrigel in a final volume of 10 μL were injected into 24 male severe combined immunodeficient (SCID) mice tibia. Intratibial injections were done using a 29-gauge, 1/2-inch needle inserted through the tibial plateau of the flexed knee. After animals recuperated from the surgery (7 days later), they were randomly divided into two groups and given oral saracatinib (25 mg/kg) or buffer (0.5% hydroxypropyl methylcellulose and 0.1% Tween 80) daily for 8 weeks from implantation. Anesthetized mice had bone lesions monitored weekly using Faxitron (Faxitron X-ray Corp.). At the end of 8 weeks, animals were sacrificed with collection of urine, sera, and tibias for biochemical and pathologic analyses. The tibias were fixed in 10% buffered formalin for 2 days, followed by decalcification in 10% EDTA solution for 2 weeks at room temperature with occasional stirring and solution changes. Specimens were paraffin embedded, sectioned, and stained with H&E and TRAP. Histomorphometric analysis was done on an Olympus system. The number of large active osteoclasts (TRAP-positive osteoclasts with three or more nuclei) per millimeter of tumor-bone interface was measured from 4-week control and treatment bone sections in five different fields at 200× magnification. Total tumor percentage in the medullary cavity of each bone from 8-week controls and treatment samples was measured at 40× magnification.

Serum pyridinoline and urine helical peptide assays

The pyridinoline cross-links and helical peptides representing the degree of bone resorption were measured by EIA assays (Metra Serum pyridinoline assay and Metra Helical Peptide EIA Kit, Quidel).

Statistics

Statistical analyses were done using unpaired Student's t tests (StatView, SAS Institute). Significant results were determined as P ≤ 0.05.

SFK inhibitor saracatinib inhibits PC-3 growth

We have reported that SFK inhibitor saracatinib efficiently inhibits the growth of most prostate cancer cells, LNCaP, PC-3, DU-145, and CWR22Rv1, with IC50 values in the low micromolar range (29). PC-3 cells are especially sensitive to saracatinib with an IC50 of 0.7 μmol/L (Fig. 1A). Inhibition of phosphorylation of Src kinase and its immediate substrate FAK was achievable with submicromolar levels of saracatinib, as was the case for MAPK-P38. Inhibition of Akt required higher doses of saracatinib, implying an indirect relationship of Src and Akt (Fig. 1B).

SFK inhibitor saracatinib inhibits PC-3 invasion

Saracatinib is very potent in inhibiting PC-3 cell migration with an IC50 value of ∼50 nmol/L. This SFK inhibitor also blocks PC-3 penetration through Matrigel in invasion assay (Fig. 2A). After 3 days, cell invasion was reduced to 57% and 25% of control in the presence of 40 and 200 nmol/L of saracatinib, respectively. PC-3 cells remained 78% and 73% viable using the same concentrations of inhibitor over the same period of time; thus, the reduction was indeed the effect of saracatinib on cell invasion. The expression of invasion-related molecules IL-8 and uPA was inhibited by saracatinib in a timely manner (Fig. 2B). Treatment of PC-3 cells with 1 μmol/L of saracatinib reduced IL-8 transcripts to 30% in 6 hours and to 18% in 24 hours (Fig. 2C). The same treatment halved uPA expression in 6 hours and further decreased to 44% after 24 hours. The level of uPA receptor was only slightly reduced (to around 85%) after 24 hours of treatment. MMP-9 activity in PC-3 cultured medium was inhibited by saracatinib in a dose-responsive manner (Fig. 2D).

Saracatinib inhibits RANKL-induced osteoclastogenesis and signaling

Murine macrophage RAW264.7 precursors for osteoclasts may be stimulated by RANKL (100 ng/mL) to undergo osteoclast differentiation. Addition of 1 μmol/L saracatinib drastically inhibited osteoclast differentiation by limiting the number of TRAP-stained multinuclear osteoclasts to less than 18% of control (Fig. 3A). Treating RAW264.7 cells with this concentration of saracatinib alone did not cause massive cell death as shown in the staining (Fig. 3B); hence, the reduction in TRAP staining is likely due to inhibition of osteoclast differentiation. It has been reported that functional RANK is expressed in prostate cancer cell lines such as PC-3 and DU-145 (30). The receptors render these prostate cancer cells under the control of bound or soluble RANKL from osteoblasts or stroma to enhance tumor growth in the bone environment. We therefore tested if saracatinib would block the RANKL/RANK pathway in PC-3 cells. Activation of Src, P38, and IκBα (a few representatives from the RANKL/RANK pathway) was detectable at 2 minutes from RANKL addition and peaked within 15 minutes (Fig. 3C). Saracatinib completely blocked phosphorylation of these kinases at 15, 30, and 60 minutes.

In vivo inhibition with a PC-3 tibial model

Based on our in vitro observations, we then implanted PC-3 cells into the tibia of SCID mice to study the effect of the Src inhibitor in vivo. PC-3 cells cause osteolysis when injected directly into mouse bones (3133). In our study, osteolysis started to appear in the control group 3 weeks after injection. Radiographs taken at 4 and 8 weeks (end points) showed that saracatinib significantly inhibited the osteolytic lesion in the treatment group compared with controls (Fig. 4A). At 5 weeks from surgery, 9 of 12 control mice showed osteolytic lesions compared with only 4 in the treatment group. TRAP staining of the tibial samples taken at 4 weeks showed growth of tumor cells leading to loss of bone architecture in the control tibias with characteristic TRAP staining signifying the presence of osteoclasts (Fig. 4B). Most of the tibias from the treatment animals retained bone integrity, bearing no signs of tumor cells or osteoclasts. The osteoclast perimeters from the 4-week bone samples were counted. Saracatinib decreased TRAP-positive osteoclasts in tibias of tumor-bearing mice by >10 fold when compared with controls. At the end of 8 weeks, all but one mouse in the control group developed severe lesions, but only half of the treatment animals had milder lesions. The areas of tumor versus bone were measured from histologic staining and plotted to show that saracatinib reduced this ratio in the treatment group to less than half of controls (Fig. 4C). In addition to bone histomorphometric measurements, the effect of saracatinib on released mouse bone metabolites, such as pyridinoline cross-links in serum and α-helical peptides in urine, were measured. Compared with the control group, treated mice had a 46% reduction in pyridinoline (P = 0.07, not statistically significant) and 60% reduction in α-helical peptide (P < 0.01; Fig. 4D).

When standard therapy for advanced prostate cancer using androgen ablation fails, there are limited treatment options for patients with prostate cancer metastatic to bone. At present, bone health is fostered by use of calcium, vitamin D, and zolendronic acid. Unbalanced bone remodeling mediated by prostate tumor cells replaces normal bone tissue with highly disorganized osteoblasts. This leads to predominantly osteoblastic lesions causing significant pain in patients with advanced prostate cancer (34). New therapeutic strategies such as small-molecule inhibitors of signaling pathways responsible for cancer progression are desirable. Src kinase has been implicated in prostate tumor proliferation, migration, survival (23, 35, 36), and progression to castration resistance (26). Many recently developed small-molecule SFK inhibitors, including bosutinib (SKI-606; ref. 37), dasatinib (BMS-354825; refs. 38, 39), and saracatinib are able to reduce the proliferation, migration, and invasion of prostate cancer cell lines in vitro and are currently in clinical trials. These inhibitors also decrease prostate cancer growth and metastasis in mouse xenograft studies (26, 29, 39). Recent studies using saracatinib showed that Src has a pivotal role in the formation and activation of human osteoclasts (40) and is involved in breast cancer bone metastasis regardless of the estrogen receptor status (41). A phase I study showed a dose-dependent decrease in bone resorption markers in men receiving saracatinib daily for 2 weeks (42). There were no significant adverse effects. Dasatinib alone and in combination therapy with docetaxel improved bone mineral density in the LNCaP C4-2B tumor–implanted tibias (43).

Using saracatinib, we were able to show a dose-dependent inhibition of proliferation and migration in PC-3 and DU-145 cells (29). Western blot analysis showed that Src phosphorylation in saracatinib-treated cells was reduced in a dose- and time-dependent manner. Intracellular Src-activating proteins, including FAK, P130Cas, and paxillin, were also inhibited, supporting the efficacy of saracatinib in preventing prostate cancer progression. In addition to the previous prostate cancer lines that lack androgen receptor, LNCaP-GRP cells that contain androgen receptor but grow independent of androgen were also sensitive to saracatinib. With the importance of Src in both bone remodeling and prostate cancer tumor progression, the use of Src inhibitors has been proposed in the treatment of prostate cancer bone metastasis (44, 45). In our study, saracatinib has shown specific inhibition of FAK and P38 kinase activation downstream of Src in PC-3 cells. This block in signaling may prevent cytoskeletal rearrangement and migration through FAK (29). We again observed the discrepancy between the doses of saracatinib required for inhibiting Src phosphorylation and cell/tumor growth as discussed in our previous article (29). In addition to speculation of partial inhibition of Src kinase on growth and its downstream signaling, we recently reported that saracatinib induced macroautophagy in cells to spare them from apoptosis by inhibiting the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway (46). Blocking autophagy with drugs such as chloroquine significantly improved the efficacy of saracatinib in treating PC-3 subcutaneous tumors, which may also be applicable to bone treatment. Saracatinib reduced the synthesis of IL-8, uPA, and MMP-9, molecules involved in angiogenesis and basement membrane degradation (47), in PC-3 cells, which is consistent with cell invasion inhibition. RANKL secreted from tumor and stroma cells is responsible for osteoclast differentiation in the tumor-bone interface. Using RAW264.7 cells as a model for osteoclast differentiation induced by RANKL, saracatinib significantly inhibited the formation of osteoclasts. Furthermore, the RANKL-induced RANK/RANKL pathway in PC-3 cells was also inhibited by saracatinib along with P38 and IκBα. Finally, in vivo experiments showed that progression of PC-3 implants in the metastatic tibial site was inhibited by saracatinib. The inhibition not only prevented proliferation of PC-3 cells but also hindered multiplication of osteoclasts in the bone environment. Osteoclast perimeter counts were significantly lower at the 4-week time point, and the tumor/bone ratio was halved at the end of the in vivo study in the inhibitor-treated specimens. Reduction in secretion of bone turnover markers pyridinoline and helical peptides in mice receiving saracatinib was consistent with fewer osteolytic lesions and decreased osteoclast proliferation. Although the majority of advanced prostate cancer patients develop osteoblastic lesions, bone resorption occurs before new bone formation. Our report on the efficacy of saracatinib on inhibiting PC-3 osteolytic bone lesions is applicable to patients with prostate cancer metastatic to bone.

Results presented herein show the utility of saracatinib in advanced prostate cancer with mechanistic and in vivo animal studies supporting clinical trials using saracatinib as a treatment for prostate cancer patients with bone metastasis.

C.P. Evans is a consultant for AstraZeneca. No other potential conflicts of interest were disclosed.

We thank Maggie Chiu of Pathology for making sections and staining the animal study samples.

Grant Support: Department of Defense PC10520 and Prostate Cancer Foundation Competitive Award (C.P. Evans).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Bubendorf
L
,
Schopfer
A
,
Wagner
U
, et al
. 
Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients
.
Human Pathol
2000
;
31
:
578
83
.
2
Keller
ET
,
Zhang
J
,
Cooper
CR
, et al
. 
Prostate carcinoma skeletal metastases: cross-talk between tumor and bone
.
Cancer Metastasis Rev
2001
;
20
:
333
49
.
3
Roato
I
,
D'Amelio
P
,
Gorassini
E
, et al
. 
Osteoclasts are active in bone forming metastases of prostate cancer patients
.
PLoS ONE
2008
;
3
:
e3627
.
4
Inoue
H
,
Nishimura
K
,
Oka
D
, et al
. 
Prostate cancer mediates osteoclastogenesis through two different pathways
.
Cancer Lett
2005
;
223
:
121
8
.
5
Whang
PG
,
Schwarz
EM
,
Gamradt
SC
,
Dougall
WC
,
Lieberman
JR
. 
The effects of RANK blockade and osteoclast depletion in a model of pure osteoblastic prostate cancer metastasis in bone
.
J Orthop Res
2005
;
23
:
1475
83
.
6
Zhang
J
,
Dai
J
,
Yao
Z
,
Lu
Y
,
Dougall
W
,
Keller
ET
. 
Soluble receptor activator of nuclear factor κB Fc diminishes prostate cancer progression in bone
.
Cancer Res
2003
;
63
:
7883
90
.
7
Boyce
BF
,
Yoneda
T
,
Lowe
C
,
Soriano
P
,
Mundy
GR
. 
Requirement of pp60c-src expression for osteoclasts to form ruffled borders and resorb bone in mice
.
J Clin Invest
1992
;
90
:
1622
7
.
8
Soriano
P
,
Montgomery
C
,
Geske
R
,
Bradley
A
. 
Targeted disruption of the c-src proto-oncogene leads to osteopetrosis in mice
.
Cell
1991
;
64
:
693
702
.
9
Insogna
KL
,
Sahni
M
,
Grey
AB
, et al
. 
Colony-stimulating factor-1 induces cytoskeletal reorganization and c-src-dependent tyrosine phosphorylation of selected cellular proteins in rodent osteoclasts
.
J Clin Invest
1997
;
100
:
2476
85
.
10
Miyazaki
T
,
Sanjay
A
,
Neff
L
,
Tanaka
S
,
Horne
WC
,
Baron
R
. 
Src kinase activity is essential for osteoclast function
.
J Biol Chem
2004
;
279
:
17660
6
.
11
Darnay
BG
,
Haridas
V
,
Ni
J
,
Moore
PA
,
Aggarwal
BB
. 
Characterization of the intracellular domain of receptor activator of NF-κB (RANK). Interaction with tumor necrosis factor receptor-associated factors and activation of NF-κb and c-Jun N-terminal kinase
.
J Biol Chem
1998
;
273
:
20551
5
.
12
Galibert
L
,
Tometsko
ME
,
Anderson
DM
,
Cosman
D
,
Dougall
WC
. 
The involvement of multiple tumor necrosis factor receptor (TNFR)-associated factors in the signaling mechanisms of receptor activator of NF-κB, a member of the TNFR superfamily
.
J Biol Chem
1998
;
273
:
34120
7
.
13
Hsu
H
,
Lacey
DL
,
Dunstan
CR
, et al
. 
Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand
.
Proc Natl Acad Sci U S A
1999
;
96
:
3540
5
.
14
Iotsova
V
,
Caamano
J
,
Loy
J
,
Yang
Y
,
Lewin
A
,
Bravo
R
. 
Osteopetrosis in mice lacking NF-κB1 and NF-κB2
.
Nat Med
1997
;
3
:
1285
9
.
15
David
JP
,
Sabapathy
K
,
Hoffmann
O
,
Idarraga
MH
,
Wagner
EF
. 
JNK1 modulates osteoclastogenesis through both c-Jun phosphorylation-dependent and -independent mechanisms
.
J Cell Sci
2002
;
115
:
4317
25
.
16
Matsumoto
M
,
Sudo
T
,
Saito
T
,
Osada
H
,
Tsujimoto
M
. 
Involvement of p38 mitogen-activated protein kinase signaling pathway in osteoclastogenesis mediated by receptor activator of NF-κB ligand (RANKL)
.
J Biol Chem
2000
;
275
:
31155
61
.
17
Wagner
EF
. 
Functions of AP1 (Fos/Jun) in bone development
.
Ann Rheum Dis
2002
;
61 Suppl 2
:
ii40
2
.
18
Koga
S
,
Yogo
K
,
Yoshikawa
K
, et al
. 
Physical and functional association of c-Src and adhesion and degranulation promoting adaptor protein (ADAP) in osteoclastogenesis in vitro
.
J Biol Chem
2005
;
280
:
31564
71
.
19
Wong
BR
,
Rho
J
,
Arron
J
, et al
. 
TRANCE is a novel ligand of the tumor necrosis factor receptor family that activates c-Jun N-terminal kinase in T cells
.
J Biol Chem
1997
;
272
:
25190
4
.
20
Xing
L
,
Venegas
AM
,
Chen
A
, et al
. 
Genetic evidence for a role for Src family kinases in TNF family receptor signaling and cell survival
.
Genes Dev
2001
;
15
:
241
53
.
21
Glantschnig
H
,
Fisher
JE
,
Wesolowski
G
,
Rodan
GA
,
Reszka
AA
. 
M-CSF, TNFα and RANK ligand promote osteoclast survival by signaling through mTOR/S6 kinase
.
Cell Death Differ
2003
;
10
:
1165
77
.
22
Sugatani
T
,
Hruska
KA
. 
Akt1/Akt2 and mammalian target of rapamycin/Bim play critical roles in osteoclast differentiation and survival, respectively, whereas Akt is dispensable for cell survival in isolated osteoclast precursors
.
J Biol Chem
2005
;
280
:
3583
9
.
23
Chang
YM
,
Kung
HJ
,
Evans
CP
. 
Nonreceptor tyrosine kinases in prostate cancer
.
Neoplasia (New York, NY)
2007
;
9
:
90
100
.
24
Guo
Z
,
Dai
B
,
Jiang
T
, et al
. 
Regulation of androgen receptor activity by tyrosine phosphorylation
.
Cancer Cell
2006
;
10
:
309
19
.
25
Lee
LF
,
Guan
J
,
Qiu
Y
,
Kung
HJ
. 
Neuropeptide-induced androgen independence in prostate cancer cells: roles of nonreceptor tyrosine kinases Etk/Bmx, Src, and focal adhesion kinase
.
Mol Cell Biol
2001
;
21
:
8385
97
.
26
Yang
JC
,
Ok
JH
,
Busby
JE
,
Borowsky
AD
,
Kung
HJ
,
Evans
CP
. 
Aberrant activation of androgen receptor in a new neuropeptide-autocrine model of androgen-insensitive prostate cancer
.
Cancer Res
2009
;
69
:
151
60
.
27
Tatarov
O
,
Mitchell
TJ
,
Seywright
M
,
Leung
HY
,
Brunton
VG
,
Edwards
J
. 
SRC family kinase activity is up-regulated in hormone-refractory prostate cancer
.
Clin Cancer Res
2009
;
15
:
3540
9
.
28
Lochter
A
,
Srebrow
A
,
Sympson
CJ
,
Terracio
N
,
Werb
Z
,
Bissell
MJ
. 
Misregulation of stromelysin-1 expression in mouse mammary tumor cells accompanies acquisition of stromelysin-1-dependent invasive properties
.
J Biol Chem
1997
;
272
:
5007
15
.
29
Chang
YM
,
Bai
L
,
Liu
S
,
Yang
JC
,
Kung
HJ
,
Evans
CP
. 
Src family kinase oncogenic potential and pathways in prostate cancer as revealed by AZD0530
.
Oncogene
2008
;
27
:
6365
75
.
30
Mori
K
,
Le Goff
B
,
Charrier
C
,
Battaglia
S
,
Heymann
D
,
Redini
F
. 
DU145 human prostate cancer cells express functional receptor activator of NFκB: new insights in the prostate cancer bone metastasis process
.
Bone
2007
;
40
:
981
90
.
31
Kim
SJ
,
Uehara
H
,
Karashima
T
,
Shepherd
DL
,
Killion
JJ
,
Fidler
IJ
. 
Blockade of epidermal growth factor receptor signaling in tumor cells and tumor-associated endothelial cells for therapy of androgen-independent human prostate cancer growing in the bone of nude mice
.
Clin Cancer Res
2003
;
9
:
1200
10
.
32
Lee
YP
,
Schwarz
EM
,
Davies
M
, et al
. 
Use of zoledronate to treat osteoblastic versus osteolytic lesions in a severe-combined-immunodeficient mouse model
.
Cancer Res
2002
;
62
:
5564
70
.
33
Miwa
S
,
Mizokami
A
,
Keller
ET
,
Taichman
R
,
Zhang
J
,
Namiki
M
. 
The bisphosphonate YM529 inhibits osteolytic and osteoblastic changes and CXCR-4-induced invasion in prostate cancer
.
Cancer Res
2005
;
65
:
8818
25
.
34
Chirgwin
JM
,
Mohammad
KS
,
Guise
TA
. 
Tumor-bone cellular interactions in skeletal metastases
.
J Musculoskelet Neuronal Interact
2004
;
4
:
308
18
.
35
Kopetz
S
,
Shah
AN
,
Gallick
GE
. 
Src continues aging: current and future clinical directions
.
Clin Cancer Res
2007
;
13
:
7232
6
.
36
Saad
F
. 
Src as a therapeutic target in men with prostate cancer and bone metastases
.
BJU Int
2009
;
103
:
434
40
.
37
Jallal
H
,
Valentino
ML
,
Chen
G
,
Boschelli
F
,
Ali
S
,
Rabbani
SA
. 
A Src/Abl kinase inhibitor, SKI-606, blocks breast cancer invasion, growth, and metastasis in vitro and in vivo
.
Cancer Res
2007
;
67
:
1580
8
.
38
Nam
S
,
Kim
D
,
Cheng
JQ
, et al
. 
Action of the Src family kinase inhibitor, dasatinib (BMS-354825), on human prostate cancer cells
.
Cancer Res
2005
;
65
:
9185
9
.
39
Park
SI
,
Gallick
GE
. 
c-Src activation in the tumor-associated endothelial cells contributes to the lymph node metastases of prostate cancer cells
.
Proc Am Assoc Cancer Res
2007
;
48
:
3002
.
40
de Vries
TJ
,
Mullender
MG
,
van Duin
MA
, et al
. 
The Src inhibitor AZD0530 reversibly inhibits the formation and activity of human osteoclasts
.
Mol Cancer Res
2009
;
7
:
476
88
.
41
Zhang
XH
,
Wang
Q
,
Gerald
W
, et al
. 
Latent bone metastasis in breast cancer tied to Src-dependent survival signals
.
Cancer Cell
2009
;
16
:
67
78
.
42
Hannon
RA
,
Clack
G
,
Rimmer
M
, et al
. 
Effects of the Src kinase inhibitor saracatinib (AZD0530) on bone turnover in healthy men: a randomized, double-blind, placebo-controlled, multiple ascending dose phase I trial
.
J Bone Miner Res
2010
;
25
:
463
71
.
43
Koreckij
T
,
Nguyen
H
,
Brown
LG
,
Yu
EY
,
Vessella
RL
,
Corey
E
. 
Dasatinib inhibits the growth of prostate cancer in bone and provides additional protection from osteolysis
.
Br J Cancer
2009
;
101
:
263
8
.
44
Frame
MC
. 
Src in cancer: deregulation and consequences for cell behaviour
.
Biochim Biophys Acta
2002
;
1602
:
114
30
.
45
Myoui
A
,
Nishimura
R
,
Williams
PJ
, et al
. 
C-SRC tyrosine kinase activity is associated with tumor colonization in bone and lung in an animal model of human breast cancer metastasis
.
Cancer Res
2003
;
63
:
5028
33
.
46
Wu
Z
,
Chang
P-C
,
Yang
JC
, et al
. 
Autophagy blockade sensitizes prostate cancer cells towards Src family kinase inhibitors
.
Genes Cancer
2010
;
1
:
40
9
.
47
Suh
J
,
Rabson
AB
. 
NF-κB activation in human prostate cancer: important mediator or epiphenomenon?
J Cell Biochem
2004
;
91
:
100
17
.