Purpose: Current evidence indicates that an osteoblast lesion in prostate cancer is preceded by osteolysis. Thus, prevention of osteolysis would reduce complications of bone metastasis. Bone marrow–derived mesenchymal stem cells have the ability to differentiate into osteoblast and produce osteoprotegerin, a decoy receptor for the receptor activator for nuclear factor κB ligand, naturally. The present study examined the potential of unmodified mesenchymal stem cells to prevent osteolytic bone lesions in a preclinical mouse model of prostate cancer.

Experimental Design: The human prostate cancer cell line PC3 was implanted in tibiae of severe combined immunodeficient mice. After establishment of the tumor, either unmodified or genetically engineered mesenchymal stem cells overexpressing osteoprotegerin was injected at the site of tumor growth. The effects of therapy were monitored by bioluminescence imaging, micro–computed tomography, immunohistochemistry, and histomorphometry.

Results: Data indicated significant (P < 0.001) inhibition of tumor growth and restoration of bone in mice treated with unmodified and modified mesenchymal stem cells. Detailed analysis suggested that the donor mesenchymal stem cell inhibited tumor progression by producing woven bone around the growing tumor cells in the tibiae and by preventing osteoclastogenesis.

Conclusions: Overcoming the limitation of the number of mesenchymal stem cells available in the bone can provide significant amelioration for osteolytic damage without further modification. (Clin Cancer Res 2009;15(23):7175–85)

Translational Relevance

Osteolytic bone damage is major cause of morbidity in several cancers. Because of the refractory nature of metastatic tumors in the bone, conventional chemotherapy combinations and radiation therapy fail to provide long-term cure. Furthermore, even the modes effects offered by these therapies fail to restore bone destruction. Thus, new therapies are needed for the management of osteolytic bone damage in cancer patients. Results of the present study show that, without any genetic modification, adult mesenchymal stem cells are capable of inducing bone formation in response to cancer-associated bone loss. Because mesenchymal stem cells are immune privileged and their use in allogenic context has entered human clinical trials, results of this preclinical study is greatly poised as a potential alternative for cancer-induced bone damage not only for prostate cancer bone metastasis, which initiates with osteolytic events, but also other cancers such carcinomas of the breast, thyroid, lung, kidney, and myeloma.

Prostate cancer is the second leading cause of cancer deaths in men behind lung cancer in the United States and metastasizes to bone in >70% of the cases during advance stages (1). Bone metastasis causes severe bone pain and pathologic fractures, and shortens life span by significant amount. Most bone metastatic cancers (breast, lung, thyroid, and kidney) generate osteolytic lesions, whereas prostate cancer generates osteoblastic phenotype, with an overall increase in bone volume (24). However, the appearance of osteoblastic lesions is preceded and/or accompanied by an osteolytic event, which is required for the establishment and growth of prostate cancer cells in the bone microenvironment (4, 5). The binding of receptor activator of nuclear factor κB ligand (RANKL) to RANK on preosteoclasts or osteoclasts is essential for their maturation and activity (6, 7). Increased expression of RANKL has been observed in osteolytic malignancies, and the inhibition of osteoclastogenesis or metastasis has been considered as an intervention strategy. Osteoprotegerin is a soluble decoy receptor for RANKL and prevents binding of RANKL to RANK, leading to the inhibition of osteoclast activity and bone metastasis (810). Osteoprotegerin therefore promises tremendous hope for potential clinical use in the management of osteolytic bone metastasis. Systemic delivery of osteoprotegerin has shown promise as a potential therapy in animal models, limiting hypercalcemia and osteolysis induced by myeloma, breast, lung, or prostate cancer, and reducing tumor establishment in bone (1117).

Homing of adult bone marrow derived mesenchymal stem cells to the sites of tumor growth is well known besides their ability to self-renew and differentiate into bone, cartilage, fat, and of other tissue types (18). Systemic administration of mesenchymal stem cells in mice has been shown to engraft within the tumor microenvironment in many cancers and thus represent an attractive cellular vehicle for cell therapy and gene therapy (19, 20). Because osteoprotegerin is constitutively produced by mesenchymal stem cells, we speculated that lack of mesenchymal stem cells in sufficient quantities in the bone microenvironment is the reason for the inability to inhibit excess osteoclastogenesis and compensate for bone loss, and overcoming this limitation would provide therapy for osteolytic bone damage. The present study determined the potential of mesenchymal stem cells that were unmodified as compared with that genetically engineered to overexpress osteoprotegerin in bone remodeling following osteolytic damage. Results indicated that naïve mesenchymal stem cells inhibited tumor growth comparable with mesenchymal stem cells overexpressing osteoprotegerin by formation of new bone around the tumor cells and by inhibiting osteoclast activation.

Cell lines and reagents

Osteolytic prostate cancer cell line PC3 expressing firefly luciferase was a generous gift from Dr. Kenneth J. Pienta (University of Michigan) and maintained in RPMI-1640 medium (Mediatech, Inc.) supplemented with 10% fetal bovine serum (Mediatech, Inc.) and penicillin/streptomycin (Mediatech, Inc.). Osteoblastic prostate cancer cell line C4-2B was a generous gift from Dr. Marco G. Cecchini, Departments of Urology and Clinical Research, University of Bern, and maintained in T-medium (Invitrogen) supplemented with 10% fetal bovine serum and antibiotics. RAW 264.7 cells were obtained from the American Type Culture Collection and maintained in α-MEM supplemented with 10% fetal bovine serum, 4 mmol/L l-glutamine and antibiotics. Mesenchymal stem cells were maintained in Stem Line medium (Sigma-Aldrich Corp.) supplemented with 10% fetal bovine serum, 4 mmol/L l-glutamine and penicillin/streptomycin. HEK293 cells were purchased from American Type Culture Collection and maintained in DMEM supplemented with 10% newborn calf serum and penicillin/streptomycin. Isolation, purification, and differentiation of mouse mesenchymal stem cells from C57BL/6 mice were carried out as described recently (21). Antibodies for cytokeratin-18 and green fluorescent protein (GFP) were purchased from Abcam Ltd. Secondary immunodetection was done using anti-rat and/or anti-rabbit ABC kits purchased from Vector Laboratories. Total RNA was isolated using Trizol (Invitrogen) and purified using a Qiagen mini kit. iScript cDNA synthesis kit was purchased from Bio-Rad. Primers for reverse transcriptase-PCR analysis were designed using the Primer 3 (version 4.0) software, and oligonucleotides were purchased from Integrated DNA Technologies, Inc. cDNA samples were analyzed in Bio-Rad iCycler (Hercules). Three-dimensional PC3 beads were prepared and supplied by Vivo Biosciences. Proliferation of PC3 cells were determined by Vybrant MTT Cell Proliferation Assay Kit (Molecular Probes, Inc.), as recommended by the manufacturer. Lentivirus encoding short-hairpin RNA (shRNA) constructs for silencing osteoprotegerin were designed and supplied by Sigma-Aldrich Corp. Alkaline phosphatase enzyme activity was measured using a commercial kit (Sigma-Aldrich Corp.), following manufacturer's instructions. Von Kossa staining was done to detect calcium deposition following standard protocols (22).

Construction of recombinant plasmid and expression analysis

The recombinant osteoprotegerin used in this study comprised the ligand-binding domain of human osteoprotegerin (1-201 amino acids) fused to the Fc domain of human IgG. The osteoprotegerin Fc fusion gene was isolated from an adenoviral construct (kindly provided by Dr. Joanne Douglas, University of Alabama at Birmingham) and subcloned into adeno-associated virus (AAV) plasmid under the control of cytomegalovirus/chicken β-actin promoter. Expression of osteoprotegerin Fc as a secreted protein from the AAV plasmid was confirmed by transient transfection into HEK293 cells using Lipofectamine-2000 reagent (Invitrogen, Inc.) and testing the supernatants on SDS-PAGE using a mouse monoclonal antibody against human osteoprotegerin (Chemicon, Inc.).

Osteoclast assay

Mesenchymal stem cells were cultured for 2 d at confluency when conditioned media was collected and centrifuged at 4,000 rpm to pellet any floating cells, and the supernatant was stored at −80°C. RAW 264.7 cells were cultured in six-well culture dishes with 25 ng/mL of RANKL (SIGMA-Aldrich), either in regular medium or mesenchymal stem cells conditioned medium for 8 d. The culture medium was replaced every alternate day. After 8 d, cells were stained for tartrate-resistant acid phosphatase to determine the effect of mesenchymal stem cells conditioned media on osteoclast formation using a leukocyte acid phosphatase kit (SIGMA-Aldrich).

Development of osteolytic bone metastasis model in the mouse

Six-week-old male severe combined immunodeficient (SCID) mice were purchased from the National Cancer Institute–Frederick Animal Production Facility. Maintenance of the animals was carried out following guidelines of the Institutional Animal Care and Use Committee, and all experimental procedures were approved by the Institutional Animal Care and Use Committee and the Occupational Health and Safety Department of the University of Alabama at Birmingham. The mice were acclimatized for a week, following which 105 osteolytic PC3 prostate cancer cells, constitutively expressing luciferase, were implanted in the tibia of right leg (n = 30) in 20 μL PBS. The left tibia served as control and was injected with only PBS. A 3/10-mL (28 gauge) insulin syringe (BD Biosciences) was used for the intratibial injection of the prostate cancer cells under isoflurane anesthesia.

Bioluminescence imaging

In vivo bioluminescence imaging was conducted in a cryogenically cooled IVIS-100 system (Xenogen Corp.) to detect luciferase expression using living imaging acquisition and analysis software (Xenogen Corp.), as described (11). For each animal, bioluminescence imaging was done before and 4 wk after the initiation of mesenchymal stem cells treatment. The intensity of light emission was represented with a pseudo–color scaling of bioluminescent images. The bioluminescent images were overlaid on black-and-white photographs of the mice collected at the same time. Bioluminescence units were converted as counts per second for each animal, and final counts were divided by the initial counts and were plotted graphically as a measure of tumor growth.

Micro–computed tomography analysis

Superficial computed tomography scanning of whole skeleton was done on live animals using MicroCAT II (Imtek, Inc.). For the determination of the three-dimensional architecture of the trabecular and cortical bones, mice were sacrificed, and tibia were harvested and analyzed in an advanced micro–computed tomography instrument (μCT 40, Scanco Medical AG). Two scans were done on each tibia, one for whole tibial bone with 16-μm resolution and one for trabecular analysis with a 6-μm resolution. For the whole tibia, the scan was composed of 1,129 slices, with a threshold value of 265. A three-dimensional reconstruction of the images was done with the region of interest consisting of trabecular and cortical areas. The scan of the trabecular bone was done below the growth plate. Each scan consisted of 209 slices, of which 100 were used for the analysis. Regions of interest were drawn on each of the 100 slices, just inside the cortical bone, to include only the trabecular bone and the marrow. Trabecular bone was set to a threshold at 327 to distinguish it from the marrow. The three-dimensional reconstruction was done on the region of interest, which only contained trabecular bone; no cortical bone was present in these regions of interest.

Bone histomorphometry

Soft tissues were fixed in 10% neutral buffered-formalin solution for 48 h before embedding in paraffin for histologic analysis. Bone tissues were decalcified in 0.5 mol/L EDTA in Ca2+- and Mg2+-free Dulbecco's PBS (Cellgro) before embedding in paraffin. Six-micrometer longitudinal serial sections were cut from the femur and tibia, and stained with hematoxylin and eosin or Goldner's trichrome stain to determine the characteristics of tumor growth in the bone and the extent of osteolysis in response to different treatments. Tartrate-resistant acid phosphatase staining was done on bone sections to determine osteoclast activity. Quantitative osteomeasurements of bones were done using an Olympus BX51 microscope and Bioqaunt Image analysis Software (R&M Biometrics).

Biomechanical testing

Mice were sacrificed 4 wk after the mesenchymal stem cells treatments, and tibiae were collected and fresh frozen. Specimens were tested to failure by three-point bending on 858 MiniBionix Materials Testing System (MTS Systems). Stiffness and peak load were calculated from the force displacement data.

Immunohistochemistry

Briefly, 6-mm paraffin sections of tibiae were deparaffinized in xylene and hydrated through graded alcohol. Antigen retrieval was done in citrate buffer (pH 6.0), under steam for 20 min. Sections were cooled to room temperature, and endogenous peroxidase was removed using 0.3% H2O2 in methanol for 30 min and blocked with 3% goat serum for 30 min. Tissue sections were then incubated with primary antibodies overnight at 4°C. Sections were washed in Phosphate-buffered saline (PBST) with 0.05% Tween-20 and again incubated at room temperature with biotin-conjugated goat anti-rabbit/anti-rat secondary antibody for 2 h. After washing, sections were incubated with streptavidin-conjugated horseradish peroxidase for 1 h at room temperature. After another wash with PBST, immunodetection was done using 3,3′-diaminobenzidine-H2O2 (Vector Labs) and counterstained with hematoxylin.

Osteoprotegerin ELISA

For determination of osteoprotegerin levels secreted by the PC3 cells, mesenchymal stem cells and mesenchymal stem cells–osteoprotegerin cells were cultured separately for 72 h; cell numbers were counted, and 100 μL of culture media was subjected to ELISA (ALPCO Diagnostics), following manufacturer's instructions. Each sample was analyzed in triplicate, and the absorbance was measured in a microplate reader (BioTek Instruments, Inc.).

Statistical analysis

Data were analyzed by one-way ANOVA. A Tukey test was also applied for multiple comparisons wherever applicable. Values provided are the mean ± SE, and the differences were considered significant if P < 0.05.

Production of osteoprotegerin by unmodified mesenchymal stem cells and inhibition of osteoclastogenesis

Total RNA was isolated from mouse mesenchymal stem cells, converted to cDNA, and subjected to real-time PCR analysis. The result indicated the expression of osteoprotegerin mRNA by the mesenchymal stem cells (Fig. 1A). Osteoprotegerin immunostaining was also done on cultured mesenchymal stem cells, which clearly indicated production of osteoprotegerin by the mesenchymal stem cells (Fig. 1B). Ability of mesenchymal stem cells to inhibit osteoclastogenesis was tested in vitro by culturing preosteoclast RAW 264.7 cells in regular medium or mesenchymal stem cells–conditioned medium in the presence of RANKL for 7 days. Tartrate-resistant acid phosphatase staining indicated significant number of osteoclasts in RAW cells cultured in regular medium comparable with RAW cells grown in mesenchymal stem cells conditioned medium (Fig. 1C). Levels of osteoprotegerin produced in culture as protein secreted by PC3 cells, mesenchymal stem cells, and mesenchymal stem cells genetically engineered to overexpress osteoprotegerin are given in Supplementary Fig. S4.

Fig. 1.

Expression of osteoprotegerin by mesenchymal stem cells and its effects on osteoclast formation in vitro. A, reverse transcriptase-PCR analysis showing osteoprotegerin mRNA expression in unmodified mouse mesenchymal stem cells. B, immunocytochemical localization of osteoprotegerin in cultured mouse mesenchymal stem cells. C, preosteoclast RAW cells were cultured in mesenchymal stem cells conditioned medium in the presence of RANKL for 7 d. Tartrate-resistant acid phosphatase staining indicated inhibition of osteoclastogenesis in RAW cells grown in mesenchymal stem cells conditioned medium compared with RAW cells grown in regular medium in the presence of RANKL. Conditioned medium from osteoprotegerin-silenced mesenchymal stem cells (osteoprotegerin–knockout (KO)–mesenchymal stem cells) failed to prevent osteoclast formation. TRAP, tartrate-resistant acid phosphatase; RM, regular media; CM, conditioned media. D, control mesenchymal stem cells and osteoprotegerin–KO–mesenchymal stem cells were tested for differentiation into osteoblast lineage using osteoblast medium for 2 wk. Von Kossa staining was done to detect calcium deposits (black) to confirm that osteoblast lineage differentiation is compromised in osteoprotegerin–KO–mesenchymal stem cells (right) as compared with unmodified mesenchymal stem cells (middle). There was no positive staining in mesenchymal stem cells culture without the osteoblast medium (left).

Fig. 1.

Expression of osteoprotegerin by mesenchymal stem cells and its effects on osteoclast formation in vitro. A, reverse transcriptase-PCR analysis showing osteoprotegerin mRNA expression in unmodified mouse mesenchymal stem cells. B, immunocytochemical localization of osteoprotegerin in cultured mouse mesenchymal stem cells. C, preosteoclast RAW cells were cultured in mesenchymal stem cells conditioned medium in the presence of RANKL for 7 d. Tartrate-resistant acid phosphatase staining indicated inhibition of osteoclastogenesis in RAW cells grown in mesenchymal stem cells conditioned medium compared with RAW cells grown in regular medium in the presence of RANKL. Conditioned medium from osteoprotegerin-silenced mesenchymal stem cells (osteoprotegerin–knockout (KO)–mesenchymal stem cells) failed to prevent osteoclast formation. TRAP, tartrate-resistant acid phosphatase; RM, regular media; CM, conditioned media. D, control mesenchymal stem cells and osteoprotegerin–KO–mesenchymal stem cells were tested for differentiation into osteoblast lineage using osteoblast medium for 2 wk. Von Kossa staining was done to detect calcium deposits (black) to confirm that osteoblast lineage differentiation is compromised in osteoprotegerin–KO–mesenchymal stem cells (right) as compared with unmodified mesenchymal stem cells (middle). There was no positive staining in mesenchymal stem cells culture without the osteoblast medium (left).

Close modal

Inhibition of tumor growth by mesenchymal stem cells

To determine the effect of mesenchymal stem cells in preventing the growth of prostate tumor in the bone, 6-week-old male SCID mice were injected intratibially with 105 osteolytic human prostate cancer cells, PC3, expressing firefly luciferase. The next day, 5 × 105 bone marrow–derived mouse mesenchymal stem cells, which were unmodified or overexpressing osteoprotegerin (mesenchymal stem cells–osteoprotegerin) were injected in the tibia in proximity to the tumor cells. Growth of prostate tumor in the bone was evaluated by bioluminescence imaging 4 weeks after the administration of the mesenchymal stem cells, which indicated almost 90% inhibition of tumor growth in both treatment groups (Fig. 2A and B). Mesenchymal stem cells–osteoprotegerin did not show any advantage over unmodified mesenchymal stem cells in preventing tumor progression.

Fig. 2.

Tumor growth following mesenchymal stem cell therapy. noninvasive total body imaging was done on the day of intratibial injection of PC3 cells (day 0) and 4 wk after the intratibial administration of the mesenchymal stem cells. A, mice on the left are the same mice that are on the right, and they maintain the same order of alignment. B, quantitative analysis of luciferase expression as a measure of tumor growth 4 wk after the treatment with mesenchymal stem cells or mesenchymal stem cells modified to overexpress osteoprotegerin. **, P < 0.001. C, when tumor cells were allowed to grow for 2 wk, followed by administration of mesenchymal stem cells, therapeutic benefits are apparent but not statistically significant (P > 0.05).

Fig. 2.

Tumor growth following mesenchymal stem cell therapy. noninvasive total body imaging was done on the day of intratibial injection of PC3 cells (day 0) and 4 wk after the intratibial administration of the mesenchymal stem cells. A, mice on the left are the same mice that are on the right, and they maintain the same order of alignment. B, quantitative analysis of luciferase expression as a measure of tumor growth 4 wk after the treatment with mesenchymal stem cells or mesenchymal stem cells modified to overexpress osteoprotegerin. **, P < 0.001. C, when tumor cells were allowed to grow for 2 wk, followed by administration of mesenchymal stem cells, therapeutic benefits are apparent but not statistically significant (P > 0.05).

Close modal

Mesenchymal stem cell therapy is ineffective for end-stage disease

To test if therapy with mesenchymal stem cells is effective after the tumor has established in the bone microenvironment with high degree of osteolytic damage, PC3 cells were injected in the tibia and allowed to grow for 2 weeks. Then, 5 × 105 mesenchymal stem cells (unmodified or overexpressing osteoprotegerin) were injected in the same location. Tumor progression was evaluated 4 weeks after the treatment, and bioluminescence imaging indicated inhibition of tumor growth in some of the treated mice compared with the untreated ones, although data was not significantly different (P > 0.05) between treated and untreated animals when all the mice were taken into consideration for comparison (Fig. 2C). Similar observations were made when histologic architecture of the tibia was studied for bone loss due to mesenchymal stem cell therapy (data not shown). These data suggest requirement of optimal number of mesenchymal stem cells to prevent osteolysis in prostate cancer bone metastasis at an earlier time.

Effect of mesenchymal stem cell therapy in bone remodeling

Micro–computed tomography of the skeleton showed significant loss of bone in the region of implantation of the PC3 cells, whereas complete restoration was observed in the tibiae treated with mesenchymal stem cells and mesenchymal stem cells–osteoprotegerin (Fig. 3A). Tibiae were harvested from the mice, and studies were done to understand the ultrastructure of the tibia. Three-dimensional micro–computed tomography data indicated a significant decline in relative bone volume and trabecular connectivity density in untreated mice compared with age-matched normal mice. Trabecular and cortical bone structures were completely restored in mice with PC3 cells in the tibia by the mesenchymal stem cell therapy (Fig. 3B). It is interesting to note that the relative bone volume and trabecular connectivity density in the treated mice significantly exceeded that observed in the tibiae of normal mice, indicating the effectiveness of the therapy. Bone restoration was highest in mice treated with mesenchymal stem cells–osteoprotegerin, which may be because of the significant inhibition of osteoclastogenesis due to overproduction of osteoprotegerin as compared with unmodified mesenchymal stem cell–treated mice (Fig. 3B; Supplementary Fig. S1A and B). In mesenchymal stem cells–osteoprotegerin–treated mice, the restoration of tibial bone resulted in limitation of marrow space and might compromise important event(s) such as hematopoiesis and hence was excluded in further experiments. Histomorphometry supported the results obtained from bioluminescence imaging and micro–computed tomography analysis. Tartrate-resistant acid phosphatase staining revealed highest number of osteoclasts in the untreated tibia, mainly at the tumor-bone interface, whereas the number and size of osteoclasts were significantly decreased in the tibia of the treated mice. The mesenchymal stem cells– and mesenchymal stem cells–osteoprotegerin–treated mice indicated similar pattern of osteoclast staining (Fig. 3C). Three-point mechanical testing of the tibial bone was done to determine the mechanical strength after treatment. Data indicated similar bone strength in mesenchymal stem cell–treated mice compared with normal mice tibia (Supplementary Fig. S2A and B).

Fig. 3.

Histomorphometric analysis of bone. A, three-dimensional scanning micro–computed tomography of the mouse skeleton showing restoration of tibia following mesenchymal stem cell therapy compared with untreated mice. B, three-dimensional transmission micro–computed tomography of the bone showing significant osteolysis in the tibia due to the growth of PC3 cells, whereas therapy with mesenchymal stem cells and mesenchymal stem cells overexpressing osteoprotegerin prevented osteolysis and reduced tumor burden significantly. When compared with normal tibia, both treated groups showed higher relative bone volume and trabecular bone density. Mesenchymal stem cells overexpressing osteoprotegerin–treated mice showed the highest bone volume and trabecular density. This is likely due to higher inhibition of osteoclastogenesis. Sections of tibia stained with Goldner's trichrome stain, wherein mineralized bone stains blue green, are at the bottom. Original magnification, ×25. C, reduction of osteoclast activity following treatment as determined by tartrate-resistant acid phosphatase staining. Mesenchymal stem cells and mesenchymal stem cells overexpressing osteoprotegerin showed significantly less osteoclast activity at the tumor-bone interface (arrowheads) as compared with untreated mice. Original magnification, ×200.

Fig. 3.

Histomorphometric analysis of bone. A, three-dimensional scanning micro–computed tomography of the mouse skeleton showing restoration of tibia following mesenchymal stem cell therapy compared with untreated mice. B, three-dimensional transmission micro–computed tomography of the bone showing significant osteolysis in the tibia due to the growth of PC3 cells, whereas therapy with mesenchymal stem cells and mesenchymal stem cells overexpressing osteoprotegerin prevented osteolysis and reduced tumor burden significantly. When compared with normal tibia, both treated groups showed higher relative bone volume and trabecular bone density. Mesenchymal stem cells overexpressing osteoprotegerin–treated mice showed the highest bone volume and trabecular density. This is likely due to higher inhibition of osteoclastogenesis. Sections of tibia stained with Goldner's trichrome stain, wherein mineralized bone stains blue green, are at the bottom. Original magnification, ×25. C, reduction of osteoclast activity following treatment as determined by tartrate-resistant acid phosphatase staining. Mesenchymal stem cells and mesenchymal stem cells overexpressing osteoprotegerin showed significantly less osteoclast activity at the tumor-bone interface (arrowheads) as compared with untreated mice. Original magnification, ×200.

Close modal

Interaction between PC3 and mesenchymal stem cells in vivo

Outcome of the previous experiment suggests that the therapeutic effects of mesenchymal stem cells are not highly apparent when administered at the advanced stages of tumor-induced osteolytic bone lesion. It is likely that, at a later stage, the number of tumor cells in the tibia outnumbered the input mesenchymal stem cells. Mesenchymal stem cell therapy did not influence the prostate tumor growth in the bone in a negative manner, even at later stages. This prompted us to study the interaction between the mesenchymal stem cells and the PC3 cells in the bone in vivo. PC3 cells (105) were injected in the tibia and allowed to grow for 1 week for detectable tumors when 5 × 105 mesenchymal stem cells were injected in the same site. Mesenchymal stem cells used here were derived from a GFP transgenic mouse. Mice were sacrificed 1 week after the injection of mesenchymal stem cells, and tibiae were harvested and subjected to histomorphometry. Results showed formation of new bone surrounding the tumor nests (Fig. 4A). When analyzed under polarized light, this newly formed bone comprised of randomly oriented collagen fibers, called woven bone and characteristic to fracture healing and prostate cancer bone metastasis in humans (Fig. 4A and B). Human epithelial cell marker cytokeratin-18 immunostaining was done to identify the prostate tumors in the bone (Fig. 4C, c), which also exposed a multiple layers of fibroblast-like cells arranged in concentric circles, separating the tumor cells from the newly formed bone, which also negatively stained for cytokeratin-18. The outermost layer of these fibroblast-like cells was often seen to be embedded or being transformed into the newly formed bone (Fig. 4C, a and c). These cells stained positively for GFP, which indicated new bone formation in the treated mice formed predominantly from exogenously administered mesenchymal stem cells (Fig. 4C, d). Formation of new bone around the tumor cells resulted in restricting the growth of prostate tumor cells in the tibia. No such woven bone formation was noticed in tibia injected with mesenchymal stem cells in the absence of PC3 cells, suggesting bone formation in the PC3 injected tibia is triggered by the prostate tumor cells (Fig. 4A). Reverse transcriptase-PCR analysis of mRNA isolated from mesenchymal stem cells obtained from a coculture experiment with PC3 cells for 10 days showed no significant upregulation of osteogenic genes in the mesenchymal stem cells, indicating that the differentiation of mesenchymal stem cells toward an osteoblastic lineage may have been driven by osteolysis because of enhanced osteoclastogenesis (Fig. 5A and B). In fact, tartrate-resistant acid phosphatase staining indicated intense osteoclast activity at the tumor-bone interface (Fig. 4C, b).

Fig. 4.

Mechanism of tumor inhibition following implantation of mesenchymal stem cells in tibiae with PC3 tumors. A, histomorphology of tibia showing presence or absence of new bone formation surrounding tumor nests in mice tibiae, following implantation of unmodified mesenchymal stem cells only or following PC3 tumor cell implantation. Polarized light microscopy showing the newly formed bone, composed of randomly oriented, mineralized collagen fibers (woven bone). Original magnification, ×100. When mesenchymal stem cells were implanted into a normal tibia without the tumor cells, no such bone formation was observed (far left). When PC3 cells were injected in the tibia followed by the implantation of mesenchymal stem cells (osteoprotegerin silenced), similarly no significant bone formation (far right) was observed, suggesting the requirement of osteoprotegerin for in vivo bone formation. B, graph showing the amount of woven bone formed in the tibia after treatment with mesenchymal stem cells, osteoprotegerin–KO–mesenchymal stem cells, or mesenchymal stem cells–osteoprotegerin. Osteoprotegerin–KO–mesenchymal stem cells resulted in least amount of woven bone, indicating a requirement for simultaneous inhibition of osteoclastogenesis while mesenchymal stem cells differentiate into bone.*, P < 0.001. C, hematoxylin and eosin staining of the tibia showing spindle-like cells of mesenchymal origin bordering the tumor and the new bone (a). Significant osteoclast activity was noticed by tartrate-resistant acid phosphatase staining at the tumor-bone interface, most likely serving as the initiating factor for the mesenchymal stem cells differentiation into osteoblasts (b). Immunostaining with the human epithelial marker cytokeratin-18 indicated tumor nests surrounded by the mesenchymal stem cells (c). Staining with GFP antibody confirmed that differentiating mesenchymal stem cells are of donor origin (d). Original magnification, ×200.

Fig. 4.

Mechanism of tumor inhibition following implantation of mesenchymal stem cells in tibiae with PC3 tumors. A, histomorphology of tibia showing presence or absence of new bone formation surrounding tumor nests in mice tibiae, following implantation of unmodified mesenchymal stem cells only or following PC3 tumor cell implantation. Polarized light microscopy showing the newly formed bone, composed of randomly oriented, mineralized collagen fibers (woven bone). Original magnification, ×100. When mesenchymal stem cells were implanted into a normal tibia without the tumor cells, no such bone formation was observed (far left). When PC3 cells were injected in the tibia followed by the implantation of mesenchymal stem cells (osteoprotegerin silenced), similarly no significant bone formation (far right) was observed, suggesting the requirement of osteoprotegerin for in vivo bone formation. B, graph showing the amount of woven bone formed in the tibia after treatment with mesenchymal stem cells, osteoprotegerin–KO–mesenchymal stem cells, or mesenchymal stem cells–osteoprotegerin. Osteoprotegerin–KO–mesenchymal stem cells resulted in least amount of woven bone, indicating a requirement for simultaneous inhibition of osteoclastogenesis while mesenchymal stem cells differentiate into bone.*, P < 0.001. C, hematoxylin and eosin staining of the tibia showing spindle-like cells of mesenchymal origin bordering the tumor and the new bone (a). Significant osteoclast activity was noticed by tartrate-resistant acid phosphatase staining at the tumor-bone interface, most likely serving as the initiating factor for the mesenchymal stem cells differentiation into osteoblasts (b). Immunostaining with the human epithelial marker cytokeratin-18 indicated tumor nests surrounded by the mesenchymal stem cells (c). Staining with GFP antibody confirmed that differentiating mesenchymal stem cells are of donor origin (d). Original magnification, ×200.

Close modal
Fig. 5.

Expression of osteogenic genes. A, mesenchymal stem cells were cultured for 10 d in either regular medium or PC3-conditioned medium. Total RNA was isolated, converted to cDNA, and analyzed for upregulation of osteogenic genes. Data showing no significant change in osteoblastic lineage differentiation after mesenchymal stem cells were cultured in conditioned media obtained from PC3 cells. BSP, bone sialoprotein; ALP, alkaline phosphatase; Runx2, runt-related transcription factor; OC, osteocalcin; OP, osteopontin. B, mesenchymal stem cells cultured in regular medium or PC3-conditioned medium showing equivalent alkaline phosphatase activity, indicating that PC3 cells did not initiate osteoblastic differentiation in the mesenchymal stem cells directly. C, PC3–mesenchymal stem cells in vitro coculture assay. PC3 cells were grown on hu-biogel matrix as three-dimensional spheroids and cultured in a 0.8-μm pore size transwell plate, along with mesenchymal stem cells in the lower chamber. After 72 h, the PC3 beads were collected and analyzed by MTT assay for cell proliferation. Data presented here are mean ± SE (n = 12 for each experimental conditions).

Fig. 5.

Expression of osteogenic genes. A, mesenchymal stem cells were cultured for 10 d in either regular medium or PC3-conditioned medium. Total RNA was isolated, converted to cDNA, and analyzed for upregulation of osteogenic genes. Data showing no significant change in osteoblastic lineage differentiation after mesenchymal stem cells were cultured in conditioned media obtained from PC3 cells. BSP, bone sialoprotein; ALP, alkaline phosphatase; Runx2, runt-related transcription factor; OC, osteocalcin; OP, osteopontin. B, mesenchymal stem cells cultured in regular medium or PC3-conditioned medium showing equivalent alkaline phosphatase activity, indicating that PC3 cells did not initiate osteoblastic differentiation in the mesenchymal stem cells directly. C, PC3–mesenchymal stem cells in vitro coculture assay. PC3 cells were grown on hu-biogel matrix as three-dimensional spheroids and cultured in a 0.8-μm pore size transwell plate, along with mesenchymal stem cells in the lower chamber. After 72 h, the PC3 beads were collected and analyzed by MTT assay for cell proliferation. Data presented here are mean ± SE (n = 12 for each experimental conditions).

Close modal

Characterization of osteoblastic phenotype in prostate cancer bone metastasis

We compared the growth kinetics of highly osteolytic PC3 cells and the osteoblastic C4-2B cells in the tibia of SCID mice. The rate of tumor progression was significantly lower in the tibia injected with C4-2B cells as compared with PC3 cells (Fig. 6A and B). Although PC3 cells induced severe osteolysis by 1 month after inoculation, C4-2B cells produced osteoblastic events by 2 months after administration. There was a gradual switch from osteoblastic to osteolytic phenotype when C4-2B cells were allowed to grow for 6 months in the tibia (Fig. 6C). These observations indicate that the growth kinetics of the cancer cells in the bone might be the determining factor favoring osteoblastic or osteolytic outcome.

Fig. 6.

A, growth kinetics of osteoblastic C4-2B cells in the tibia of SCID mice. B, growth kinetics of PC3 cells in the tibia of SCID mice. C, C4-2B–injected tibia showing osteolytic lesions when allowed to grow for 6 mo.

Fig. 6.

A, growth kinetics of osteoblastic C4-2B cells in the tibia of SCID mice. B, growth kinetics of PC3 cells in the tibia of SCID mice. C, C4-2B–injected tibia showing osteolytic lesions when allowed to grow for 6 mo.

Close modal

Role of mesenchymal stem cell–produced osteoprotegerin in bone formation

To test the significance of osteoprotegerin in this process, a mesenchymal stem cell line was generated wherein osteoprotegerin expression was silenced using a lentivirus producing shRNA targeting (Supplementary Fig. S3). Osteoprotegerin-silenced mesenchymal stem cells failed to differentiate into osteoblast in vitro, as determined by von Kossa staining (Fig. 1D), and no significant new bone formation was observed in the tibia when these mesenchymal stem cells were tested against PC3 cells in a similar experiment mentioned in the previous section (Fig. 4A). Moreover, conditioned medium from osteoprotegerin-silenced mesenchymal stem cells failed to inhibit osteoclastogenesis when RAW cells were cultured for 8 days in the presence of RANKL (Fig. 1C), suggesting the requirement of osteoprotegerin for osteoblast differentiation and inhibition of osteoclastogenesis.

Effects of bone marrow microenvironment on the growth of PC3 cells

To test the effects of mesenchymal stem cells on the prostate cancer cells, PC3 cells were cocultured in a three-dimensional matrix with mesenchymal stem cells in the presence or absence of bone marrow conditioned medium for 3 days. MTT assay was done to determine cell proliferation. Addition of naïve mesenchymal stem cells to the coculture did not alter PC3 cell proliferation compared with PC3 cultured only in bone marrow conditioned medium. An increase in cell proliferation was noted only when PC3 cells were cultured in bone marrow conditioned media along with mesenchymal stem cells overexpressing osteoprotegerin (P < 0.001; Fig. 5C), supporting that osteoprotegerin is also a survival factor for the prostate cancer cells (23). This suggests that the inhibitory effect of mesenchymal stem cells on the growth of prostate cancer in vivo is an indirect effect and is mediated by the inhibition of osteoclastogenesis and differentiation into osteoblasts.

Results of the present study indicate the therapeutic potential of unmodified mesenchymal stem cells in inhibiting the growth of prostate tumor in the bone and prevention of bone loss. Mesenchymal stem cells did not induce direct apoptosis of tumor cells, instead the inhibition of tumor growth in the bone was mediated by new bone formation around them. Although significant therapeutic advantage can be obtained in osteolytic bone metastasis using this approach, absence of a direct killing mechanism may help the metastasis re-establish itself in the course of time. The beneficial effects of mesenchymal stem cells can thus be further amplified by modifying them ex vivo to express tumoricidal genes besides retaining their ability to differentiate into bone. A recent study used mesenchymal stem cells expressing urokinase-type plasminogen antagonist amino-terminal fragment and showed inhibition of tumor growth by inhibiting angiogenesis prevented bone loss (24). Most studies indicated that mesenchymal stem cells promote tumor growth by participating in tumor stroma formation and establishment of premetastatic niche. Our in vitro studies showed an increase in proliferation rate of prostate cancer cells only when cocultured with the mesenchymal stem cells overexpressing osteoprotegerin. However, the same mesenchymal stem cells overexpressing osteoprotegerin tested provided a therapeutic effect in vivo. Hence, the observed in vitro effects could be attributed only to secretory proteins in the coculture system and may not include other events in the tumor microenvironment. Mesenchymal stem cells inhibited the growth of prostate tumor in the bone in vivo by laying down new bone around the cancer cells, which slowed their rapid growth. Other factors such as cell contact in vivo may have also played important roles in addition to the effects of osteoprotegerin in increasing osteogenesis.

Although the difference in tumor volume between unmodified mesenchymal stem cells (mesenchymal stem cells–GFP) and mesenchymal stem cells overexpressing osteoprotegerin was not statistically significant, there was an observable difference in the mesenchymal stem cells–GFP group, which showed less tumor growth compared with mesenchymal stem cells–osteoprotegerin group. Because osteoprotegerin may serve as a survival factor for tumor cells, this observation also indicates the importance of regulated expression of osteoprotegerin for restoration of bone damage following osteolytic bone metastasis. Furthermore, identification of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binding and RANK-binding domains on osteoprotegerin would allow genetic modifications in osteoprotegerin to abolish TRAIL binding.

The present study also highlights the formation of woven bone in osteoblastic metastases (2527). In our study, the therapeutic effect of the mesenchymal stem cells is initially imparted by the woven bone formation surrounding the tumor nests. Formation of woven bone also characterizes osteoblastic metastases in prostate cancer and normal-to-fracture healing. Roudier et al. (2) did a detailed histomorphic analysis of bone samples obtained from prostate cancer patients with bone metastasis and observed equal representation of osteoblastic and osteolytic components. They also showed that the woven bone formation in the osteoblastic metastases originated from the skeletal mesenchymal stem cells. The pattern of woven bone formation in our experiment truly matches the histologic pattern observed in patient with prostate cancer bone metastasis, as reported by Roudier et al. (2). We speculate that the presentation of woven bone in prostate cancer bone metastasis is an endogenous therapeutic response by the resident mesenchymal stem cells rather than a pathologic outcome. Continuous presence of tumor cells in the bone induces osteolysis, which in turn signals the mesenchymal stem cells to initiate osteoblastogenesis and manifest the formation of woven bone as part of an early therapeutic response. As we compared growth kinetics of highly osteolytic PC3 cells versus osteoblastic C4-2B cells in the tibia of SCID mice, the rate of tumor growth is significantly lower in tibia injected with C4-2B cells compared with PC3 cells. Therefore, it is likely that low turnover of C4-2B cells provides a therapeutic window for the endogenous mesenchymal stem cells to induce bone formation to make up for the lost bone due to initial osteolytic event required by the cancer cells to colonize in the bone microenvironment. At this point, we are unable to comment on other factors responsible for the slow in vivo turnover of the C4-2B cells. When mice were sacrificed 6 months after the implantation of the C4-2B cells, all the lesions in the tibiae were of osteolytic nature. Determination of osteoblastic and/or osteolytic phenotype may be dependent on factors such as growth kinetics of the cancer cells and phenotypic changes of the cancer cells, and events such as hypoxia in the tumor microenvironment. Hypoxia is known to promote osteolytic bone metastasis and suppresses osteoblast differentiation (28); therefore, slower turnover of C4-2B cells may have delayed the onset of hypoxia compared to PC3 cells, which may have delayed the formation of osteolytic lesions. Nyambo et al. (29) reported that osteoprotegerin produced by the bone marrow stromal cells inhibits TRAIL-induced apoptosis of the tumor cells and favors the growth of prostate cancer in vitro. Our in vitro coculture assay showed that the bone marrow microenvironment and osteoprotegerin produced by the mesenchymal stem cells favored PC3 cell proliferation. Interestingly, mesenchymal stem cells or mesenchymal stem cells–osteoprotegerin imparted therapeutic benefits when applied in vivo. Based on these findings, we conclude that primary mesenchymal stem cells has the potential to provide therapeutic advantage in limiting the establishment of prostate cancer in the bone at an early stage by the virtue of its ability to differentiate into bone and inhibit osteoclastogenesis. These data signify that relatively abundant amounts of mesenchymal stem cells in the tumor microenvironment can provide therapeutic effects by activating osteoprotegerin and/or other factors through interactions with prostate cancer cells. Because the amount of mesenchymal stem cells in the bone microenvironment is extremely low (1 in 108 cells), strategies to endogenously mobilize and proliferate mesenchymal stem cells upon bone metastasis of osteolytic cancers may provide significant therapy and reduce morbidity and mortality encountered in late-stage prostate cancer patients. The aim of the present study was to determine potential of adult bone marrow–derived mesenchymal stem cells for the prevention of cancer osteolysis. Based on the limitations of the model used, it is imperative that additional studies should be done with other osteolytic bone metastasis models having the bone defect in the entire skeleton in immunocompetent animals before translating the findings for humans.

No potential conflicts of interest were disclosed.

We thank Dr. Maria Johnson and Xingsheng Li for the excellent technical assistance in micro–computed tomography measurements.

1
Jemal
A
,
Siegel
R
,
Ward
E
,
Murray
T
,
Xu
H
,
Thun
MJ
. 
Cancer statistics
.
CA Cancer J Clin
2007
;
57
:
43
66
.
2
Roudier
MP
,
Morrissey
C
,
True
LD
,
Higano
CS
,
Vessella
RL
,
Ott
SM
. 
Histopathological assessment of prostate cancer bone osteoblastic metastases
.
J Urol
2008
;
180
:
1154
60
.
3
Ye
L
,
Kynaston
HG
,
Jiang
WG
. 
Bone metastasis in prostate cancer: molecular and cellular mechanisms
.
Int J Mol Med
2007
;
20
:
103
11
.
4
Guise
TA
,
Yin
JJ
,
Mohammad
KS
. 
Role of endothelin-1 in osteoblastic bone metastases
.
Cancer
2003
;
97
:
779
84
.
5
Choueiri
MB
,
Tu
SM
,
Yu-Lee
LY
,
Lin
SH
. 
The central role of osteoblasts in the metastasis of prostate cancer
.
Cancer Metastasis Rev
2006
;
25
:
601
9
.
6
Blair
JM
,
Zhou
H
,
Seibel
MJ
,
Dunstan
CR
. 
Mechanisms of disease: roles of OPG, RANKL and RANK in the pathophysiology of skeletal metastasis
.
Nat Clin Pract Oncol
2006
;
3
:
41
9
.
7
Wittrant
Y
,
Theoleyre
S
,
Chipoy
M
, et al
. 
RANKL/RANK/OPG: new therapeutic targets in bone tumours and associated osteolysis
.
Biochim Biophys Acta
2004
;
1704
:
49
57
.
8
Dougall
WC
,
Chaisson
M
. 
The RANK/RANKL/OPG triad in cancer-induced bone diseases
.
Cancer Metastasis Rev
2006
;
25
:
541
9
.
9
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
.
10
Simonet
WS
,
Lacey
DL
,
Dunstan
CR
, et al
. 
Osteoprotegerin: a novel secreted protein involved in the regulation of bone density
.
Cell
1997
;
89
:
309
19
.
11
Chanda
D
,
Isayeva
T
,
Kumar
S
, et al
. 
Systemic osteoprotegerin gene therapy restores tumor-induced bone loss in a therapeutic model of breast cancer bone metastasis
.
Mol Ther
2008
;
16
:
871
8
.
12
Holen
I
,
Shipman
CM
. 
Role of osteoprotegerin (OPG) in cancer
.
Clin Sci (Lond)
2006
;
110
:
279
91
.
13
Onyia
JE
,
Galvin
RJ
,
Ma
YL
, et al
. 
Novel and selective small molecule stimulators of osteoprotegerin expression inhibit bone resorption
.
J Pharmacol Exp Ther
2004
;
309
:
369
79
.
14
Vanderkerken
K
,
De Leenheer
E
,
Shipman
C
, et al
. 
Recombinant osteoprotegerin decreases tumor burden and increases survival in a murine model of multiple myeloma
.
Cancer Res
2003
;
63
:
287
9
.
15
Zhang
J
,
Dai
J
,
Qi
Y
, et al
. 
Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone
.
J Clin Invest
2001
;
107
:
1235
44
.
16
Morony
S
,
Capparelli
C
,
Sarosi
I
,
Lacey
DL
,
Dunstan
CR
,
Kostenuik
PJ
. 
Osteoprotegerin inhibits osteolysis and decreases skeletal tumor burden in syngeneic and nude mouse models of experimental bone metastasis
.
Cancer Res
2001
;
61
:
4432
6
.
17
Capparelli
C
,
Kostenuik
PJ
,
Morony
S
, et al
. 
Osteoprotegerin prevents and reverses hypercalcemia in a murine model of humoral hypercalcemia of malignancy
.
Cancer Res
2000
;
60
:
783
7
.
18
Kumar
S
,
Chanda
D
,
Ponnazhagan
S
. 
Therapeutic potential of genetically modified mesenchymal stem cells
.
Gene Ther
2008
;
15
:
711
15
.
19
Studeny
M
,
Marini
FC
,
Dembinski
JL
, et al
. 
Mesenchymal stem cells: potential precursors for tumor stroma and targeted delivery vehicles for anticancer agents
.
J Natl Cancer Inst
2004
;
96
:
1593
603
.
20
Nakamura
K
,
Ito
Y
,
Kawano
Y
, et al
. 
Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model
.
Gene Ther
2004
;
11
:
1155
64
.
21
Tropel
P
,
Noël
D
,
Platet
N
,
Legrand
P
,
Benabid
AL
,
Berger
F
. 
Isolation and characterisation of mesenchymal stem cells from adult mouse bone marrow
.
Exp Cell Res
2004
;
295
:
395
406
.
22
Kumar
S
,
Ponnazhagan
S
. 
Bone homing of mesenchymal stem cells by ectopic α4 integrin expression
.
FASEB J
2007
;
21
:
3917
27
.
23
Holen
I
,
Croucher
PI
,
Hamdy
FC
,
Eaton
CL
. 
Osteoprotegerin (OPG) is a survival factor for human prostate cancer cells
.
Cancer Res
2002
;
62
:
1619
23
.
24
Fritz
V
,
Noël
D
,
Bouquet
C
, et al
. 
Antitumoral activity and osteogenic potential of mesenchymal stem cells expressing the urokinase-type plasminogen antagonist amino-terminal fragment in a murine model of osteolytic tumor
.
Stem Cells
2008
;
26
:
2981
90
.
25
Dotan
ZA
. 
Bone imaging in prostate cancer
.
Nat Clin Pract Urol
2008
;
5
:
434
44
.
26
Kingsley
LA
,
Fournier
PG
,
Chirgwin
JM
,
Guise
TA
. 
Molecular biology of bone metastasis
.
Mol Cancer Ther
2007
;
6
:
2609
17
.
27
Guise
TA
,
Mohammad
KS
,
Clines
G
, et al
. 
Basic mechanisms responsible for osteolytic and osteoblastic bone metastases
.
Clin Cancer Res
2006
;
12
:
6213
6
.
28
Hiraga
T
,
Kizaka-Kondoh
S
,
Hirota
K
,
Hiraoka
M
,
Yoneda
T
. 
Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer
.
Cancer Res
2007
;
67
:
4157
63
.
29
Nyambo
R
,
Cross
N
,
Lippitt
J
, et al
. 
Human bone marrow stromal cells protect prostate cancer cells from TRAIL-induced apoptosis
.
J Bone Miner Res
2004
;
19
:
1712
21
.

Competing Interests

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.

Supplementary data