8
[CANCER RESEARCH 59, 1987–1993, April 15, 1999] Severe Combined Immunodeficient-hu Model of Human Prostate Cancer Metastasis to Human Bone 1 Jeffrey A. Nemeth, John F. Harb, Ubirajara Barroso, Jr., Zhanquan He, David J. Grignon, and Michael L. Cher 2 Program in Urologic Oncology (J. A. N., D. J. G., M. L. C.) and Cancer Biology (X. H.), Barbara Ann Karmanos Cancer Institute, and Departments of Urology (J. A. N., J. F. H., U. B., M. L. C.) and Pathology (D. J. G, M. L. C.), Wayne State University School of Medicine, Detroit, Michigan 48201 ABSTRACT Commonly used in vivo models of prostate cancer metastasis include syngeneic rodent cancers and xenografts of human cancer in immuno- deficient mice. However, the occurrence of osseous metastases in these models is rare, and in xenograft models, species-specific factors may limit the ability of human cells to metastasize to rodent bones. We have mod- ified the severe combined immunodeficient (SCID)-human model to test the ability of circulating human prostate cancer cells to home to macro- scopic fragments of human bone and other organs previously implanted into SCID mice. We have also compared the growth of human prostate cancer cells in various human and mouse tissue microenvironments in vivo. Macroscopic fragments of human fetal bone, lung, or intestine (16 –22 weeks gestation) or mouse bone were implanted s.c. into male CB.17 SCID mice. Four weeks later, human prostate cancer cells were injected either i.v. via the tail vein (circulating cell colonization assay) or directly into the implanted tissue fragments transdermally (end organ growth assay). Tumor growth was followed for 6 weeks by palpation and magnetic resonance imaging. After 6 weeks, tumors were enumerated in implanted human and mouse organ fragments and native mouse tissue. Tumors were characterized by histology, immunohistochemistry, and chromosomal analysis. After i.v. injection, circulating PC3 cells success- fully colonized implanted human bone fragments in 5 of 19 mice. Tumors were easily followed by palpation and imaging and had an average volume of 258 mm 3 at autopsy. Histological examination revealed osteolysis and a strong desmoplastic stromal response, which indicated intense stromal- epithelial interaction. Bone tumors were subcultured, and chromosomal analysis demonstrated that the tumors were derived from the parental prostate cancer cell line. Microscopic tumor colonies were also found in a few mouse lungs after i.v. injection of PC3, DU145, and LNCaP cells, however the volume of the lung nodules was less than 1 mm 3 in all of the cases. No colonization of human lung or intestine implants, the mouse skeleton, or other mouse organs was detected, demonstrating a species- and tissue-specific colonization of human bone by PC3 cells. Direct injec- tion of 10 4 prostate cancer cells into human bone implants resulted in large tumors in 75–100% of mice. PC3 and DU145 bone tumors were primarily osteolytic, whereas LNCaP bone tumors were both osteoblastic and osteolytic. PC3 and LNCaP bone tumors showed a desmoplastic stromal response, which indicated intense stromal-epithelial interaction. All three of the cell lines formed tumors in implanted human lung tissue; however, the tumors were all <10 mm 3 in volume and showed minimal stromal involvement. No tumors formed after either s.c. injection or injection of cells into implanted mouse bone demonstrating both species- and tissue-specific enhancement of growth of human prostate cancer cells by human bone. The severe combined immunodeficient-human model provides a useful system to study species-specific mechanisms involved in the homing of human prostate cancer cells to human bone and the growth of human prostate cancer cells in human bone. INTRODUCTION Bone has long been recognized to be the most common target organ of prostate cancer metastasis (1), and the phenomenon of osseous metastasis signals the final, incurable stage of disease. The reasons underlying the proclivity of prostate cancer to metastasize to bone remain unclear; however, the competing hypotheses can be divided into two categories: anatomical/hemodynamic factors (Batson’s plexus) and “seed and soil.” In 1940, Batson (2) described a venous portal system between the plexus of veins draining the prostate and the plexus of veins surrounding the lumbar spine. In animal experi- ments and in human cadavers, Batson demonstrated that dye injected into the deep dorsal vein of the penis passed through the prostatic plexus and continued through longitudinal, valveless veins to the sinusoidal venous structures of the lumbar spine (2, 3). These exper- iments suggested that passive hemodynamic patterns may contribute to the tendency of prostate cancer to metastasize to the lumbar spine. The alternative hypothesis has been attributed to Paget, who surmised that the metastatic pattern of a given type of carcinoma is dependent on the interaction of individual tumor cells with the microenvironment of the target tissue (4). Thus the high frequency of osseous metastases in prostate cancer is not due to passive hemodynamic patterns but to a molecular affinity between prostate cancer cells and the bone marrow microenvironment. The phenomenon of prostate cancer metastasis to bone has been difficult to mimic in animal models. In syngeneic animal prostate cancer models such as the Dunning rat model (5) and in the transgenic mouse model TRAMP (6), lymph node and pulmonary metastases are common, but the occurrence of bone metastasis from a primary tumor site is rare. It has also been difficult to mimic clinical patterns of osseous metastasis in immunodeficient murine xenograft models of human prostate cancer. For example, i.v. injection of up to 1 3 10 6 PC3 or LNCaP human prostate cancer cells does not typically result in the development of bone metastases (7, 8). Increased osseous metastasis of prostate cancer cell lines has been achieved by various experimental manipulations, including coinjection of tumor cells with bone stromal cells (9), orthotopic injection of prostate tumor cells (10), or brief occlusion of the vena cava during i.v. injection of cells (11). Nonetheless, these xenograft models all share the fact that human cells must metastasize to and grow in mouse organs. It is possible that many of the molecules involved in the metastatic process such as proteinases, adhesion molecules, chemotactic factors, and growth factors and their receptors, are species-specific, which may contribute to the difficulty in achieving bone metastases in xenograft models of prostate cancer. Recent reports have described the use of the SCID-hu 3 system to study the behavior of metastatic human tumor cells. It was found that small cell lung carcinoma cells that did not grow in murine organs after i.v. injection specifically colonized human fetal lung and bone marrow tissues implanted in SCID mice (12). Application of the SCID-hu 3 system to the study of human colon carcinoma has also led Received 10/9/98; accepted 2/16/99. 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 Supported by the American Cancer Society CRTG-97-044-01-EDT (to M. L. C.). 2 To whom requests for reprints should be addressed, at Departments of Urology and Pathology, Wayne State University School of Medicine, 540 East Canfield Road, Scott Hall #9112, Detroit, MI 48201. Phone: (313) 577-2879; Fax: (313) 577-0057; E-mail: [email protected]. 3 The abbreviations used are: SCID-hu, severe combined immunodeficient-human; MR, magnetic resonance; MRI, magnetic resonance imaging; FOV, field of view; CGH, comparative genomic hybridization; PSA, prostate-specific antigen. 1987 Research. on March 16, 2021. © 1999 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

[CANCER RESEARCH 59, 1987–1993, April 15, 1999]

Severe Combined Immunodeficient-hu Model of Human Prostate Cancer Metastasisto Human Bone1

Jeffrey A. Nemeth, John F. Harb, Ubirajara Barroso, Jr., Zhanquan He, David J. Grignon, and Michael L. Cher2

Program in Urologic Oncology (J. A. N., D. J. G., M. L. C.) and Cancer Biology (X. H.), Barbara Ann Karmanos Cancer Institute, and Departments of Urology (J. A. N., J. F. H.,U. B., M. L. C.) and Pathology (D. J. G, M. L. C.), Wayne State University School of Medicine, Detroit, Michigan 48201

ABSTRACT

Commonly used in vivo models of prostate cancer metastasis includesyngeneic rodent cancers and xenografts of human cancer in immuno-deficient mice. However, the occurrence of osseous metastases in thesemodels is rare, and in xenograft models, species-specific factors may limitthe ability of human cells to metastasize to rodent bones. We have mod-ified the severe combined immunodeficient (SCID)-human model to testthe ability of circulating human prostate cancer cells to home to macro-scopic fragments of human bone and other organs previously implantedinto SCID mice. We have also compared the growth of human prostatecancer cells in various human and mouse tissue microenvironmentsinvivo. Macroscopic fragments of human fetal bone, lung, or intestine(16–22 weeks gestation) or mouse bone were implanted s.c. into maleCB.17 SCID mice. Four weeks later, human prostate cancer cells wereinjected either i.v. via the tail vein (circulating cell colonization assay) ordirectly into the implanted tissue fragments transdermally (end organgrowth assay). Tumor growth was followed for 6 weeks by palpation andmagnetic resonance imaging. After 6 weeks, tumors were enumerated inimplanted human and mouse organ fragments and native mouse tissue.Tumors were characterized by histology, immunohistochemistry, andchromosomal analysis. After i.v. injection, circulating PC3 cells success-fully colonized implanted human bone fragments in 5 of 19 mice. Tumorswere easily followed by palpation and imaging and had an average volumeof 258 mm3 at autopsy. Histological examination revealed osteolysis and astrong desmoplastic stromal response, which indicated intense stromal-epithelial interaction. Bone tumors were subcultured, and chromosomalanalysis demonstrated that the tumors were derived from the parentalprostate cancer cell line. Microscopic tumor colonies were also found in afew mouse lungs after i.v. injection of PC3, DU145, and LNCaP cells,however the volume of the lung nodules was less than 1 mm3 in all of thecases. No colonization of human lung or intestine implants, the mouseskeleton, or other mouse organs was detected, demonstrating a species-and tissue-specific colonization of human bone by PC3 cells. Direct injec-tion of 104 prostate cancer cells into human bone implants resulted inlarge tumors in 75–100% of mice. PC3 and DU145 bone tumors wereprimarily osteolytic, whereas LNCaP bone tumors were both osteoblasticand osteolytic. PC3 and LNCaP bone tumors showed a desmoplasticstromal response, which indicated intense stromal-epithelial interaction.All three of the cell lines formed tumors in implanted human lung tissue;however, the tumors were all<10 mm3 in volume and showed minimalstromal involvement. No tumors formed after either s.c. injection orinjection of cells into implanted mouse bone demonstrating both species-and tissue-specific enhancement of growth of human prostate cancer cellsby human bone. The severe combined immunodeficient-human modelprovides a useful system to study species-specific mechanisms involved inthe homing of human prostate cancer cells to human bone and the growthof human prostate cancer cells in human bone.

INTRODUCTION

Bone has long been recognized to be the most common target organof prostate cancer metastasis (1), and the phenomenon of osseousmetastasis signals the final, incurable stage of disease. The reasonsunderlying the proclivity of prostate cancer to metastasize to boneremain unclear; however, the competing hypotheses can be dividedinto two categories: anatomical/hemodynamic factors (Batson’splexus) and “seed and soil.” In 1940, Batson (2) described a venousportal system between the plexus of veins draining the prostate andthe plexus of veins surrounding the lumbar spine. In animal experi-ments and in human cadavers, Batson demonstrated that dye injectedinto the deep dorsal vein of the penis passed through the prostaticplexus and continued through longitudinal, valveless veins to thesinusoidal venous structures of the lumbar spine (2, 3). These exper-iments suggested that passive hemodynamic patterns may contributeto the tendency of prostate cancer to metastasize to the lumbar spine.The alternative hypothesis has been attributed to Paget, who surmisedthat the metastatic pattern of a given type of carcinoma is dependenton the interaction of individual tumor cells with the microenvironmentof the target tissue (4). Thus the high frequency of osseous metastasesin prostate cancer is not due to passive hemodynamic patterns but toa molecular affinity between prostate cancer cells and the bonemarrow microenvironment.

The phenomenon of prostate cancer metastasis to bone has beendifficult to mimic in animal models. In syngeneic animal prostatecancer models such as the Dunning rat model (5) and in the transgenicmouse model TRAMP (6), lymph node and pulmonary metastases arecommon, but the occurrence of bone metastasis from a primary tumorsite is rare. It has also been difficult to mimic clinical patterns ofosseous metastasis in immunodeficient murine xenograft models ofhuman prostate cancer. For example, i.v. injection of up to 13 106

PC3 or LNCaP human prostate cancer cells does not typically resultin the development of bone metastases (7, 8). Increased osseousmetastasis of prostate cancer cell lines has been achieved by variousexperimental manipulations, including coinjection of tumor cells withbone stromal cells (9), orthotopic injection of prostate tumor cells(10), or brief occlusion of the vena cava during i.v. injection of cells(11). Nonetheless, these xenograft models all share the fact thathuman cells must metastasize to and grow in mouse organs. It ispossible that many of the molecules involved in the metastatic processsuch as proteinases, adhesion molecules, chemotactic factors, andgrowth factors and their receptors, are species-specific, which maycontribute to the difficulty in achieving bone metastases in xenograftmodels of prostate cancer.

Recent reports have described the use of the SCID-hu3 system tostudy the behavior of metastatic human tumor cells. It was found thatsmall cell lung carcinoma cells that did not grow in murine organsafter i.v. injection specifically colonized human fetal lung and bonemarrow tissues implanted in SCID mice (12). Application of theSCID-hu3 system to the study of human colon carcinoma has also led

Received 10/9/98; accepted 2/16/99.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby markedadvertisementin accordance with18 U.S.C. Section 1734 solely to indicate this fact.

1 Supported by the American Cancer Society CRTG-97-044-01-EDT (to M. L. C.).2 To whom requests for reprints should be addressed, at Departments of Urology and

Pathology, Wayne State University School of Medicine, 540 East Canfield Road, ScottHall #9112, Detroit, MI 48201. Phone: (313) 577-2879; Fax: (313) 577-0057; E-mail:[email protected].

3 The abbreviations used are: SCID-hu, severe combined immunodeficient-human;MR, magnetic resonance; MRI, magnetic resonance imaging; FOV, field of view; CGH,comparative genomic hybridization; PSA, prostate-specific antigen.

1987

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

to the identification of adhesion molecules that may be important incolon cancer metastasis (13). These studies clearly illustrate the im-portance of species- and tissue-specific interactions in the design ofinvivo metastasis models. Using the SCID-hu system, we have foundthat circulating human prostate cancer cell lines preferentially colo-nized implanted human bone tissue. In addition, growth of humanprostate cancer cells was specifically enhanced by direct interactionwith the human bone marrow microenvironment.

MATERIALS AND METHODS

Cell Lines and Cell Culture. DU145, LNCaP, and PC3 human prostatecancer cell lines were purchased from the American Type Culture Collection.DU145 cells were maintained in culture in DMEM-10% fetal bovine serum,and LNCaP and PC3 cells were maintained in RPMI 1640–10% fetal bovineserum. All of the culture reagents were purchased from Life Technologies, Inc.(Gaithersburg, MD). Cell lines were used at low passage number in all of theexperiments.

Animal Care and Human Tissue Implantation. Male homozygousC.B-17 scid/scidmice, aged 5 weeks, were purchased from Taconic Farms(Germantown, NY). Mice were maintained according to the NIH standardsestablished in the “Guidelines for the Care and Use of Experimental Animals,”and all of the experimental protocols were approved by the Animal Investi-gation Committee of Wayne State University. Human male fetal tissues wereobtained with informed consent according to regulations issued by each stateinvolved and the federal government. Methoxyflurane anesthesia was usedduring all of the surgical procedures. Implantation of 6-week-old mice withhuman fetal lung fragments (SCID-hu-L) or human fetal intestine fragments(SCID-hu-I) was performed as described by Shtivelman (12). Briefly, humanfetal lungs or intestines of 16–22 weeks of gestation were cut into 23 2 3 2mm fragments and implanted s.c. in the ventral surface of the mouse via asmall skin incision. Implantation of mice with human fetal human bonefragments (SCID-hu-B) was performed as described previously(14). Briefly,human fetal femurs and humeri of 16–22 weeks of gestation were divided inhalf longitudinally and then again in half transversely into four fragmentsapproximately 1 cm long and 3 or 4 mm in diameter. These bone fragmentswere implanted s.c. in the flank through a small skin incision with the openedmarrow cavity against the mouse muscle. As a control for species-specificgrowth of prostate cells in human bone, femurs were harvested from normalSCID mice, and bone fragments were implanted in the flank of recipient micein an identical fashion (SCID-m-B). All of the mice received at least twodifferent tissue implants; human bone in the flank and human lung or intestinein the abdomen or human bone and mouse bone in opposite flanks.

Circulating Cell Colonization Assay. Cultured prostate cancer cell lineswere harvested by incubation with trypsin/EDTA and washed twice in Hank’sbalanced salt solution. Cell number and viability were measured by trypan blueexclusion. Forin vivo circulating cell colonization experiments, 13 106 cellsin a volume of 200 ml were injected into SCID-hu mice via lateral tail veinusing a 27-g needle. In all of the experiments, cancer cells were injected 4weeks after tissue implantation because this time point was found to be optimalfor colonization of human tissues by carcinoma cells in previous reports (12).

End Organ Tumor Growth Assay. For experiments measuring growth ofhuman prostate cancer cells in human metastatic organ microenvironmentsinvivo, 13 104 cells in a volume of 20ml were injected directly into implantedhuman or mouse tissue fragments through the skin using a 27-g needle. Again,injections were performed 4 weeks after tissue implantation. For the bonefragments, the opened marrow surface was targeted with the needle. Somecontrol mice also received the same number of cells injected s.c. in theshoulder region as a further control for tumor growth. For the LNCaP cell line,a second group of mice received a larger inoculum of 53 104 cells (see“Results”).

MRI. MR images were acquired on a Bruker AVANCE spectrometerequipped with 4.7-T horizontal-bore magnet and actively shielded gradients.During MR observation, the mouse was anesthetized (1.5% v/v halothane) andimmobilized on a bed that was heated by circulating temperature-controlledwater. A whole-body RF transceiver coil (70-mm diameter) was used totransmit homogeneous RF excitation and hold the animal bed. A RF surfacetransceiver coil (30-mm diameter) was placed on the back of the mouse to

receive MR signals because it produced images with increased signal:noiseratios compared with the whole-body coil. The surface coil and whole-bodycoil were actively decoupled to avoid signal interference. High-resolutionT1-weighted spin echo images (TE5 15 ms, TR5 545 ms, flip angle5 90`,8.0 cm FOV, and 2563 256 matrix size) were acquired in coronal sections toserve as the detail anatomical reference for tumor position. However, tumorcannot be easily distinguished from surrounding normal tissue in these T1-weighted images. To enhance contrast between tumor and normal tissue, amultislice T2-weighted spin echo sequence (TE5 60 ms, TR5 1225 ms, flipangle5 90`, 2 averages) with fat saturation was used. T2-weighted images of10 contiguous slices covering the whole tumor were obtained in two sections(axial and coronal), yielding in-plane resolution,0.1 mm2 with a slicethickness of 1.0 mm (6.4-cm or 8.0-cm FOV, 2563 256 matrix size).

Data Analysis and Histology. Mice were killed 6 weeks after injection oftumor cells by cervical dislocation while under methoxyflurane anesthesia.Human tissues and certain mouse tissues (lumbar vertebrae, ribs, visible lymphnodes, and lungs) were dissected, photographed, and measured with calipers.Tumor volume was estimated by the formulaa 3 b2/2, wherea is the longestdimension andb is the width. All of the other organs and the mouse skeletonwere visually inspected. Mouse lungs were stained in Bouin’s fluid to revealtumor nodules. Portions of each tissue were fixed in 10% buffered formalin,decalcified (bone specimens), embedded in paraffin, sectioned, and stainedwith H&E for routine histological examination. Immunohistochemical stainingwas also performed to confirm the presence of tumor cells in bone tissues.Briefly, sections were deparaffinized and rehydrated through graded alcohols.For antigen retrieval (15), slides were placed in 10 mM citrate buffer and boiledby microwave heating for 5 min. Nonspecific sites were blocked by incubationwith Superbloc (ScyTek, Logan, UT). Sections were incubated with eithermonoclonal antipan cytokeratin antibody (C-2562, Sigma, St. Louis, MO) ornonspecific mouse immunoglobulins at 1:200 dilution. Positive immunoreac-tive sites were detected using a Vectastain alkaline phosphatase kit (Vector,Burlingame, Ca) and Sigma Fast Red substrate, and nuclei were brieflycounterstained with hematoxylin.

Mean tumor volumes were calculated for each cell line test group and tumorsite, and statistical comparisons of mean tumor volumes between groups wereperformed using Student’st test (one-tailed analysis).

Subculture of Tumors and Genetic Analysis.Tumors arising in im-planted human bone tissues after i.v. injection of tumor cells were analyzed toconfirm the cell of origin. Small portions were minced under sterile conditionsand cultured in Keratinocyte-SFM (Life Technologies, Inc.) containing EGF,bovine pituitary extract, and 10% fetal bovine serum until a confluent mono-layer of cells was established. This medium was chosen to retard the growth offibroblastic cells. After the first and second passages, cultures were stained forcytokeratin as described above (“Histology”) to assess the purity of the tumorcell populations. By the second passage, the cultured tumor cells were nearly100% cytokeratin-positive, suggesting minimal stromal cell survival in culture.At this point, cells were analyzed by CGH.

CGH analysis was performed essentially as described previously (16, 17).DNA was prepared separately from bone tumor subcultures and parental celllines by proteinase K digestion and phenol/chloroform extraction. Onemg oftumor DNA or reference normal DNA was nick-translated in the presence ofFITC-12-dUTP (green fluorescence) or Texas Red-5-dUTP (red fluorescence,DuPont NEN, Boston, MA), respectively. Labeled tumor and normal DNAwere hybridized together with 30mg of Cot-1 DNA (Life Technologies, Inc.)onto normal male metaphase spreads (Vysis, Downers Grove, IL) for 2–3 days.After washing, slides were counterstained with 0.1 mM DAPI. Ten to 15metaphase images were acquired for each tumor-normal hybridization, ofwhich the best images were chosen for quantitative analysis. Quantitativeimage analysis was performed as described previously using the QUIPS CGHsystem (Vysis Inc., Downers Grove, IL; Ref. 18).

Serum PSA Measurements.Serum samples were taken from mice bearingLNCaP tumors in implanted human bone fragments at the time the mice werekilled. Total PSA levels were measured by routine methods at the ClinicalChemistry Laboratory of the Detroit Medical Center.

RESULTS

Implantation of Human Fetal Tissues. SCID-hu mice were usedfor circulating cell colonization and end organ growth experiments

1988

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

3–4 weeks after the implantation of human tissues. Previous studiesdemonstrated higher rates of colonization of implanted human tissueswhen cells were injected at early time points, before the recovery ofhematopoiesis in the bone marrow compartment (12). To assureviability of implanted human fetal tissues at the time of injection, fourSCID-hu mice bearing different human tissue implants were killed 4weeks after implantation, and the tissues were examined histologi-cally. The marrow compartment of human bone implants containedmainly stromal elements and some residual hematopoietic cells, andevidence of new bone formation could be seen (Fig. 1A). Small bloodvessels containing RBCs were clearly visible in the tissue sections.Human lung implants grew in size, and the tissue appeared to becomemore differentiated with evidence of alveolar structures, columnarepithelium lining bronchioles, and cartilage (Fig. 1B). Likewise, im-planted human fetal intestine appeared healthy, with a well-differen-tiated epithelial lining (Fig. 1C). These results are consistent withprevious reports describing the biology of implanted human fetaltissue fragments (12, 14) and suggest that the implanted tissues werehealthy and adequately vascularized.

Colonization of Human Tissue Implants by Circulating TumorCells. The ability of circulating prostate cancer cells to colonizehuman tissuesin vivo was tested by the injection of DU145, LNCaP,or PC3 cells into SCID-hu mice via the lateral tail vein. Three weeksafter the injection of cancer cells, 5 (26%) of 19 mice receiving PC3cells developed grossly palpable and measurable tumors in implantedhuman bone fragments (Table 1). Six weeks after injection, tumorswere confirmed and measured at autopsy. Histological analysis re-vealed large solid tumors composed of cohesive nests of tumor cellsintermingled with a cellular desmoplastic stromal response (Fig. 2B).Close examination showed areas of bone destruction and resorption,which indicated an osteolytic tumor phenotype (Fig. 2C).

Immunohistochemical staining for cytokeratin confirmed the pres-ence of epithelial tumor cells (not shown). DU145 and LNCaP cellsdid not form macroscopic palpable tumors in human bone tissues aftertail vein injection (Table 1), and this observation was confirmedhistologically and by immunohistochemical staining for cytokeratin(not shown). Histological evaluation of implanted human lung andintestine tissues did not reveal any evidence of colonization by any ofthe cell lines tested (Table 1), which suggested that circulating PC3cells prefer to colonize human bone as opposed to other humantissues, and the colonization of human bone by PC3 cells was notmerely an effect of the implantation procedure.

Because cells injected i.v. via the tail vein must pass throughthe mouse lung before entering the general circulation, we examinedthe mouse lungs for signs of colonization. In a small number of mice,the three cell lines formed between 5 and 15 colonies visible on thelung surface (Table 1), however these colonies were always less than1 mm3 in volume and did not cause any adverse respiratory effects. Of

the two mice with PC3 tumors in the mouse lungs, one occurred in amouse that also had a human bone tumor. A representative lungcolony formed by circulating PC3 tumor cells is shown in Fig. 2D.

None of the mice experienced limb paralysis, which was consistentwith a lack of spinal metastases, and all of the mouse tissues examinedwere clear of tumor deposits by gross and histological inspection. Theabsence of metastasis to mouse bone suggested that species-specificfactors favored targeting to the human bone environment.

To confirm the origin of tumors arising in implanted human bonetissues after i.v. injection of PC3 cells, explant cultures of thesetumors were established. After the second passage, the cultures weregreater than 98% epithelial cells, as judged by cellular morphologyand positive staining for cytokeratin (not shown), confirming that thetumors were of epithelial origin. One of the resulting cell populations,termed PC3-HB1, was characterized by CGH analysis. We foundgains in the regions of chromosome 7p, 8q, and 11q, and losses in theregions of chromosome 4q, 9p, and 10p (not shown). Analysis of theparental PC3 cells revealed the same pattern of chromosomal alter-ations as PC3-HB1. This pattern of gains and losses was consistentwith the recent findings of Nupponenet al.(19). Genetic analysis thusconfirmed that the bone tumors that were formed in the circulatingcell colonization assay originated from circulating PC3 cells.

The ability of circulating PC3-HB1 cells to form tumors inSCID-hu mice was also tested. Injection of 13 106 cells intoSCID-hu mice via the lateral tail vein did not result in any detectabletumors in human bone or lung tissues nor in the mouse lungs (Table1). The reason for this result was unclear.

End Organ Tumor Growth Assay. To measure the effects ofvarious human tissue microenvironments on thein vivo growth ofhuman prostate cancer cells, we used a direct injection approach toensure uniform seeding of the tissues with the tumor cells. Theinjection of 13 104 DU145, PC3, or PC3-HB1 cancer cells directlyinto implanted human bone fragments resulted in grossly evident

Fig. 1. SCID-hu tissue implants. Histological appearance of human fetal tissues 4 weeks after implantation into SCID mice.A, the marrow compartment of human bone implantscontained mainly stromal elements and some residual hematopoietic cells, and evidence of new bone formation could be seen (arrow). 340. B, implanted human lung with alveolarstructures (arrow) and columnar epithelium lining bronchioles (arrowhead).340. C, implanted human fetal intestine appeared healthy, with a well-differentiated epithelial lining(arrow). 340. No evidence of necrosis was seen in any tissue examined.

Table 1 Circulating cell colonization assay

Tumor incidence was measured in implanted human tissues and host mouse lungs 6weeks after i.v. injection of 13 106 prostate cancer cells.

Cell line

Target organ

Humanbone

Humanlung

Humanintestine

Mouse lunga

(average no. of colonies/mouse)

DU145 0/12 0/12 0/7 5/12 (12.5)LNCaP 0/12 0/6 0/3 1/12 (6)PC3 5/19b 0/16 0/5 2/19 (4.5)PC3-HB1 0/12 0/12 NDc 0/12a Volume of individual prostate cancer cell lung nodules was less than 1 mm3 in all of

the cases.b Average PC3 bone tumor volume5 258.0 mm3.c ND, not done.

1989

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

tumors in a majority of mice (Table 2). The bone tumors were easilypalpable through the skin and could be measured with calipers.

To test the accuracy of tumor measurement before sacrifice, MRIwas performed on two mice bearing PC3 tumors in human bonetissue. The tumors appeared as regions of high signal intensity on T2

weighted images (Fig. 3). Residual bony tissue could be discerned asdark punctate areas within the tumor mass. Measurements taken fromthese images agreed with direct measurement of the same bone tumorsharvested the following day.

The average size of bone tumors formed by PC3 cells was larger thantumors formed by DU145 (P, 0.05), which indicated more rapid growthof PC3 cells (Table 2). Injection of 13 104 LNCaP cells did not resultin detectable bone tumors (Table 2), however increasing the inoculumsize to 53 104 cells resulted in tumors in 6 (75%) of 8 mice. The bonetumors formed by LNCaP cells were also large and similar in size to thePC3 and PC3-HB1 bone tumors (P . 0.5 for both comparisons). Incontrast, no tumors were formed in any group after s.c. injection of thesame low number of cancer cells, which indicated that the human boneenvironment enhanced prostate tumor growthin vivo.

Histological examination of the bone tumors formed by each of thecell lines showed large, poorly differentiated tumors with a variabledegree of stromal-epithelial interaction (Fig. 4,A, B, andC). DU145cells formed solid, cellular tumor masses without central necrosis(Fig. 4A). A limited desmoplastic stromal response was present at thetumor-normal tissue interface. In contrast, the tumors formed by PC3cells showed a striking desmoplastic stromal response with large areasof central necrosis (Fig. 4C). Fragments of necrotic bone could beseen within the tumor masses. The differential distribution of tumorand stromal cells was more clearly demonstrated by immunostainingfor cytokeratin (Fig. 4,D, E, andF). Tumors formed by DU145 andPC3 also appeared to be osteolytic in nature. In DU145 tumors, areas

of bone resorption could clearly be seen adjacent to tumor cell nests(Fig. 4A, arrow); and in many of the PC3 tumors, the calcified bonetissue was almost totally replaced by tumor cells and fibrous stroma(Fig. 4C). LNCaP cells formed solid tumors in bone with a moderatedesmoplastic stromal response (Fig. 4B). The tumors formed byLNCaP cells appeared to be of a mixed osteoblastic/osteolytic type(Fig. 4B), and regions of bone resorption and deposition of new bonematrix (arrow) could be seen. The level of total PSA was alsomeasured in serum samples taken from two mice bearing LNCaPtumors in human bone tissues and was found to average 4.5 ng/ml.This result further confirmed the presence of LNCaP tumor in thesemice and suggested that LNCaP cells retained the ability to producePSA while growing in a human bone microenvironmentin vivo.

To control for the possibility that any implanted human tissue couldenhance tumor growth, prostate cancer cell lines were also injectedinto implanted human lung tissues. Tumor deposits formed in aportion of implanted human lung tissues directly injected with pros-tate tumor cells, and in all of the cases, the tumor nodules wereextremely small in comparison with tumors formed in the boneimplants (Table 2). For all of the three cell lines, the tumors that grewin implanted lung tissue consisted of solid masses of tumor cells withintervening vasculature (Fig. 4,G, H, and I). Very little interactionwith stromal cells could be seen, and many of the tumor nodules weresurrounded by a thin capsule.

To control for species specificity, prostate cancer cell lines wereinjected into s.c. implanted mouse bone fragments. For each cell line,four mouse bone implants were injected, and in no case did a bonetumor result (Table 2). This provided further evidence of the impor-tance of human-human stromal-epithelial interactions with regard toprostate cancer cells growing within bone.

Fig. 2. Circulating cell colonization assay. Repre-sentative tumor formed in SCID-hu mouse after i.v.injection of PC3 cells.A, gross appearance of tumor inhuman bone fragment. Tumor appeared as a smoothmass (bracket) extending from residual normal bonetissue.Bar, 1 cm. B, low power view of tumor mass,showing residual bone tissue at the periphery (arrow).320. C, close examination revealed cohesive nests oftumor cells in a desmoplastic stromal response. Evi-dence of osteolytic activity could also be seen (arrow-head).3100. D, representative colony of PC3 tumorcells in mouse lung, consisting of a small number ofcells (arrow).3100.

Table 2 End organ tumor growth assay

Tumor incidence and mean tumor volume were measured 6 weeks after the direct injection of prostate cancer cells into implanted human or mouse tissues.

Cell Line (no. of cells injected)

Injection site

Human bonea Human lunga Mouse bone implant Subcutaneous

DU145 (13 104) 7/9 (113.56 26.0) 4/9 (4.06 1.0) 0/4 0/7LNCaP (13 104) 0/7 5/10 (10.56 1.5) 0/4 0/8LNCaP (53 104) 6/8 (305.56 62.5) NDb ND 0/6PC3 (13 104) 16/18 (407.56 106.5) 3/14 (2.56 1.0) 0/4 0/8PC3-HB1 (13 104) 10/10 (235.56 92.0) 3/10 (1.06 0.5) ND ND

a Tumor volume (in parentheses) expressed in mm3 6 SE.b ND, not done.

1990

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

DISCUSSION

When designing animal models of human prostate cancer metasta-sis, it is important to take into account the possible contribution ofhuman-human stromal-epithelial interactions to the establishment andgrowth of prostate tumors in the target bone tissue. The results of thisstudy demonstrate that circulating human prostate cancer cells wereable to specifically and preferentially colonize implanted human bonetissue in SCID mice, and that human bone provided a more favorablegrowth environment for human prostate cancer cells than either hu-man lung or mouse bone tissue.

Circulating PC3 cells formed tumors in 5 (26%) of 19 human boneimplants but in only 2 (11%) of 19 mouse lungs. In addition, the

human bone tumors were very large compared with the microscopicmouse lung tumors, and we found no other metastatic deposits in thenative mouse organs. This is remarkable for several reasons: (a)i.v.-injected tumor cells must pass through the mouse lung capillarybeds before entering the general circulation; and (b) the mass ofhuman tissue available to the circulating cancer cells is quite small incomparison with the mouse skeleton and other organs. These obser-vations further support the notion that the colonization of human boneinvolved species- and tissue-specific mechanisms and was not due tothe passive lodging of tumor cells in the bone. Studies on other tumorcell lines have clearly demonstrated that selective colonization ofspecific target organs is not simply the result of nonspecific trapping

Fig. 3. MRI. T2-weighted spin echo MR images (TE5 60ms, TR 5 1225 ms) of a mouse bearing a PC3 tumor inimplanted human bone tissue.A, coronal section,B, axialsection.Arrows, tumor location.

Fig. 4. End organ growth assay. Representative photomicrographs demonstrating histological features of tumors formed in human bone tissue (A-F) and human lung tissue (G-I)6 weeks after the direct injection of human prostate cancer cell lines.A, DU145 cells formed solid tumors with evidence of osteolytic activity (arrow). 3100. A mild desmoplasticstromal response was visible in some areas of bone tumors and was revealed by red staining of tumor cells with a pancytokeratin antibody (D). 3200.B, bone tumors formed by LNCaPalso showed a desmoplastic stromal response, and evidence of both osteoblastic (arrow) and osteolytic activity (arrowhead) could be seen by routine staining.3100.E, the interminglingof red-stained LNCaP cells and stromal cells could be better seen after cytokeratin immunostaining.3200. C, PC3 cells formed solid osteolytic tumors, and bone tissue was oftencompletely destroyed.3100. A strong desmoplastic stromal response was visible in these tumors, revealed both by routine H&E staining (C) and by red staining for cytokeratin (F).3200. DU145, LNCaP, and PC3 cell lines all formed small solid tumors in human lung tissue and demonstrated little stromal-epithelial interaction (G, H, andI, respectively).3100.

1991

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

of tumor cells by the organ vasculature. One of the best studiedexamples is the B16 melanoma cell line, in which sublines have beenisolated that preferentially metastasize to the lung and liver (20).Similarly, in vitro selection combined with repeatedin vivo passagingof the PC3 human prostate cancer cell line has resulted in sublines thatreliably colonize distinct organ sites (7), and sublines of the LNCaPcell line with the ability to metastasize to bone have been isolated (9).A natural selection process in the original patients may also accountfor the differential ability of the three cell lines used in our study tocolonize human bone implants; PC3 cells were derived from a met-astatic bone lesion (21), whereas the LNCaP and DU145 cell lineswere derived from lymph node and brain metastases, respectively(22, 23).

The ability of circulating PC3 cells to colonize human bones in25% of our mice is also remarkable considering that we used routinelycultured PC3 cells at a low passage number after purchase, withoutany prior selective pressure. In previous studies, metastasis of PC3cells to bone has been achieved, but only after repeated rounds ofinvivo selection (7, 24) or through forced seeding of the lumbar verte-brae by simultaneous tail vein injection and caval occlusion (11).Taken in this light, our observed tumor rate in human bone clearlyexceeds what would be expected for the metastasis of PC3 cells tomouse bone.

The colonization of bone marrow by prostate cancer cells is likelyto involve adhesion to specific ligands found either on endothelial orparenchymal cells or within the extracellular matrix of the bonemarrow environment. It has been demonstrated that various bonecomponents can stimulate tumor cell adhesion and chemotaxis (25),and recently Kostenuiket al. (26) found that prostate cancer cellsadhere to type I collagen in osteoblast extracellular matrix via thea2b1 integrin. In addition, Pasqualini and Ruoslahti (27) have sug-gested the existence of organ-selective vascular address molecules.Using in vivo screening of random peptides, they identified aminoacid sequences that specifically target the vascular endothelium oftarget organs, through binding to either integrins or other undefinedmolecules. It is possible that prostate tumor cells use a similar mech-anism to adhere specifically to endothelium in human bone, allowingcolonization and eventual tumor formation. Such a mechanism hasbeen demonstrated for the homing of lymphocytes and granulocytes tospecific tissue sites (28). Conversely, a lack of expression of theseimportant molecules by DU145 and LNCaP could explain the inabil-ity of these circulating cells to colonize the human bone implants. Weare currently investigating specific cell-surface molecules that mayallow PC3 cells to colonize human bone.

The failure of circulating prostate cancer cells to form gross skeletaltumors in the mouse skeleton could be related to an inability of thecells to adhere to molecular targets in the mouse marrow compartmentvia mechanisms mentioned above. Alternatively, single tumor cells orsmall cell aggregates may have been present in the marrow but wereunable to proliferate and form detectable skeletal tumors because of alack of favorable environmental factors that are present in humanbone tissue. In fact, results of earlier studies have shown that whetherinjected via intraarterial or i.v. routes, tumor cells could be detected inorgan sites throughout the test animals, regardless of the ultimateformation of metastatic deposits (29). Rapid growth of human prostatecancer cells in mouse bone has been demonstrated after intrafemoralinjection (30); however, larger numbers of prostate cancer cells wereused in this study, making it difficult to determine whether tumorgrowth in that system was actually dependent on the bone marrowenvironment. Our observations show that a relatively small inoculumof 1 3 104 prostate cancer cells failed to grow when seeded directlyinto implanted mouse bone but formed tumors reliably in human boneimplants. Therefore, both specific adhesion mechanisms and species-

specific factors may help explain the difficulty in obtaining skeletalmetastasis in other rodent models.

Once metastatic cells arrive in the bone marrow compartment, theymust grow to form a tumor. Results from our end organ growth assayclearly showed that the three human prostate cancer cell lines pre-ferred to proliferate and form tumors in the human bone environmentin comparison with the human lung, mouse bone, and mouse s.c.environments. In 1889, Paget (4) noted that breast cancer consistentlymetastasized to specific organs. This observation led to the seed-and-soil hypothesis of metastasis, whereby certain organs provide a fa-vorable environment for the growth of certain tumor cells. Our ob-servations are consistent with the hypothesis of Paget and suggest thathuman bone provides a supportive environment for both the targetingand growth of human prostate cancer cells. Results of a number ofstudies have demonstrated the inductive capacity of bone and bone-derived factors on the growth of prostate cancer cells. Gleaveet al.(31) noted that soluble factors produced by bone fibroblasts couldinduce tumor formation by LNCaP in nude mice, suggesting thatparacrine interactions might be involved. Tissue culture studies havealso shown that soluble factors derived from bone marrow (32) orosteoblast-like cells (33) could stimulate growth of various prostatecancer cell lines. Using coculture techniques, Langet al. (34) foundthat close contact with bone marrow stromal cells in culture canstimulate the growth of cells derived from primary prostate tumors.Close physical interaction with bone cells has also been shown toenhance tumorigenicity of normally nontumorigenic LNCaP prostatecancer cells (35). s.c. tumors formed in nude mice after coinjection ofLNCaP cells with either bone or prostate fibroblasts but not withkidney or lung fibroblasts. Our present observations also suggest thathuman bone provides a more supportive environment than humanlung. A combination of the SCID-hu system andin vitro studies islikely to be useful in defining the factors important to prostate cancergrowth in bonein vivo.

The choice to use fetal bones rather than adult bones in this studywas based on the success of previous studies using this system (12,13) and the observations of Heikeet al. (36), who found that im-planted adult human bone fragments did not stimulate an angiogenicresponse and required constant cytokine infusion to prevent degener-ation and fibrosis of the marrow compartment. It is possible that somebiological factors are different between adult and fetal bones. Forexample, hematopoietic cells from implanted human marrow retainsome fetal characteristics (14). Nonetheless, the SCID-human bonesystem is still a valuable tool to begin addressing specific human-human interactions involved in bone metastasis, including growthfactors, adhesion molecules, extracellular matrix components, andcell-cell contact. In addition, the surgical procedures are simple,measurements are easy and accurate, and the resulting tumors causeminimal discomfort to the animals, allowing longer-term studies. Useof the SCID-hu model may also allow more accurate prediction of theresponse of metastatic cancer cells in the bone to therapeutic agents(37) and will be helpful in the development of new therapies targetedat sites of interaction between prostate cancer cells and the humanbone environment.

ACKNOWLEDGMENTS

We thank Dr. Jeffrey L. Evelhoch for valuable advice concerning MRI analysis.

REFERENCES

1. Abrams, H. L., Spiro, R., and Goldstein, N. Metastases in carcinoma: analysis of 1000autopsied cases. Cancer (Phila.),3: 74–85, 1950.

2. Batson, O. V. Function of vertebral veins and their role in the spread of metastases.Ann. Surg.,112: 138–149, 1940.

1992

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

3. Batson, O. V. The role of vertebral veins in metastatic processes. Ann. Intern. Med.,16: 38–45, 1942.

4. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet,1: 571–573, 1889.

5. Isaacs, J. T., Isaacs, W. B., Feitz, W. F. J., and Scheres, J. Establishment andcharacterization of seven Dunning rat prostatic cancer cell lines and their use indeveloping methods for predicting metastatic abilities of prostatic cancers. Prostate,9: 261–281, 1986.

6. Gingrich, J. R., Barrios, R. J., Morton, R. A., Boyce, B. F., DeMayo, F. J., Finegold,M. J., Angelopoulou, R., Rosen, J. M., and Greenberg, N. M. Metastatic prostatecancer in a transgenic mouse. Cancer Res.,56: 4096–4102, 1996.

7. Wang, M., and Stearns, M. E. Isolation and characterization of PC-3 human prostatictumor sublines which preferentially metastasize to select organs in S. C. I. D. mice.Differentiation,48: 115–125, 1991.

8. Dumont, P., Petein, M., Lespagnard, L., Tueni, E., and Coune, A. Unusual behaviourof the LNCaP prostate tumour xenografted in nude mice. In Vivo (Athens),7:167–180, 1993.

9. Thalmann, H. N., Anezinis, P. E., Chang, S. M., Zhau, H. E., Kim, E. E., Hopwood,V. L., Pathak, S., von Eschenbach, E. A., and Chung, L. W. K. Androgen-independentcancer progression and bone metastasis in the LNCaP model of human prostatecancer. Cancer Res.,54: 2577–2581, 1994.

10. Sato, N., Gleave, M. E., Bruchovsky, N., Rennie, P. S., Beraldi, E., and Sullivan,L. D. A metastatic and androgen-sensitive human prostate cancer model usingintraprostatic inoculation of LNCaP cells in SCID mice. Cancer Res.,57: 1584–1589,1997.

11. Shevrin, D. H., Kukreja, S. C., Ghosh, L., and Lad, T. E. Development of skeletalmetastasis by human prostate cancer in athymic nude mice. Clin. Exp. Metastasis,6: 401–409, 1988.

12. Shtivelman, E., and Namikawa, R. Species-specific metastasis of human tumor cellsin the severe combined immunodeficiency mouse engrafted with human tissue. Proc.Natl. Acad. Sci. USA,92: 4661–4665, 1995.

13. Sampson-Johannes, A., Wang, W., and Shtivelman, E. Colonization of human lunggrafts in SCID-hu mice by human colon carcinoma cells. Int. J. Cancer,65: 864–869,1996.

14. Kyoizumi, S., Baum, C. M., Kaneshima, H., McCune, J. M., Yee, E. J., andNamikawa, R. Implantation and maintenance of functional human bone marrow inSCID-hu mice. Blood,79: 1704–1711, 1992.

15. Shi, S-R., Key, M. E., and Kalra, K. L. Antigen retrieval in formalin-fixed, paraffinembedded tissues: an enhancement method for immunohistochemical staining basedon microwave oven heating of tissue sections. J. Histochem. Cytochem.,39: 741–748,1991.

16. Cher, M. L., Bova, G. S., Moore, D. H., Small, E. J., Carroll, P. R., Pin, S. S., Epstein,J. I., Isaacs, W. B., and Jensen, R. H. Genetic alterations in untreated metastases andandrogen independent prostate cancer detected by comparative genomic hybridizationand allelotyping. Cancer Res.,56: 3091–3098, 1996.

17. Cher, M. L., Lewis, P. E., Banerjee, M., Sakr, W., Grignon, D. J., and Powell, I. J.Similar pattern of chromosomal alterations in prostate cancers from African Ameri-cans and Caucasian Americans. Clin. Cancer Res.,4: 1273–1278, 1998.

18. Moore, D. H., Pallavicini, M., Cher, M. L., and Gray, J. W. A t-statistic for objectiveinterpretation of comparative genomic hybridization (CGH) profiles. Cytometry,28:183, 1997.

19. Nupponen, N. N., Hyytinen, E. R., Kallioniemi, A. H., and Visakorpi, T. Geneticalterations in prostate cancer cell lines detected by comparative genomic hybridiza-tion. Cancer Genet. Cytogenet.,101: 53–57, 1998.

20. Fidler, I. J., and Hart, I. R. Biological diversity in metastatic neoplasms: origins andimplications. Science (Washington DC),217: 998–1003, 1982.

21. Kaighn, M. E., Narayan, K. S., Ohnuki, Y., Lechner, J. F., and Jones, L. W.Establishment and characterization of a human prostatic carcinoma cell line (PC-3).Invest. Urol.,17: 16–23, 1979.

22. Horoszewicz, J. S., Leong, S. S., Kawinski, E., Karr, J. P, Rosenthal, H., Chu, T. M.,Miranda, E. A., and Murphy, G. P. LNCaP model of human prostatic carcinoma.Cancer Res.,43: 1809–1818, 1983.

23. Mickey, D. D., Stone, K. R., Wunderli, H., Mickey, G., and Paulson, D. F. Charac-terization of a human prostate adenocarcinoma cell line (DU-145) as a monolayerculture and a solid tumor in athymic mice. Progr. Clin. Biol. Res.,37: 67–84, 1980.

24. Pettaway, C. A., Pathak, S., Greene, G., Ramirez, E., Wilson, M. R., Killion, J. J., andFidler, I. J. Selection of highly metastatic variants of different human prostaticcarcinomas using orthotopic implantation in nude mice. Clin. Cancer Res.,2: 1627–1636, 1996.

25. Magro, C., Orr, F. W., Manishen, W., Sivananthan, K., and Mokashi, S. Adhesion,chemotaxis, and aggregation of Walker carcinosarcoma cells in response to productsof resorbing bone. J. Natl. Cancer Inst.,74: 29–34, 1985.

26. Kostenuik, P. J., Sanchez-Sweatman, O., Orr, F. W., Singh, G. Bone cell matrixpromotes the adhesion of human prostatic carcinoma cells via the a2b1 integrin. Clin.Exp. Metastasis,14: 19–26, 1996.

27. Pasqualini, R., and Ruoslahti, E. Organ targetingin vivo using phage display peptidelibraries. Nature (Lond.),380: 364–366, 1996.

28. Springer, T. A. Traffic signals for lymphocyte recirculation and leukocyte emigration:the multistep paradigm. Cell,76: 301–314, 1994.

29. Potter, K. M., Juacaba, S. F., Price, J. E., and Tarin, D. Observations on organdistribution of fluorescein-labelled tumour cells released intravascularly. InvasionMetastasis,3: 221–233, 1983.

30. Soos, G., Jones, R. F., Haas, G. P., and Wang, C. Y. Comparative intraosseal growthof human prostate cancer cell lines LNCaP and PC-3 in the nude mouse. AnticancerRes.,17: 4253–4258, 1997.

31. Gleave, M., Hsieh, J-T., Gao, C., von Eschenbach, A. C., and Chung, L. W. K.Acceleration of human prostate cancer growthin vivoby factors produced by prostateand bone fibroblasts. Cancer Res.,51: 3753–3761, 1991.

32. Chackal-Roy, M., Niemeyer, C., Moore, M., and Zetter, B. R. Stimulation of humanprostatic carcinoma cell growth by factors present in human bone marrow. J. Clin.Invest.,84: 43–50, 1989.

33. Lang, S. H., Miller, W. R., and Habib, J. K. Stimulation of human prostate cancer celllines by factors present in human osteoblast-like cells but not in bone marrow.Prostate,27: 287–293, 1995.

34. Lang, S. H., Clarke, N. W., George, N. J. R., Allen, T. D., and Testa, N. G. Interactionof prostate epithelial cells from benign and malignant tumor tissue with bone-marrowstroma. Prostate,34: 203–213, 1998.

35. Gleave, M., Hsieh, J-T., Gao, C., von Eschenbach, A. C., and Chung, L. W. K.Acceleration of human prostate cancer growthin vivoby factors produced by prostateand bone fibroblasts. Cancer Res.,51: 3753–3761, 1991.

36. Heike, Y., Ohira, T., Takahashi, M., and Nagahiro, S. Long-term human hematopoi-esis in SCID-hu mice bearing transplanted fragments of adult bone and bone marrowcells. Blood,86: 524–530, 1995.

37. Wilmanns, C., Fan, D., O’Brian, C. A., Bucana, C. D., and Fidler, I. J. Orthotopicand ectopic organ environments differentially influence the sensitivity of murinecolon carcinoma cells to doxorubicin and 5-fluorouracil. Int. J. Cancer,52:98 –104, 1992.

1993

SCID-hu MODEL OF PROSTATE CANCER BONE METASTASIS

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Severe Combined Immunodeficient-hu Model of Human ......control mice also received the same number of cells injected s.c. in the shoulder region as a further control for tumor growth

1999;59:1987-1993. Cancer Res   Jeffrey A. Nemeth, John F. Harb, Ubirajara Barroso, Jr., et al.   Prostate Cancer Metastasis to Human BoneSevere Combined Immunodeficient-hu Model of Human

  Updated version

  http://cancerres.aacrjournals.org/content/59/8/1987

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/59/8/1987.full#ref-list-1

This article cites 33 articles, 12 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/59/8/1987.full#related-urls

This article has been cited by 30 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/59/8/1987To request permission to re-use all or part of this article, use this link

Research. on March 16, 2021. © 1999 American Association for Cancercancerres.aacrjournals.org Downloaded from