Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cell and Tumor Biology

Simultaneous Blockade of Platelet-Derived Growth Factor-Receptor and Epidermal Growth Factor-Receptor Signaling and Systemic Administration of Paclitaxel as Therapy for Human Prostate Cancer Metastasis in Bone of Nude Mice

Sun Jin Kim, Hisanori Uehara, Sertac Yazici, Robert R. Langley, Junqin He, Rachel Tsan, Dominic Fan, Jerald J. Killion and Isaiah J. Fidler
Sun Jin Kim
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hisanori Uehara
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sertac Yazici
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert R. Langley
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Junqin He
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rachel Tsan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Dominic Fan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jerald J. Killion
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Isaiah J. Fidler
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-03-3763 Published June 2004
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Once prostate cancer metastasizes to bone, conventional chemotherapy is largely ineffective. We hypothesized that inhibition of phosphorylation of the epidermal growth factor receptor (EGF-R) and platelet-derived growth factor receptor (PDGF-R) expressed on tumor cells and tumor-associated endothelial cells, which is associated with tumor progression, in combination with paclitaxel would inhibit experimental prostate cancer bone metastasis and preserve bone structure. We tested this hypothesis in nude mice, using human PC-3MM2 prostate cancer cells. PC-3MM2 cells growing adjacent to bone tissue and endothelial cells within these lesions expressed phosphorylated EGF-R and PDGF-Rα and -β on their surfaces. The percentage of positive endothelial cells and the intensity of receptor expression directly correlated with proximity to bone tissue. Oral administration of PKI166 inhibited the phosphorylation of EGF-R but not PDGF-R, whereas oral administration of STI571 inhibited the phosphorylation of PDGF-R but not EGF-R. Combination therapy using oral PKI166 and STI571 with i.p. injections of paclitaxel induced a high level of apoptosis in tumor vascular endothelial cells and tumor cells in parallel with inhibition of tumor growth in the bone, preservation of bone structure, and reduction of lymph node metastasis. Collectively, these data demonstrate that blockade of phosphorylation of EGF-R and PDGF-R coupled with administration of paclitaxel significantly suppresses experimental human prostate cancer bone metastasis.

INTRODUCTION

Prostate cancer is the second greatest cause of cancer-related death among men in North America (1 , 2) . Metastasis to the bone, which is resistant to conventional therapies, causes devastating symptoms such as intractable bone pain, nerve compression, and pathological fractures (3 , 4) . Despite progress in the detection of prostate cancer, ∼24% of patients have metastatic lesions at initial diagnosis, and in an additional 30% of the patients, metastases are discovered during surgical staging. A significant number of patients with clinically localized disease who are treated with radical prostatectomy may also develop metastasis (5, 6, 7, 8) . Clearly then, the development of new treatment modalities for metastatic prostate cancer is important to the many men who will have metastatic prostate cancer.

Developing such a treatment requires an understanding of the progression of prostate cancer. To produce metastasis in the bone, prostate cancer cells must complete a series of sequential and highly selective steps (5 , 9 , 10) . We recently confirmed the “seed and soil” hypothesis of Stephen Paget (11) that certain tumor cells have a specific affinity for the milieu of certain organs. Specifically, we reported that the expression of various cytokines, growth factors, and their receptors on prostate cancer cells and endothelial cells differ in different organ microenvironments (12 , 13) . Moreover, cell response to protein tyrosine kinase receptor inhibitors, such as epidermal growth factor receptor (EGF-R; Ref. 12 ) and platelet-derived growth factor receptor (PDGF-R; Ref. 13 ), depends on the host organ microenvironment. EGF and EGF-R pathways have been reported to be related to cell proliferation (14 , 15) , activation of antiapoptotic effects (16) , and disease progression (17, 18, 19, 20, 21) . The PDGF and PDGF-R pathways are involved in cell division (22, 23, 24) , cell migration (25) , angiogenesis (26) , and cell survival (27) .

In our animal model, endothelial cells in tumors growing in the bone expressed phosphorylated EGF-R or PDGF-R on their surfaces, whereas endothelial cells in tumors growing in the muscle or endothelial cells in unaffected tissues did not (12 , 13) . Oral administration of the protein tyrosine kinase inhibitors PKI166 (28) or STI571 (29 , 30) has been found to inhibit phosphorylation of EGF-R and PDGF-R, respectively, and when each was combined with i.p. injection of paclitaxel, these inhibitors significantly inhibited tumor growth (12 , 13) . Because human prostate cancer cells growing adjacent to bone tissue express both EGF and PDGF and both tumor cells and tumor-associated endothelial cells express phosphorylated EGF-R and PDGF-R, we hypothesized that simultaneous targeting of both receptors combined with systemic injections of paclitaxel would produce additive therapeutic results. We report here the therapeutic effects of combining PKI166 with STI571 and paclitaxel on experimental human prostate cancer bone metastases in the tibias of nude mice.

MATERIALS AND METHODS

PC-3MM2 Metastatic Variant of Human Prostate Cancer Cell Line.

The highly metastatic cell line PC-3MM2 (31) was maintained as monolayer cultures in DMEM (Life Technologies, Inc., Grand Island, NY) supplemented with 10% fetal bovine serum, sodium pyruvate, nonessential amino acids, l-glutamine, a 2-fold vitamin solution (Life Technologies, Inc.), and penicillin-streptomycin (Flow Laboratories, Rockville, MD) in 5% CO2–95% air at 37°C. The cultures were free of Mycoplasma and the following murine viruses: reovirus type 3, pneumonia virus, K virus, Theiler’s encephalitis virus, ectomelia virus, and lactate dehydrogenase virus (assayed by M. A. Bioproducts, Walkersville, MD).

Animals.

Male athymic nude mice (NCI-nu) were purchased from the Animal Production Area of the National Cancer Institute-Frederick Cancer Research Facility (Frederick, MD). The mice were housed and maintained in specific pathogen-free conditions. The facilities were approved by the American Association for Accreditation of Laboratory Animal Care and met all current regulations and standards of the United States Department of Agriculture, United States Department of Health and Human Services, and the NIH. The mice were used in accordance with institutional guidelines when they were 6 to 8 weeks of age.

Intratibial Injection of Tumor Cells.

To produce bone tumors, PC-3MM2 cells were harvested from subconfluent cultures by brief exposure to 0.25% trypsin and 0.02% EDTA. Trypsinization was stopped with medium containing 10% fetal bovine serum, and the cells were washed once in serum-free medium and resuspended in Ca2+- and Mg2+-free HBSS. Cell viability was determined by trypan blue exclusion, and only single-cell suspensions with >95% viability were used to produce tumors in the tibias of mice.

Nude mice were anesthetized with Nembutal (0.5 mg/g of body weight; Abbott Laboratories, Chicago, IL). A percutaneous intraosseal injection was made by drilling a 27-gauge needle into the tibia immediately proximal to the tuberositas tibia. After penetration of the cortical bone, the needle was inserted into the shaft of the tibia, and 20 μl of the cell suspension (2 × 105 cells) were deposited in the bone cortex by use of a calibrated, push-button-controlled dispensing device (Hamilton Syringe Co., Reno, NV). To prevent leakage of cells into the surrounding muscles, a cotton swab was held for 1 min over the site of injection. The animals tolerated the surgical procedure well, and no anesthesia-related deaths occurred.

Therapy for Human Prostate Cancer Cells Growing in the Tibias of Athymic Nude Mice.

PKI166 (4-phenethylamino-6-[hydroxyl]phenyl-7H-pyrrolo[2.3-d]-pyrimidine), a novel EGF-R tyrosine kinase inhibitor, was synthesized and provided by Novartis Pharma (Basel, Switzerland). For oral administration (three times per week), PKI166 was dissolved in DMSO–0.5% Tween 80 and then diluted 1:20 in water (12) . The PDGF-R tyrosine kinase inhibitor STI571 (imatinib meslate, Gleevec), synthesized and provided by Novartis Pharma (Basel, Switzerland), was dissolved in distilled water at 6.25 mg/ml for daily oral administration (13) . Paclitaxel (Taxol) purchased from Bristol-Myers Squibb (Princeton, NJ) was diluted in water for once per week i.p. injection.

Three days after the implantation of tumor cells into the tibias, five mice were killed, and the presence of actively growing cancer cells was confirmed by histology. The remaining mice were randomized into eight treatment groups (n = 10) as follows: (a) control mice receiving daily administration of distilled water and one i.p. injection per week of distilled water (n = 5) or oral administration three times per week of water diluted 1:20 with DMSO–0.5% Tween 80 and a once-per-week i.p. injection of distilled water (n = 5); (b) once-per-week i.p. injection of paclitaxel at 8 mg/kg and daily oral administration of distilled water (n = 5) or oral administration three times per week of water diluted 1:20 with DMSO–0.5% Tween 80 (n = 5); (c) oral administration three times per week of 100 mg/kg PKI166 and once per week i.p. injection of distilled water; (d) daily administration of 50 mg/kg STI571 and once per week i.p. injection of distilled water; (e) oral administration three times per week of 100 mg/kg PKI166, a daily oral administration of STI571 at 50 mg/kg, and once per week i.p. injection of distilled water; (f) oral administration three times per week of 100 mg/kg PKI166 and once per week i.p. injection of paclitaxel at 8 mg/kg; (g) a daily oral administration of STI571 at 50 mg/kg and once per week i.p. injection of paclitaxel at 8 mg/kg; and (h) oral administration three times per week of 100 mg/kg PKI166, daily oral administrations of STI571 at 50 mg/kg, and once per week i.p. injection of paclitaxel at 8 mg/kg. All mice were treated for 5 weeks. Tumor size and status of injected bone (lysis) were evaluated by gross observation and digital radiography as described below.

Digital Radiography and Harvest of Tumors.

After 3 and 4 weeks of treatment, three mice selected randomly from each of the different treatment groups were anesthetized with Nembutal (0.5 mg/g body weight) and placed in a prone position. Digital radiography was carried out with a Faxitron (Faxitron X-Ray Corporation, Wheeling, IL). On week 6 of the study (5 weeks of treatment), all mice were euthanized by injection of Nembutal (1.0 mg/g) and weighed. Digital radiography was carried out on the hind limbs of each mouse, and tumor incidence and size were recorded. The tumor-bearing leg and the tumor-free contralateral leg of each mouse were resected at the head of the femur and weighed. The net tumor weight was calculated by subtraction. Macroscopically enlarged lymph nodes were harvested and prepared for histological examination to determine the presence of metastasis.

Reagents for Immunohistochemistry and Terminal Deoxynucleotidyltransferase-Mediated Nick End Labeling (TUNEL) Assay.

All antibodies for immunohistochemistry were purchased as follows: rabbit anti-EGF-R (Santa Cruz Biotechnology, Santa Cruz, CA); rabbit anti-phospho-EGF-R (activated EGF-R, Tyr845; Cell Signaling Technology, Inc., Beverly, MA); rabbit anti-PDGF-Rα and -β (Santa Cruz Biotechnology); goat polyclonal anti-phospho-PDGF-Rβ (activated PDGF-Rβ; Santa Cruz Biotechnology); rat antimouse CD31-PECAM-1 (PharMingen, San Diego, CA); mouse anti-proliferative cellular nuclear antigen (PCNA) clone PC-10 (DAKO A/S, Copenhagen, Denmark); peroxidase-conjugated goat antirabbit IgG, peroxidase-conjugated goat antirat IgG, Texas Red-conjugated goat antirat IgG, and FITC-conjugated goat antirabbit IgG (Jackson Research Laboratories, West Grove, CA); peroxidase-conjugated rat antimouse IgG2a (Serotec; Harlan Bioproducts for Science, Inc., Indianapolis, IN); and Alexa Fluor 594-conjugated goat antirabbit IgG and Alexa Fluor 594-conjugated rabbit antigoat IgG (Molecular Probes, Eugene, OR). Stable 3,3′-diaminobenzidine (Research Genetics, Huntsville, AL) and Gill’s hematoxylin (Sigma, St. Louis, MO) were used for visualization of immunohistochemical (IHC) reaction and counterstaining, respectively. TUNEL was assayed with a commercial apoptosis detection kit (Promega Corp., Madison, WI).

Preparation of Tissues.

The tibias and surrounding muscles were fixed in 10% buffered formalin for 24 h at room temperature, washed with PBS for 30 min, decalcified with 10% EDTA (pH 7.4) for 7–10 days at 4°C, sectioned, and embedded in paraffin. The method described by Morris et al. (32) was used for preparation of frozen sections with the following modifications: The tibias and surrounding muscles were fixed in 4% paraformaldehyde containing 0.075 m lysine and 0.01 m sodium periodate solution for 24 h, washed with PBS for 30 min, decalcified with 10% EDTA (pH 7.4) for 7–10 days, and washed three times, once with PBS containing 10% sucrose for 4 h, once with PBS containing 15% sucrose for 4 h, and once with PBS containing 20% sucrose for 16 h. All procedures were carried out at 4°C. The tissues were then embedded in OCT compound (Miles, Inc., Elkhart, IN), rapidly frozen in liquid nitrogen, and stored at −70°C.

IHC Determination of EGF-R, Activated EGF-R, PDGF-R, Activated PDGF-R, PCNA, and CD31/PECAM-1.

Paraffin-embedded tissues were sectioned (4- to 6-μm thick) and used to detect expression of EGF-R, activated EGF-R, PDGF-Rα and -β, activated PDGF-Rβ, and PCNA. Sections were mounted on positively charged Superfrost slides (Fisher Scientific Co., Houston, TX) and dried overnight on a hot (65°C) plate. Sections were deparaffinized in xylene, dehydrated with a graded series of alcohol [100, 95, and 80% (v/v) ethanol in doubly distilled H2O], and then rehydrated in PBS (pH 7.5). Sections analyzed for PCNA were microwaved for 5 min to improve “antigen retrieval.” Periodate-lysine-paraformaldehyde-fixed frozen tissues used for CD31/PECAM-1 were sectioned (8–10 μm), mounted on positively charged Plus slides (Fisher Scientific), and air-dried for 30 min. Frozen sections were washed three times with PBS. IHC procedures were performed as described previously (12 , 13) . Control samples exposed to secondary antibody alone showed no specific staining. Dilutions of primary antibodies were as follows: EGF-R (1:50), phosphorylated EGF-R (1:50), PDGF-Rα and -β (1:100), phosphorylated PDGF-Rβ (1:100), PCNA (1:100), and CD31/PECAM-1 (1:400). The samples for EGF-R, activated EGF-R, PDGF-Rα and -β, CD31/PECAM-1, and PCNA were incubated with protein-blocking solution containing 5% normal horse serum and 1% normal goat serum for 20 min, and the sample for activated PDGF-Rβ was incubated with protein-blocking solution containing 4% fish gel (Cold Water Fish Skin Gelatin, 40%, Aurion; Electron Microscopy Science, Fort Washington, PA) in PBS for 20 min.

We used stable 3,3′-diaminobenzidine (Research Genetics) and Gill’s hematoxylin (Sigma) for visualization of the IHC reaction with EGF-R or PDGF-Rα, PDGF-Rβ, PCNA, and CD31/PECAM-1 and counterstaining or Alexa Fluor 594-conjugated secondary antibody for immunohistochemistry of EGF-R, PDGF-R, phosphorylated EGF-R, and phosphorylated PDGF-Rβ. The sections stained with immunofluorescence-tagged secondary antibody were rinsed with distilled water and mounted with Vectashield (mounting medium with 4′,6-diamidino-2-phenylindole; Vector Laboratories, Inc., Burlingame, CA), which gave blue fluorescence nuclear staining. Immunofluorescence microscopy was performed with an ×20 or ×40 objective on an epifluorescence microscope equipped with narrow band-pass excitation filters mounted on a filter wheel (Ludl Electronic Products, Hawthorne, NY).

Distance from bone fragments was marked, and cells were counted in ×100 fields (×10 objective and ×10 ocular; 0.14 mm2/field) to calculate the percentage of cells stained positively for EGF-R or PDGF-Rα and -β (% positive cells = number of positive cells in 0.14 mm2/total number of cells in 0.14 mm2 × 100%). The intensity of staining was measured in the same fields where positivity of cells was calculated by use of ImageQuant analyzer and Optima software program (Bioscan, Edmonds, WA). Cell counting and intensity measurements were performed in six fields of each slide and analyzed statistically.

Immunofluorescence Double Staining for CD31/PECAM-1 (Endothelial Cells) and EGF-R, PDGF-R, or TUNEL.

Pyridoxal phosphate-fixed tissues were sectioned (8–10 μm), mounted on positively charged slides, and air-dried for 30 min. The samples were washed three times with PBS, incubated with protein-blocking solution containing 5% normal horse serum and 1% normal goat serum in PBS for 20 min at room temperature, and then incubated with a 1:400 dilution of rat monoclonal antimouse CD31 antibody (human cross-reactive) over 18 h at 4°C. After the samples were rinsed four times with PBS for 3 min each, the slides were incubated with a 1:200 dilution of secondary goat antirat antibody conjugated to Texas Red for 1 h at room temperature in the dark. Samples were then washed twice with PBS containing 0.1% Brij and once with PBS for 5 min. EGF-R, PDGF-Rα, and PDGF-Rβ immunostaining was performed after CD31 staining. Samples were incubated with protein-blocking solution for 5 min at room temperature and incubated with a 1:50 dilution of anti-EGF-R antibody or 1:100 dilution of anti-PDGF-Rα or -PDGF-Rβ antibody for 18 h at 4°C. The samples were then rinsed four times with PBS for 3 min each, and the slides were incubated with a 1:200 dilution of secondary goat antirabbit antibody conjugated to FITC for 1 h at room temperature. Samples were washed twice with PBS containing 0.1% Brij and once with PBS for 5 min and mounted with Vectashield. Endothelial cells were identified by red fluorescence, and EGF-R, PDGF-Rα, and PDGF-Rβ were identified by green fluorescence. Colocalization of endothelial cells/EGF-R or endothelial cells/PDGF-Rα, PDGF-Rβ (endothelial cells, red + EGF-R, PDGF-Rα, or PDGF-Rβ green = yellow) was obtained by superimposing two images. We measured the distance of endothelial cells expressing EGF-R or PDGF-Rα and -β (yellow) from the cells expressing EGF-R or PDGF-Rα and -β (green) with the Euclidian distance map (33 , 34) .

TUNEL was performed using an apoptosis detection kit with the following modification: samples were fixed with 4% paraformaldehyde (methanol free) for 10 min at room temperature, washed twice with PBS for 5 min, and then incubated with 0.2% Triton X-100 for 15 min at room temperature. After being washed twice with PBS for 5 min, the samples were incubated with equilibration buffer (from the kit) for 10 min at room temperature. The equilibrium buffer was drained, and reaction buffer containing equilibration buffer, nucleotide mixture, and terminal deoxynucleotidyl transferase enzyme was added to the tissue sections. The sections were incubated in a humidified atmosphere at 37°C for 1 h in the dark. The reaction was terminated by immersing the samples in 2× SSC for 15 min, and samples were washed three times for 5 min to remove unincorporated fluorescein-dUTP.

For quantification of endothelial cells, the samples were incubated with 300 μg/ml Hoechst stain for 10 min at room temperature. Fluorescence bleaching was minimized by treating the slides with an enhancing reagent (Prolong Solution, Prolong Anifade Kit; Molecular Probes). Immunofluorescence microscopy was performed with a ×40 objective on a Zeiss epifluorescence microscope equipped with narrow band-pass excitation filters mounted on a filter wheel (Ludl Electronic Products) to individually select for green, red, and blue fluorescence. Images were captured by use of a Sony 3-chip camera (Sony Corporation of America, Montvale, NJ) mounted on a Zeiss universal microscope (Carl Zeiss, Thornwood, NY) and Optimas Image Analysis software (Bioscan, Edmond, WA) installed on a Compaq computer with Pentium chip, a frame grabber, an optical disk storage system, and a Sony Mavigraph UP-D7000 digital color printer (Tokyo, Japan). To produce prints, images were further processed by use of Adobe PhotoShop software (Adobe Systems, Mountain View, CA). Endothelial cells were identified by red fluorescence, and DNA fragmentation was detected by localized green and yellow fluorescence within the nuclei of apoptotic cells. Apoptosis in endothelial cells was expressed as an average of the ratio of apoptotic endothelial cells to total number of endothelial cells in 5–10 random 0.011-mm2 fields at ×400 magnification.

Quantification of Mean Vessel Density and PCNA.

For quantification of microvessel density, 10 random 0.159-mm2 fields in the bone at ×100 magnification were captured for each tumor, and the vessel fields were counted according to the method described previously (35) . Cells positively stained with anti-PCNA antibody were counted in the same fields.

Statistical Analysis.

Tumor incidence (χ2 test), weight, positivity and intensity of EGF-R and PDGF-R (Mann–Whitney U test), incidence of lymph node metastasis (χ2 test), and PCNA-positive cells, mean vessel density (CD31/PECAM-1), and ratio of CD31/PECAM-1/TUNEL colocalized cells (unpaired Student’s t test) were compared.

RESULTS

Inhibition of Prostate Cancer Cell Growth and Metastasis.

We determined the therapeutic efficacy of PKI166, STI571, and paclitaxel alone or in different combinations on the growth and metastasis of human prostate cancer cells implanted in the tibias of nude mice and the bone structure. The data from two independent experiments were very similar and were therefore combined and analyzed together (Table 1) ⇓ . All control mice had large tumors in the tibias and surrounding muscles (median weight, 4.0 g; range, 2.5–6.4 g) with 100% incidence of lymph node metastasis. All mice receiving only paclitaxel also developed large tumors (median weight, 3.4 g; range, 1.2–5.0 g) with 100% incidence of lymph node metastasis. Similar to our previous reports (12 , 13) , oral administrations of PKI166 (three times per week) significantly (P < 0.05) decreased tumor incidence, tumor size, and lymph node metastasis. The daily oral administration of STI571 also significantly (P < 0.05) decreased tumor incidence, tumor size, and lymph node metastasis. Tumor incidence, size of tumor, and lymph node metastasis were decreased even more (P < 0.01) in mice receiving the combination of PKI166 and paclitaxel. Similarly, therapy with STI571 and paclitaxel significantly decreased (P < 0.01) tumor incidence, median tumor weight, and lymph node metastasis. Combination treatment with PKI166 and STI571 did not produce any additive effects over either PKI166 or STI571 administered as single agents; however, combination therapy with PKI166, STI571, and paclitaxel produced the greatest improvement (P < 0.001). Specifically, tumor incidence, median tumor weight, and incidence of lymph node metastasis were all further reduced.

View this table:
  • View inline
  • View popup
Table 1

Results of treatment with PKI166, STI571, and paclitaxel for human prostate carcinoma growing in the bones of nude mice

PC-3MM2 cells (2 × 105) were injected into the tibias of nude mice. Three days later, treatment began with paclitaxel (8 mg/kg, i.p., once/week), PKI166 (100 mg/kg, oral, 3 times/week), and STI571 (50 mg/kg, oral, daily). The mice were killed after 5 weeks of treatment. Tumor incidence, weight, and regional lymph node metastasis were determined.

Digital radiographs of representative hind legs of mice from the eight treatment groups are shown in Fig. 1 ⇓ . Control mice and mice treated with paclitaxel alone had severe lysis of the tibia. In mice given oral PKI166 or STI571, lysis of the bone was less pronounced. In mice treated with oral PKI166 and STI571, the lysis of bone was reduced. Mice treated with oral PKI166 and injectable Taxol and mice treated with oral STI571 and injectable Taxol exhibited a significant decrease in bone lysis. In mice given the combination of PKI166, STI571, and paclitaxel, the bone structure was remarkably preserved (Fig. 1) ⇓ .

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Digital radiographs of hind legs of nude mice. Nude mice implanted with human PC-3MM2 cells in the tibia received 5 weeks of treatment with single or various combinations of vehicle solution (Control), paclitaxel (8 mg/kg dose; i.p. injection once per week), PKI166 (100 mg/kg; oral administration three times per week), and STI571 (50 mg/kg; daily oral administration). The mice were then euthanized with 1 mg/g Nembutal and analyzed by digital radiography in a prone position. Tibias of mice treated with PKI166 and/or STI571 with paclitaxel were well preserved. Combination therapy of PKI166, STI571, and Taxol produced the most significant inhibition of tumor growth and the best preservation of the bone structure.

Site-Specific Expression of Growth Factors and Receptors.

PC-3MM2 prostate cancer cells can produce lysis of bone and invade the surrounding musculature where they proliferate into large lesions. In this study as well as our previous reports using PKI166 plus Taxol (12) or STI571 plus Taxol (13) , PC-3MM2 tumor cells growing in the muscle did not respond to therapy. We therefore investigated the reason that PC-3MM2 cells growing in the muscle did not respond to therapy with either PKI166 or STI571. Because targeted therapy requires that tumor cells (or endothelial cells) express a target, i.e., phosphorylated protein tyrosine kinase (34) , we determined whether the level and expression pattern of EGF-R and PDGF-R on tumor cells growing adjacent to and distant from bone tissue can provide an explanation (Fig. 2) ⇓ . The percentage of cells positive for EGF-R decreased with distance from the nearest bone tissue, from 84 ± 10% (mean ± SD) in tumors within 0.5 mm from the nearest bone fragment to 4% ± 2% in tumors 2.0–2.5 mm from nearest bone fragment (P < 0.001, Mann–Whitney U test; Table 2 ⇓ ). The intensity of staining for EGF-R was significantly decreased in cells growing more than 1.0 mm away from the nearest bone fragment (P < 0.001, Mann-Whitney U test). The percentage of positive cells stained with PDGF-R also correlated with distance from nearest bone fragment-tissue (Table 3) ⇓ . The percentage of positive tumor cells growing more than 2 mm away from the nearest bone tissue was significantly decreased (P < 0.001, Mann-Whitney U test). Staining intensity also inversely correlated with distance from the bone tissue (Table 3) ⇓ .

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Immunohistochemical analyses to determine expression of epidermal growth factor receptor (EGF-R) and platelet-derived growth factor receptor α (PDGF-Rα) and β (PDGF-Rβ) in PC-3MM2 tumor cells growing in the tibias of nude mice. Samples were stained with anti-EGF-R or anti-PDGF-Rα or -β antibodies. The distance from bone was marked, and the staining intensity in several 0.14-mm2 fields was analyzed by ImageQuant analyzer and Optima software program. The percentage of positively stained cells was calculated using the formula: % positive cells = number of positive cells/total number of cells × 100%.

View this table:
  • View inline
  • View popup
Table 2

Epidermal growth factor receptor expression on PC-3MM2 cells in bone lesions

Distance from the bone was marked, and cells were counted in multiple × 100 fields (× 10 objective and × 10 ocular; 0.14-mm2/field), and the percentage of positive cells stained with epidermal growth factor receptor was calculated. The intensity of staining was measured in the same fields by use of ImageQuant analyzer and Optima software program (Bioscan). Cell counting and intensity measurements were performed in six fields of each slide and analyzed statistically.

View this table:
  • View inline
  • View popup
Table 3

Platelet-derived growth factor receptor (PDGF-R) expression on PC-3MM2 cells in bone lesions

Distance from the bone was marked, and cells were counted in multiple × 100 fields (×10 objective and × 10 ocular; 0.14-mm2/field), and the percentage positive cells stained with PDGF-Rα or PDGF-Rβ was calculated. The intensity of staining was measured in the same fields by use of an ImageQuant analyzer and Optima software program (Bioscan). Cell counting and intensity measurements were performed in six fields of each slide and analyzed statistically.

We next examined whether the expression of EGF-R, PDGF-Rα, and PDGF-Rβ on endothelial cell surfaces correlated with expression on tumor cells. Endothelial cells expressing EGF-R on their surfaces were mostly located within 20 μm (19.6 ± 8.0 μm) of tumor cells expressing EGF-R on their surfaces (Fig. 3) ⇓ . Endothelial cells expressing PDGF-Rα or PDGF-Rβ were located within 36–37 μm (36.6 ± 11.4 and 37.4 ± 10.6 μm, respectively) of tumor cells positive for this receptor.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Double immunofluorescence staining (colocalization) of CD31/PECAM-1 and epidermal growth factor receptor (EGF-R) or platelet-derived growth factor receptor α (PDGF-Rα) or β (PDGF-Rβ). Samples were stained with anti-CD31/PECAM-1 antibody (red) and anti-EGF-R or anti-PDGF-Rα, -β (green). Colocalization of CD31/PECAM-1 and EGF-R or CD31/PECAM-1 and PDGF-R α or β is yellow. Distance was assessed with a Euclidian distance map.

Specificity of Tyrosine Kinase Inhibitors.

As shown in Fig. 4 ⇓ , PKI166 and STI571 treatments were specific for cells that expressed the corresponding receptor. Control treatments did not inhibit phosphorylation (activation) of EGF-R or PDGF-Rβ. Treatment with PKI166 inhibited phosphorylation only of EGF-R, treatment with STI571 inhibited phosphorylation only of PDGF-Rβ, and the combination of PKI166 and STI571 inhibited phosphorylation of both EGF-R and PDGF-Rβ (Fig. 4) ⇓ . Similar data were obtained with PDGF-Rα (data not shown).

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Specificity of protein tyrosine kinase inhibition. Immunohistochemical staining of phosphorylated epidermal growth factor receptor (EGF-R) or platelet-derived growth factor receptor β (PDGF-Rβ) in PC-3MM2 human prostate cancer cells growing in the tibias of nude mice. Samples were stained with anti-phospho-EGF-R or anti-phospho-PDGF-Rβ.

Cell Proliferation and Apoptosis.

We next determined the number of PCNA+ and TUNEL+ cells in PC-3MM2 tumors harvested from the tibias of control and treated mice (Table 4) ⇓ . Tumors from control mice had 130 ± 21 PCNA+ cells. Mice in all other treatment groups had significant reductions in the number of PCNA+ cells, with the combination of all three agents producing the greatest reduction (32 ± 12; P < 0.001).

View this table:
  • View inline
  • View popup
Table 4

Immunohistochemical analysis of human prostate carcinoma in bones of control and treated nude mice

Human prostate cancer cells were injected into the tibias of nude mice. Three days later, groups of mice were treated by oral administration of PKI166 (100 mg/kg, 3 times/week) and STI571 (50 mg/kg, daily), and once/week i.p. injection of paclitaxel (8 mg/kg).

Control mice had 7 ± 2 TUNEL+ cells in tumors growing adjacent to bone. Mice in all of the treatment groups had significant increases in apoptosis (TUNEL+ cells; Table 4 ⇓ ). The largest increase occurred with the combination of all three agents (83 ± 20 TUNEL+ cells; P < 0.001).

Mean Vessel Density.

In the next set of IHC studies, we determined the mean vessel density in the bone tumors as indicated by the number of CD31+ cells (Table 4) ⇓ . There were 55 ± 14 CD31+ endothelial cells in control mice. All of the various treatments produced significant reductions, with the largest decrease (to 12 ± 6) in CD31+ cells occurring in mice given a combination of all three agents (P < 0.001).

Immunofluorescence Double Staining for CD31/PECAM-1 (Endothelial Cells) and TUNEL (Apoptotic Cells).

Using the results of our previous study, we determined whether the combination of all three agents would significantly increase apoptosis in endothelial cells. The CD31/TUNEL fluorescent double-labeling technique (colocalization) revealed a median percentage of apoptotic endothelial cells in control mice of 2% (range, 0–7%). Mice in the treatment groups had median percentages of 7% (range, 0–15%) to 19% (range, 5–26%) apoptotic endothelial cells (Table 4) ⇓ .

DISCUSSION

The progressive growth of metastases is dependent on the presence of an adequate blood supply and the development of vasculature, i.e., angiogenesis is a rate-limiting factor for this process (36) . In previous studies, we reported that human prostate cancer cells growing adjacent to bone tissue express high levels of EGF and PDGF and that the tumor cells and tumor-associated endothelial cells, but not endothelial cells in uninvolved bone, expressed EGF-R and PDGF-R (12 , 13) . These data confirmed that targeting the tumor cells and the organ microenvironment can produce significant therapeutic benefits (37) . Because prostate cancer cells growing adjacent to bone tissue and endothelial cells within these lesions expressed both EGF-R and PDGF-R, it raised the question of whether simultaneous inhibition of phosphorylation of both receptors combined with systemic administration of paclitaxel could produce additive or synergistic therapeutic benefits.

In the first set of experiments, we determined the relationship of tumor cell phenotype to specific organ microenvironment. By detailed IHC analysis, we demonstrated that the expression of EGF-R and PDGF-R on the surface of human prostate cancer cells directly correlates with their proximity to bone tissue. Tumor cells that proliferate in bone tissue induce inflammatory changes. Inflammation or damage to bone tissue is associated with expression of transforming growth factor-β, which in turn induces expression of EGF (38, 39, 40) and PDGF (41 , 42) in osteoblasts and osteoclasts. The production of EGF and PDGF can up-regulate (and activate) the expression of their respective receptors in an autocrine (tumor cells) and paracrine manner (tumor-associated endothelial cells). Indeed, our present study demonstrates that endothelial cells expressing EGF-R on their surface are located within 20 μm of tumor cells expressing EGF/EGF-R. Endothelial cells expressing PDGF-Rα or -β are located within 36–37 μm of tumor cells expressing PDGF/PDGF-R. These slight differences may well be due to either the concentration of the ligands or to differences in their diffusion coefficients within solid tissues.

The expression of EGF or PDGF and their receptors has been shown to correlate with progressive growth of human carcinomas of the prostate (43 , 44) , ovary (45 , 46) , lung (47 , 48) , colon (49 , 50) , stomach (51 , 52) , and esophagus (53 , 54) , breast (55 , 56) as well as gliomas (57 , 58) and melanomas (59 , 60) ; therefore, blockade of their signaling pathways has been developed as a new therapeutic strategy (61, 62, 63, 64, 65) . Specifically, blockade of EGF-R signaling has been shown to produce arrest at the G1 restriction point (66 , 67) and to increase apoptosis (68 , 69) . Administration of STI571, which inhibits phosphorylation of PDGF-R, has been shown to inhibit depolymerization of microtubules (70 , 71) .

Whether protein tyrosine kinase inhibitors have specific, restricted activity has been controversial (61) . Our present data directly addressed this question. We report that the systemic administration of PKI166 inhibited phosphorylation of EGF-R but not PDGF-R in tumor cells and endothelial cells, whereas systemic administration of STI571 inhibited phosphorylation of PDGF-R but not EGF-R in tumor cells and endothelial cells. Systemic administration of both agents blocked the phosphorylation of both receptors.

Blockade of EGF-R or PDGF-R can alter phosphatidylinositol 3′-kinase activity, leading to a change in interstitial fluid homeostasis, which results in a reduction of interstitial hypertension with an increase in transcapillary transport (72, 73, 74) . The roles of EGF and PDGF in angiogenesis have been reported for several human tumors (75, 76, 77, 78) , and destruction of the vasculature within neoplasms can produce necrosis of actively growing tumors (79 , 80) . The induction of apoptosis in tumor-associated endothelial cells suggests that this regimen of therapy is directed against tumor vasculature.

Activation of the PDGF-R has been shown to increase expression of Bcl2 and P13K/Akt and to decrease the level of caspase in cells, i.e., increase resistance to apoptosis (81) . Because treatment of cells with STI571 increases their sensitivity to an anticycling drug, e.g., Taxol, the results support the hypothesis that PDGF could be a survival factor for tumor cells and endothelial cells. The proliferation rate of endothelial cells within the vasculature of normal organs is <0.01%, whereas 2–9% of endothelial cells in tumor-associated vessels divide daily (82) . Because the activation of the PDGF-R on tumor-associated endothelial cells can enhance their resistance to anticycling drugs (36) , the administration of protein tyrosine kinase inhibitors such as STI571 can inhibit the resistance of dividing endothelial cells to anticycling drugs, such as paclitaxel. Indeed, the oral administration of PKI166 and STI571 without Taxol completely blocked the phosphorylation of EGF-R and PDGF-R but did not produce therapeutic effects, whereas the combination of PKI166, STI571, and paclitaxel produced the best therapeutic outcome. These results support the rationale that a heterogeneous disease such as cancer should be treated with combined modalities that target both tumor cells and host microenvironment factors, such as unique vasculature and thus support the rationale that cancer (5) should be treated by combination therapy targeting both tumor cells and factors of the host microenvironment, such as the tumor vasculature.

Acknowledgments

We thank Walter Pagel for critical editorial review and Lola López for expert preparation of this manuscript.

Footnotes

  • Grant support: Cancer Center Support Core Grant CA16672 and SPORE in Prostate Cancer Grant CA90270 from the National Cancer Institute, NIH.

  • 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.

  • Requests for reprints: Isaiah J. Fidler, Department of Cancer Biology, Unit 173, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: (713) 792-8577; Fax: (713) 792-8747; E-mail: ifidler{at}mdanderson.org

  • Received December 2, 2003.
  • Revision received March 10, 2004.
  • Accepted April 9, 2004.
  • ©2004 American Association for Cancer Research.

References

  1. ↵
    Landis SH, Murray T, Bolden-Wingo PA Cancer statistics, 1999. Cancer (Phila), 49: 8-31, 1999.
  2. ↵
    Soh S, Kattan MW, Berkman S, Wheeler TM, Scardino PT Has there been a recent shift in the pathological features and prognosis of patients treated with radical prostatectomy?. J. Urol, 157: 2212-8, 1997.
    OpenUrlCrossRefPubMed
  3. ↵
    Barrettoni BA, Carter JR Mechanisms of cancer metastasis to bone. J Bone Joint Surg Am, 68A: 308-12, 1986.
    OpenUrl
  4. ↵
    Jacobs SC Spread of prostatic cancer to bone. Urology, 21: 337-44, 1983.
    OpenUrlCrossRefPubMed
  5. ↵
    Fidler IJ Critical factors in the biology of human cancer metastasis: twenty-eighth G. H. A. Clowes Memorial Award lecture. Cancer Res, 50: 6130-8, 1990.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Grignon DJ, Hammond EH College of American Pathologists Conference XXVI on Clinical Relevance of Prognostic Markers in Solid Tumors: report of the Prostate Cancer Working Group. Arch Pathol Lab Med, 119: 1122-6, 1995.
    OpenUrlPubMed
  7. ↵
    Sgrinoli AR, Walsh PC, Steinberg GD, Steiner MS, Epstein JI Prognostic factors in men with stage D1 prostate cancer: identification of patients less likely to have prolonged survival after radical prostatectomy. J Urol, 152: 1077-81, 1994.
    OpenUrlPubMed
  8. ↵
    Partin AW, Yoo J, Bellentine CH, et al The use of prostate-specific antigen, clinical stage, and Gleason score to predict pathological stage in men with localized prostate cancer. J Urol, 150: 110-4, 1994.
    OpenUrl
  9. ↵
    Fidler IJ Modulation of the organ microenvironment for the treatment of cancer metastasis. J Natl Cancer Inst (Bethesda), 87: 1588-92, 1995.
    OpenUrlFREE Full Text
  10. ↵
    Fidler IJ, Radinsky R Genetic control of cancer metastasis. J Natl Cancer Inst (Bethesda), 82: 166-8, 1990.
    OpenUrlFREE Full Text
  11. ↵
    Paget S The distribution of secondary growths in cancer of the breast. Lancet, 1: 571-3, 1889.
    OpenUrl
  12. ↵
    Kim SJ, Uehara H, Karashima T, Shepherd DL, Killion JJ, Fidler IJ Blockade of epidermal growth factor receptor signaling in tumor cells and tumor-associated endothelial cells for therapy of androgen-independent human prostate cancer growing in the bone of nude mice. Clin Cancer Res, 9: 1200-10, 2003.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Uehara H, Kim SJ, Karashima T, et al Effects of blocking platelet-derived growth factor-receptor signaling in a mouse model of experimental prostate cancer bone metastases. J Natl Cancer Inst (Bethesda), 95: 558-70, 2003.
    OpenUrl
  14. ↵
    Ulrich A, Schlessinger J Signal transduction by receptors with tyrosine kinase activity. Cell, 61: 203-12, 1990.
    OpenUrlCrossRefPubMed
  15. ↵
    Yarden Y, Ulrich A Growth factor receptor tyrosine kinases. Annu Rev Biochem, 57: 443-78, 1988.
    OpenUrlCrossRefPubMed
  16. ↵
    Wells A EGF receptor. Int J Biochem Cell Biol, 31: 637-43, 1999.
    OpenUrlCrossRefPubMed
  17. ↵
    Uhlman DL, Nguyen P, Manivel JC, et al Epidermal growth factor receptor and transforming growth factor-α expression in papillary and nonpapillary renal cell carcinoma: correlation with metastatic behavior and prognosis. Clin Cancer Res, 8: 913-20, 1995.
    OpenUrl
  18. ↵
    Atlas I, Mendelsohn J, Baselga J, Fair WR, Masui H, Kumar R Growth regulation of human renal carcinoma cells: role of transforming growth factor-α. Cancer Res, 52: 3335-9, 1992.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Moch H, Sautter G, Buchholz N, et al Epidermal growth factor receptor expression is associated with rapid tumor cell proliferation in renal cell carcinoma. Hum Pathol, 11: 1255-9, 1997.
  20. ↵
    Hofmockel G, Riess S, Bassukas ID, Dammrich J Epidermal growth factor family and renal cell carcinoma: expression and prognostic impact. Eur Urol, 4: 478-84, 1997.
    OpenUrl
  21. ↵
    Yoshida K, Tosaka A, Takeuchi S, Kobayashi N Epidermal growth factor receptor content in human renal cell carcinomas. Cancer (Phila), 73: 1913-8, 1994.
  22. ↵
    Xie J, Aszterbaum M, Zhang X, et al A role of PDGF-Rβ in basal cell carcinoma proliferation. Proc Natl Acad Sci USA, 98: 9255-9, 2001.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Funa K, Papanicolaou V, Juhlin C, et al Expression of platelet-derived growth factor alpha-receptors on stromal tissue cells in human carcinoid tumors. Cancer Res, 50: 748-53, 1990.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Liu YC, Chen SC, Chang C, Leu CM, Hu CP Platelet-derived growth factor is an autocrine stimulator for the growth and survival of human esophageal carcinoma cell lines. Exp Cell Res, 228: 206-11, 1996.
    OpenUrlCrossRefPubMed
  25. ↵
    Bornfeldt KE, Raines EW, Nakano T, Graves LM, Krebs EG, Ross R Insulin-like growth factor-I and platelet-derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J Clin Investig, 93: 1266-74, 1994.
  26. ↵
    Plate KH, Breier G, Farrell CL, Risau W Platelet-derived growth factor receptor-β is induced during tumor development and upregulated during tumor progression in endothelial cells in human gliomas. Lab Investig, 67: 529-34, 1992.
    OpenUrlPubMed
  27. ↵
    Zhang SX, Gozal D, Sachleben LR, Jr, Rane M, Klein JB, Gozal E Hypoxia induces on autocrine-paracrine survival pathway via platelet-derived growth factor (PDGF)-β/PDGF-β receptor/phosphatidylinositol 3-kinase/Akt signaling in RN46A neuronal cells. FASEB J, 17: 1709-11, 2003.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Traxler P, Bold G, Buchdunger E, et al Tyrosine kinase inhibitors: from rational design to clinical trials. Med Res Rev, 21: 499-512, 2001.
    OpenUrlCrossRefPubMed
  29. ↵
    Druker BJ, Tamura S, Buchdunger E, et al Effects of selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med, 2: 561-6, 1996.
    OpenUrlCrossRefPubMed
  30. ↵
    McCormick F New-age drug meets resistance. Nature (Lond), 412: 281-2, 2001.
    OpenUrlCrossRefPubMed
  31. ↵
    Pettaway CA, Pathak S, Greene G, et al Selection of highly metastatic variants of different human prostate carcinomas utilizing orthotopic implantation in nude mice. Clin Cancer Res, 2: 1627-36, 1996.
    OpenUrlAbstract
  32. ↵
    Morris S, Sawai T, Teshima T, Kyogoku M A new decalcifying technique for immunohistochemical studies of calcified tissue, especially applicable to cell surface marker demonstration. J Histochem Cytochem, 36: 111-4, 1988.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Russ JC . The image processing handbook, 2nd ed. p. 463-9, CRC Press Boca Raton, FL 1995.
  34. ↵
    Fidler IJ, Yano S, Zhang R, Fujimaki T, Bucana CD The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol, 3: 53-7, 2002.
    OpenUrlCrossRefPubMed
  35. ↵
    Bruns CJ, Solorzano CC, Harbison MT, et al Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res, 60: 2926-35, 2000.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Folkman MJ The role of angiogenesis in tumor growth. Semin Cancer Biol, 3: 65-71, 1992.
    OpenUrlPubMed
  37. ↵
    Baker CH, Kedar D, McCarty MF, et al Blockade of epidermal growth factor receptor signaling on tumor cells and tumor-associated endothelial cells for therapy of human carcinomas. Am J Pathol, 161: 929-38, 2002.
    OpenUrlPubMed
  38. ↵
    Hou X, Johnson AC, Rosner MR Induction of epidermal growth factor receptor gene transcription by transforming growth factor beta 1: association with loss of protein binding to a negative regulatory element. Cell Growth Differ, 5: 801-9, 1994.
    OpenUrlAbstract
  39. ↵
    Saha D, Datta PK, Sheng H, et al Synergistic induction of cyclooxygenase-2 by transforming growth factor-β1 and epidermal growth factor inhibits apoptosis in epithelial cells. Neoplasia, 1: 508-17, 1999.
    OpenUrlCrossRefPubMed
  40. ↵
    Vinals F, Pouyssegur J Transforming growth factor (TGF)-β1 promotes endothelial cell survival during in vitro angiogenesis via an autocrine mechanism implicating TGF-α signaling. Mol Cell Biol, 21: 7218-30, 2001.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Soory M, Virdi H Implications of minocycline, platelet-derived growth factor, and transforming growth factor-β on inflammatory repair potential in the periodontium. J Periodontol, 70: 1136-43, 1999.
    OpenUrlCrossRefPubMed
  42. ↵
    Seifert RA, Coats SA, Raines EW, Ross R, Bowen-Pope DF Platelet-derived growth factor (PDGF)-receptor α-subunit mutant and reconstituted cell lines demonstrate that transforming growth factor-β can be mitogenic through PDGF A-chain-dependent and -independent pathways. J Biol Chem, 269: 13951-5, 1994.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Lorenzo GD, Tortora G, D’Armiento FP, et al Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res, 8: 3438-44, 2002.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Fudge K, Bostwick DG, Stearns ME Platelet-derived growth factor A and B chains and α and β receptors in prostatic intraepithelial neoplasia. Prostate, 29: 282-6, 1996.
    OpenUrlCrossRefPubMed
  45. ↵
    Fujimura M, Hidaka T, Saito S Selective inhibition of the epidermal growth factor receptor by ZD1839 decreases the growth and invasion of ovarian clear cell adenocarcinoma cells. Clin Cancer Res, 8: 2448-54, 2002.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Henriksen R, Funa K, Wilander E, Backstrom T, Ridderheim M, Oberg K Expression and prognostic significance of platelet-derived growth factor and its receptors in epithelial ovarian neoplasms. Cancer Res, 53: 4550-4, 1993.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Mukohara T, Kudoh S, Yamauchi S, et al Expression of epidermal growth factor-receptor (EGF-R) and downstream-activated peptides in surgically excised non-small-cell lung cancer (NSCLC). Lung Cancer, 41: 123-30, 2003.
    OpenUrlPubMed
  48. ↵
    Antoniades HN, Galanopoulos T, Neville-Golden J, O’Hara CJ Malignant epithelial cells in primary human lung carcinomas coexpress in vivo platelet-derived growth factor (PDGF) and PDGF-receptor mRNAs and their protein products. Proc Natl Acad Sci USA, 89: 3942-6, 1992.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Layfield LJ, Bernard PS, Goldstein NS Color multiplex polymerase chain reaction for quantitative analysis of epidermal growth factor receptor genes in colorectal adenocarcinoma. J Surg Oncol, 83: 227-31, 2003.
    OpenUrlCrossRefPubMed
  50. ↵
    Lindmark G, Sundberg C, Glimelius B, Pahlman L, Rubin K, Gerdin B Stromal expression of platelet-derived growth factor beta-receptor and platelet-derived growth factor B-chain in colorectal cancer. Lab Investig, 69: 682-9, 1993.
    OpenUrlPubMed
  51. ↵
    Garcia I, Vizoso F, Martin A, et al Clinical significance of the epidermal growth factor receptor and HER2 receptor in resectable gastric cancer. Ann Surg Oncol, 10: 234-41, 2003.
    OpenUrlCrossRefPubMed
  52. ↵
    Chung CK, Antoniades HN Expression of c-sis/platelet-derived growth factor B, insulin-like growth factor I, and transforming growth factor-α messenger RNAs and their respective receptor messenger RNAs in primary human gastric carcinomas: in vivo studies with in situ hybridization and immunocytochemistry. Cancer Res, 52: 3453-9, 1992.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Friess H, Fukuda A, Tang WH, et al Concomitant analysis of the epidermal growth factor receptor family in esophageal cancer: overexpression of epidermal growth factor receptor mRNA but not of c-erbB-2 and c-erbB-3. World J Surg, 23: 1010-8, 1999.
    OpenUrlCrossRefPubMed
  54. ↵
    Yoshida K, Kuniyasu H, Yasui W, Kitadai Y, Toge T, Tahara E Expression of growth factors and their receptors in human esophageal carcinomas: regulation of expression by epidermal growth factor and transforming growth factor alpha. J Cancer Res Clin Oncol, 119: 401-7, 1992.
    OpenUrl
  55. ↵
    Tsutsui S, Ohno S, Murakami S, Kataoka A, Kinoshita J, Hachitanda Y Prognostic significance of the combination of biological parameters in breast cancer. Surg Today, 33: 151-4, 2003.
    OpenUrlCrossRefPubMed
  56. ↵
    Seymour L, Dajee D, Bezwoda WR Tissue platelet derived-growth factor (PDGF) predicts for shortened survival and treatment failure in advanced breast cancer. Breast Cancer Res Treat, 26: 247-52, 1993.
    OpenUrlCrossRefPubMed
  57. ↵
    Zhou YH, Tan F, Hess KR, Yung WK The Expression of PAX6, PTEN, vascular endothelial growth factor, and epidermal growth factor-receptor in gliomas: relationship to tumor grade and survival. Clin Cancer Res, 9: 3369-75, 2003.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Hermanson M, Funa K, Koopmann J, et al Association of loss of heterozygosity on chromosome 17p with high platelet-derived growth factor alpha-receptor expression in human malignant gliomas. Cancer Res, 56: 164-71, 1996.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    de Wit PE, Moretti S, Koenders PG, et al Increasing epidermal growth factor receptor expression in human melanocytic tumor progression. J Investig Dermatol, 99: 168-73, 1992.
    OpenUrlCrossRefPubMed
  60. ↵
    Palumbo C, van Roozendaal K, Gillis AJ, et al Expression of the PDGFα-receptor 1.5 kb transcript, OCT-4, and c-KIT in human normal and malignant tissues: implications for the early diagnosis of testicular germ cell tumors and for our understanding of regulatory mechanisms. J Pathol, 196: 467-77, 2002.
    OpenUrlCrossRefPubMed
  61. ↵
    Levitzki A Tyrosine kinases as targets for cancer therapy. Eur J Cancer, 38(Suppl 5): S11-8, 2002.
  62. ↵
    Laird AD, Cherrington JM Small molecule tyrosine kinase inhibitors: clinical development of anticancer agents. Expert Opin Investig Drugs, 12: 51-64, 2003.
    OpenUrlCrossRefPubMed
  63. ↵
    Ghosh S, Liu XP, Zheng Y, Uckun FM Rational design of potent and selective EGF-R tyrosine kinase inhibitors as anticancer agents. Curr Cancer Drug Targets, 1: 129-40, 2001.
    OpenUrlCrossRefPubMed
  64. ↵
    Capdeville R, Buchdunger E, Zimmermann J, Matter A Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov, 1: 493-502, 2002.
    OpenUrlCrossRefPubMed
  65. ↵
    Manley PW, Cowan-Jacob SW, Buchdunger E, et al Imatinib: a selective tyrosine kinase inhibitor. Eur J Cancer, 38(Suppl 5): S19-27, 2002.
  66. ↵
    Ulrich A, Coussens L, Hayflick JS, et al Human epidermal growth factor receptor cDNA sequence and aberrant expression of the amplified gene in A431 epidermoid carcinoma cells. Nature (Lond), 309: 418-25, 1984.
    OpenUrlCrossRefPubMed
  67. ↵
    Ciardiello F, Damiano V, Bianco R, et al Antitumor activity of combined blockade of epidermal growth factor receptor and protein kinase A. J Natl Cancer Inst (Bethesda), 88: 1770-6, 1996.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    Uckun FM, Narla RK, Zeren T, et al In vivo toxicity, pharmacokinetics, and anticancer activity of Genistein linked to recombinant human epidermal growth factor. Clin Cancer Res, 4: 1125-34, 1988.
    OpenUrl
  69. ↵
    Uckun FM, Narla RK, Jun X, et al Cytotoxic activity of epidermal growth factor-genistein against breast cancer cells. Clin Cancer Res, 4: 901-12, 1988.
    OpenUrl
  70. ↵
    Thyberg J The microtubular cytoskeleton and the initiation of DNA synthesis. Exp Cell Res, 155: 1-8, 1984.
    OpenUrlCrossRefPubMed
  71. ↵
    Yoon SY, Tefferi A, Li CY Cellular distribution of platelet-derived growth factor, transforming growth factor-β, basic fibroblast growth factor, and their receptors in normal bone marrow. Acta Haematol, 104: 151-7, 2000.
    OpenUrlCrossRefPubMed
  72. ↵
    Marcoux N, Vuori K EGF receptor mediates adhesion-dependent activation of the Rac GTPase: a role for phosphatidylinositol 3-kinase and Vav2. Oncogene, 22: 6100-6, 2003.
    OpenUrlCrossRefPubMed
  73. ↵
    Pietras K, Ostman A, Sjoquist M, et al Inhibition of platelet-derived growth factor receptors reduces interstitial hypertension and increases transcapillary transport in tumors. Cancer Res, 61: 2929-34, 2001.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    Heuchel R, Berg A, Tallquist M, et al Platelet-derived growth factor beta-receptor regulates interstitial fluid homeostasis through phosphatidylinositol-3′ kinase signaling. Proc Natl Acad Sci USA, 96: 11410-5, 1999.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    Yano S, Nishioka Y, Goto H, Sone S Molecular mechanisms of angiogenesis in non-small cell lung cancer, and therapeutics targeting related molecules. Cancer Sci, 94: 479-85, 2003.
    OpenUrlCrossRefPubMed
  76. ↵
    Suhardja A, Hoffman H Role of growth factors and their receptors in proliferation of microvascular endothelial cells. Microsc Res Tech, 60: 70-5, 2003.
    OpenUrlCrossRefPubMed
  77. ↵
    Cao R, Brakenhielm E, Li X, et al Angiogenesis stimulated by PDGF-CC, a novel member in the PDGF family, involves activation of PDGF-AA and -AB receptors. FASEB J, 16: 1575-83, 2002.
    OpenUrlAbstract/FREE Full Text
  78. ↵
    Battegay EJ, Rupp J, Iruela-Arispe L, Sage EH, Pech M PDGF-BB modulates endothelial proliferation and angiogenesis in vitro via PDGFβ-receptors. J Cell Biol, 125: 917-28, 1994.
    OpenUrlAbstract/FREE Full Text
  79. ↵
    Morioka H, Weissbach L, Vogel T, et al Anti-angiogenesis treatment combined with chemotherapy produces chondrosarcoma necrosis. Clin Cancer Res, 9: 1211-7, 2003.
    OpenUrlAbstract/FREE Full Text
  80. ↵
    Saharinen P, Alitalio K Double target for tumor mass destruction. J Clin Investig, 111: 1277-80, 2003.
    OpenUrlCrossRefPubMed
  81. ↵
    Aimakajornboon N, Szerlip NJ, Gozal E, Anonetapipat JH, Gozal D In vivo PDGFβ receptor activation in the dorsocaudal brain stem of the rat prevents hypoxia-induced apoptosis via activation of Akt and Bad. Brain Res, 895: 111-8, 2001.
    OpenUrlCrossRefPubMed
  82. ↵
    Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH, Augustin HG Heterogeneity of angiogenesis and blood vessel maturation in human tumors: implications for antiangiogenic tumor therapies. Cancer Res, 60: 1388-93, 2000.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Cancer Research: 64 (12)
June 2004
Volume 64, Issue 12
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Simultaneous Blockade of Platelet-Derived Growth Factor-Receptor and Epidermal Growth Factor-Receptor Signaling and Systemic Administration of Paclitaxel as Therapy for Human Prostate Cancer Metastasis in Bone of Nude Mice
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Simultaneous Blockade of Platelet-Derived Growth Factor-Receptor and Epidermal Growth Factor-Receptor Signaling and Systemic Administration of Paclitaxel as Therapy for Human Prostate Cancer Metastasis in Bone of Nude Mice
Sun Jin Kim, Hisanori Uehara, Sertac Yazici, Robert R. Langley, Junqin He, Rachel Tsan, Dominic Fan, Jerald J. Killion and Isaiah J. Fidler
Cancer Res June 15 2004 (64) (12) 4201-4208; DOI: 10.1158/0008-5472.CAN-03-3763

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Simultaneous Blockade of Platelet-Derived Growth Factor-Receptor and Epidermal Growth Factor-Receptor Signaling and Systemic Administration of Paclitaxel as Therapy for Human Prostate Cancer Metastasis in Bone of Nude Mice
Sun Jin Kim, Hisanori Uehara, Sertac Yazici, Robert R. Langley, Junqin He, Rachel Tsan, Dominic Fan, Jerald J. Killion and Isaiah J. Fidler
Cancer Res June 15 2004 (64) (12) 4201-4208; DOI: 10.1158/0008-5472.CAN-03-3763
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Papillomavirus E2 Protein Induces Expression of the Matrix Metalloproteinase-9 via the Extracellular Signal-Regulated Kinase/Activator Protein-1 Signaling Pathway
  • Lysyl Oxidase Regulates Breast Cancer Cell Migration and Adhesion through a Hydrogen Peroxide–Mediated Mechanism
  • Apoptotic Cells Initiate Endothelial Cell Sprouting via Electrostatic Signaling
Show more Cell and Tumor Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement