Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Res ; 64(12): 4201-8, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15205332

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-R alpha and -beta 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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , ErbB Receptors/antagonists & inhibitors , Paclitaxel/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzamides , Bone Neoplasms/metabolism , Cell Division/drug effects , Enzyme Inhibitors/administration & dosage , ErbB Receptors/metabolism , ErbB Receptors/physiology , Humans , Imatinib Mesylate , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Phosphorylation , Piperazines/administration & dosage , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/physiology , Substrate Specificity , Xenograft Model Antitumor Assays
2.
Gynecol Oncol ; 93(1): 78-86, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047217

ABSTRACT

OBJECTIVES: We determined the expression of platelet-derived growth factor (PDGF), PDGF-receptor (PDGF-R), and phosphorylated PDGF-R (p-PDGF-R) on tumor cells and tumor-associated endothelial cells in clinical specimens of human ovarian carcinoma and human ovarian cancer cells growing in culture and in the peritoneal cavity of nude mice. METHODS: Ten specimens of high-grade serous ovarian carcinoma were analyzed using immunohistochemistry (IHC). IHC was used to detect ligand and receptor expression in the human ovarian cancer cells from Hey A8 and SKOV3ip1 growing in culture. Cells from these lines were also implanted orthotopically into the peritoneal cavity of nude mice. IHC was used to determine ligand and receptor expression in tumors that formed in the peritoneal cavity. RESULTS: All 10 evaluable samples expressed both PDGF AA and BB on tumor cells. Tumor cells were positive for PDGF-Ralpha in 10/10 samples, PDGF-Rbeta in 8/10 samples, p-PDGF-Ralpha in 6/10 samples, and p-PDGF-Rbeta in 4/10 samples. p-PDGF-Ralpha was positive in 4/10 tumor-associated endothelial cell samples and p-PDGF-Rbeta was positive in 3/10 samples. Human ovarian cancer cells expressed PDGF, PDGF-R, and p-PDGF-R when growing in culture or in the peritoneal cavity of nude mice. PDGF-R and p-PDGF-R were also present on tumor-associated endothelial cells as demonstrated by simultaneous staining with CD31 antibody. CONCLUSIONS: PDGF and the corresponding receptors were expressed in autochthonous human ovarian cancer lesions on both tumor cells and tumor-associated endothelial cells. The ligand and receptor were also present on Hey A8 and SKOV3ip1 human ovarian cancer cells growing in vitro and in the peritoneal cavity of nude mice.


Subject(s)
Ovarian Neoplasms/metabolism , Platelet-Derived Growth Factor/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Animals , Becaplermin , Cell Line, Tumor , Disease Progression , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transplantation, Heterologous
3.
Clin Cancer Res ; 10(3): 897-908, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871965

ABSTRACT

PURPOSE: We sought to determine whether blockade of platelet-derived growth factor receptor (PDGF-R) activation by oral administration of a PDGF-R tyrosine kinase inhibitor (STI571) alone or in combination with i.p. paclitaxel can inhibit the progression of tumors caused by human ovarian carcinoma cells growing in the peritoneal cavity of female nude mice. EXPERIMENTAL DESIGN: In several different experiments, paclitaxel-sensitive and paclitaxel-resistant metastatic human ovarian carcinoma cells were injected into the peritoneal cavity of nude mice. Seven days later, groups (n = 10) of mice began receiving a control treatment, STI571 alone, paclitaxel alone, or a combination of STI571 and paclitaxel. The mice were necropsied after 45 days of treatment. RESULTS: Treatment with combination therapy significantly reduced tumor weight (relative to control or single-agent therapy) in all three human ovarian cancer cell lines. Immunohistochemical analyses revealed that PDGF-R activation was blocked by STI571 administered alone or in combination with paclitaxel. Tumor-associated endothelial cells expressed both PDGF-R and phosphorylated PDGF-R. In mice receiving combination therapy, tumor-associated endothelial cells underwent apoptosis, leading to decreases in microvessel density and tumor cell proliferation relative to control and single-agent therapy. CONCLUSIONS: These results show that administration of a PDGF-R tyrosine kinase inhibitor in combination with paclitaxel impairs the progression of ovarian cancer in the peritoneal cavity of nude mice, in part, by blockade of PDGF, an endothelial cell survival factor, which results in the increased apoptosis of tumor-associated endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/pathology , Receptors, Platelet-Derived Growth Factor/chemistry , Administration, Oral , Animals , Apoptosis , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Endothelium, Vascular/pathology , Enzyme Inhibitors/pharmacology , Female , Humans , Imatinib Mesylate , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , Paclitaxel/pharmacology , Phosphorylation , Piperazines/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/biosynthesis , Receptors, Platelet-Derived Growth Factor/metabolism
4.
Clin Cancer Res ; 9(17): 6534-44, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14695158

ABSTRACT

PURPOSE: We evaluated the expression of platelet-derived growth factor (PDGF) ligands and receptors in clinical specimens of human pancreatic adenocarcinomas and determined the therapeutic effect of STI571 (Gleevec), a protein tyrosine kinase inhibitor of PDGF receptor (PDGFR), on human pancreatic carcinoma cells growing in the pancreas and liver of nude mice. EXPERIMENTAL DESIGN: Immunohistochemical staining for PDGF-AA and -BB ligands, PDGFR-alpha and -beta, and phosphorylated PDGFR-alpha and -beta was performed on 31 specimens of human pancreatic cancer and L3.6pl human pancreatic adenocarcinoma cell line. To determine the in vivo effects of STI571, nude mice with L3.6pl cells injected into the pancreas were randomized 7 days later to receive one of the following treatments: sterile water p.o. (control), STI571, gemcitabine, or a combination of STI571 and gemcitabine. RESULTS: In 29 of 31 clinical specimens of human pancreatic adenocarcinoma, both tumor cells and tumor-associated endothelial cells expressed phosphorylated PDGFR-alpha and -beta. L3.6pl cells growing in culture expressed moderate amounts of PDGF-AA and little to no PDGFR-alpha or -beta, whereas L3.6pl cells growing in the pancreas of nude mice expressed a high level of PDGF and receptors. Colocalization immunohistochemical analysis demonstrated expression of activated PDGFR-beta by tumor-associated endothelial cells in both the pancreas and in liver metastases. Tumors of mice treated for 4 weeks with STI571 (50 mg/kg or 100 mg/kg p.o. daily) were slightly smaller than controls. Tumors treated with gemcitabine and STI571 (50 mg/kg) were >70% smaller than tumors in control mice and 36% smaller than those in mice treated with gemcitabine only (P < 0.0002 and P < 0.04, respectively). Combination therapy also inhibited spontaneous metastasis to the liver. Tumors from mice treated with both STI571 and gemcitabine had decreased expression of activated (phosphorylated) PDGFR-alpha and -beta, decreased mean vessel density, decreased cell proliferation, and increased apoptosis of tumor cells. CONCLUSIONS: Collectively, these data show that activated PDGFR on tumor cells and tumor-endothelial cells can be a novel target for therapy of pancreatic carcinoma.


Subject(s)
Carcinoma/pathology , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Adenocarcinoma/metabolism , Animals , Antineoplastic Agents/pharmacology , Becaplermin , Benzamides , Blotting, Western , Cell Division , Cell Line, Tumor , Humans , Imatinib Mesylate , Immunohistochemistry , In Situ Nick-End Labeling , Ligands , Mice , Mice, Nude , Microscopy, Fluorescence , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Platelet-Derived Growth Factor/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Time Factors
5.
Clin Cancer Res ; 9(5): 1858-67, 2003 May.
Article in English | MEDLINE | ID: mdl-12738744

ABSTRACT

PURPOSE: We determined whether chronic administration of IFN-alpha at optimal biological dose inhibits angiogenesis of human pancreatic carcinoma growing in the pancreas of nude mice. EXPERIMENTAL DESIGN: Cells of the human pancreatic cancer cell line L3.6pl were implanted into the pancreas of nude mice. Seven days later, groups of mice received s.c. injection with IFN-alpha alone (50,000 units biweekly or 10,000 units daily), i.p. injection with gemcitabine alone (125 mg/kg biweekly), or injection with both daily IFN-alpha and biweekly gemcitabine for 35 days. In a survival study, the mice were treated until they became moribund. RESULTS: Biweekly treatments with 50,000 units of IFN-alpha alone were ineffective. In contrast, daily injections of IFN-alpha (10,000 units/day) alone, biweekly injections of gemcitabine alone, or the combination of IFN-alpha and gemcitabine reduced tumor volume by 53%, 70%, and 87%, respectively. Immunohistochemical analysis revealed that treatment with IFN-alpha alone or with IFN-alpha plus gemcitabine inhibited expression of the proangiogenic molecules basic fibroblast growth factor and matrix metalloproteinase 9 more than did treatment with gemcitabine alone. These treatments also decreased the staining of proliferating cell nuclear antigen within the tumor and induced apoptosis in tumor-associated mouse endothelial cells (staining with CD31/terminal deoxynucleotidyl transferase-mediated nick end labeling), leading to a decrease in microvessel density. CONCLUSIONS: These data show that administration of IFN-alpha at optimal biological dose and schedule in combination with gemcitabine induced apoptosis in tumor-associated endothelial cells and decreased growth of human pancreatic cancer cells in the pancreas, leading to a significant increase in survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Endothelium, Vascular/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Cell Division/drug effects , Deoxycytidine/administration & dosage , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Implants, Experimental , Interferon-alpha/administration & dosage , Interleukin-8/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental , Pancreatic Neoplasms/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Survival Rate , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Gemcitabine
6.
Clin Cancer Res ; 9(3): 1200-10, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631626

ABSTRACT

PURPOSE: We determined whether blockade of the epidermal growth factor receptor (EGF-R) signaling pathway by oral administration of the EGF-R tyrosine kinase inhibitor (PKI 166) alone or in combination with injectable Taxol inhibits the growth of PC-3MM2 human prostate cancer cells in the bone of nude mice. EXPERIMENTAL DESIGN: Male nude mice implanted with PC-3MM2 cells in the tibia were treated with oral administrations of PKI 166 or PKI 166 plus injectable Taxol beginning 3 days after implantation. The incidence and size of bone tumors and destruction of bone were determined by digitalized radiography. Expression of epidermal growth factor (EGF), EGF-R, and activated EGF-R in tumor cells and tumor-associated endothelial cells was determined by immunohistochemistry. RESULTS: Oral administration of PKI 166 or PKI 166 plus injectable Taxol reduced the incidence and size of bone tumors and destruction of bone. Immunohistochemical analysis revealed that PC-3MM2 cells growing adjacent to the bone expressed high levels of EGF and activated EGF-R, whereas tumor cells in the adjacent musculature did not. Moreover, endothelial cells within the bone tumor lesions, but not in uninvolved bone or tumors in the muscle, expressed high levels of activated EGF-R. Treatment with PKI 166 and more so with PKI 166 plus Taxol significantly inhibited phosphorylation of EGF-R on tumor and endothelial cells and induced significant apoptosis and endothelial cells within tumor lesions. CONCLUSIONS: These data indicate that endothelial cells exposed to EGF produced by tumor cells express activated EGF-R and that targeting EGF-R can produce significant therapeutic effects against prostate cancer bone metastasis.


Subject(s)
ErbB Receptors/metabolism , Signal Transduction , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Bone Neoplasms/pathology , Bone and Bones/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/biosynthesis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , Fibroblast Growth Factor 2/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/biosynthesis , Interleukin-8/biosynthesis , Lymphokines/biosynthesis , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Metastasis , Neoplasm Transplantation , Paclitaxel/pharmacology , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Proliferating Cell Nuclear Antigen/biosynthesis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
J Natl Cancer Inst ; 95(6): 458-70, 2003 Mar 19.
Article in English | MEDLINE | ID: mdl-12644539

ABSTRACT

BACKGROUND: Expression of platelet-derived growth factor (PDGF) and activation (by autophosphorylation) of its receptor (PDGF-R), a tyrosine kinase, are associated with the growth of metastatic prostate tumor cells in the bone parenchyma. The tyrosine kinase inhibitor STI571 blocks the PDGF signaling pathway by inhibiting PDGF-R autophosphorylation. We examined the effects of STI571, given alone or with paclitaxel (Taxol), on tumor growth in a mouse model of prostate cancer metastasis. METHODS: Human prostate cancer PC-3MM2 cells were injected into the tibias of male nude mice. Three days later the mice (20 per group) were randomly assigned to 5 weeks of treatment with oral and injected water (control), daily oral STI571, weekly injected paclitaxel, or STI571 plus paclitaxel. Lesions in bone and the surrounding muscles were then harvested and analyzed by histology, western blotting (for PDGF-R phosphorylation), immunohistochemistry (for expression of proangiogenic molecules), and double immunofluorescence (to identify endothelial cells and apoptotic tumor cells). Growth of bone lesions was monitored by digital radiography. Bone lesions from control mice were used to establish short-term cell cultures for analysis of PDGF-R phosphorylation. All statistical tests were two-sided. RESULTS: PC-3MM2 cells cultured from bone lesions and treated in vitro with STI571 had less phosphorylated PDGF-R than untreated cells. In control mice, bone lesions expressed high levels of PDGF and activated (i.e., phosphorylated) PDGF-R, whereas lesions in the adjacent musculature did not. Activated PDGF-R was present on the surface of endothelial cells within the bone lesions but not in endothelial cells of uninjected bone. Mice treated with STI571 or STI571 plus paclitaxel had a lower tumor incidence, smaller tumors, and less bone lysis and lymph node metastasis than mice treated with water or paclitaxel alone (P<.001 for all). Mice treated with STI571 or STI571 plus paclitaxel had less phosphorylated PDGF-R on tumor cells and tumor-associated endothelial cells, less tumor cell proliferation, statistically significantly more apoptotic tumor cells (all P<.001), and fewer tumor-associated endothelial cells (P<.001) than control mice. CONCLUSIONS: Endothelial cells appear to express phosphorylated PDGF-R when they are exposed to tumor cells that express PDGF. Using STI571 to inhibit PDGF-R phosphorylation may, especially in combination with paclitaxel, produce substantial therapeutic effects against prostate cancer bone metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Platelet-Derived Growth Factor/metabolism , Prostatic Neoplasms/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzamides , Blotting, Western , Bone Neoplasms/blood supply , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/enzymology , Bone Neoplasms/secondary , Cell Division/drug effects , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Microcirculation/drug effects , Neoplasms, Experimental , Paclitaxel/administration & dosage , Phosphorylation/drug effects , Piperazines/administration & dosage , Platelet-Derived Growth Factor/drug effects , Prostatic Neoplasms/metabolism , Pyrimidines/administration & dosage , Radiographic Image Enhancement , Receptors, Platelet-Derived Growth Factor/drug effects , Tumor Cells, Cultured
8.
Clin Cancer Res ; 8(11): 3592-600, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429651

ABSTRACT

We determined whether blockade of the epidermal growth factor-receptor (EGF-R) signaling pathway by oral administration of the EGF-R tyrosine kinase inhibitor PKI166 can inhibit angiogenesis and growth of SN12PM6 human renal cell carcinoma (HRCC) in the kidney of nude mice and whether gemcitabine can potentiate these effects. In vitro treatment of HRCC cells with PKI166 inhibited EGF-R autophosphorylation, which correlated with a decrease in expression of Bcl-xl protein and phosphorylation of signal transducers and activators of transcription, particularly signal transducers and activators of transcription 3. PKI166 also decreased expression of vascular endothelial growth factor and basic fibroblast growth factor in a dose-dependent manner. Oral administration of PKI166 or PKI166 and injected gemcitabine or gemcitabine alone beginning 7 days after implantation of SN12PM6 cells into the kidney of athymic nude mice reduced the volume of tumors by 26, 61, and 23%, respectively. In another experiment 28 days after the orthotopic implantation of SN12PM6 cells, nephrectomy was performed followed by 4 weeks of treatment. Treatment with PKI166 and, more so, PKI166 plus gemcitabine significantly inhibited lung metastasis, corresponding to a significant increase in overall length of survival. EGF-R activation was significantly blocked by therapy with PKI166 and was associated with a significant reduction in expression of vascular endothelial growth factor and interleukin-8, decreased microvessel density, decreased staining of proliferating cell nuclear antigen, and increased tumor cell apoptosis. Collectively, the data indicate that targeting activation of EGF-R on HRCC produces significant therapeutic benefits.


Subject(s)
Carcinoma, Renal Cell/pathology , Deoxycytidine/analogs & derivatives , ErbB Receptors/antagonists & inhibitors , Neovascularization, Pathologic , Signal Transduction , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , DNA-Binding Proteins/metabolism , Deoxycytidine/pharmacology , Down-Regulation , Endothelial Growth Factors/metabolism , Enzyme Inhibitors/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/metabolism , Kidney/pathology , Lung/pathology , Lymphokines/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor , Time Factors , Trans-Activators/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Gemcitabine
9.
Cancer Res ; 62(20): 5720-6, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384530

ABSTRACT

We evaluated whether treatment of orthotopic human prostate cancer in nude mice with pegylated IFN-alpha-2b (PEG-IFN-alpha-2b) and docetaxel could represent a two-compartment targeting of primary tumor (tumor cells and tumor-associated endothelial cells) and inhibition of regional lymph node metastasis. The antiangiogenic properties of IFN were combined with the cytotoxic properties of docetaxel, resulting in apoptosis of both tumor cells and endothelium and hence significant inhibition of primary tumor growth. We first determined the optimal biological dose of PEG-IFN-alpha-2b (70,000 IU/week) necessary to down-regulate the expression of basic fibroblast growth factor, matrix metalloprotease-9, and matrix metalloprotease-2. The therapeutic dose of docetaxel (10 mg/kg/week) was determined by efficacy and minimal body weight loss. Therapy beginning 3 days after orthotopic implantation of PC3-MM2 prostate cancer cells reduced tumor weight by 37% in mice treated with PEG-IFN-alpha-2b, by 60% in mice treated with docetaxel, and by 83% in those given both drugs. PEG-IFN-alpha-2b also induced apoptosis of tumor-associated endothelial cells and hence a significant decrease in microvessel density. Our data indicate that the combination of PEG-IFN-alpha and docetaxel inhibits neoplastic angiogenesis by inducing a decrease in the local production of proangiogenic molecules by tumor cells, resulting in increased apoptosis of tumor-associated endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interferon-alpha/pharmacology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Polyethylene Glycols/pharmacology , Prostatic Neoplasms/drug therapy , Taxoids , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cadherins/biosynthesis , Cell Division/drug effects , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Fibroblast Growth Factor 2/biosynthesis , Humans , Immunohistochemistry , Interferon alpha-2 , Interferon-alpha/administration & dosage , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Nude , Neoplasm Metastasis/prevention & control , Paclitaxel/administration & dosage , Polyethylene Glycols/administration & dosage , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Random Allocation , Recombinant Proteins , Xenograft Model Antitumor Assays
10.
Arch Otolaryngol Head Neck Surg ; 128(8): 875-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12162763

ABSTRACT

BACKGROUND: Overexpression of epidermal growth factor receptor (EGF-R) is associated with increased malignant potential and correlates with poor clinical outcome in head and neck cancer. Therefore, inhibition of the EGF-R pathway provides an ideal target for molecular therapy. We examined in vitro and in vivo effects of PKI166, an orally administered EGF-R inhibitor, on 2 human squamous cell carcinoma of the oral cavity cell lines, Tu159 and MDA1986. STUDY DESIGN: Basic science, laboratory investigation. RESULTS: For Western blotting, Tu159 and MDA1986 cells were pretreated for 1 hour and then stimulated with EGF. The EGF-R-specific tyrosine kinase autophosphorylation was inhibited completely by PKI166 at all doses tested (1-10 micro g/mL). By means of a tetrazolium-based viable cell assay, PKI166 was shown to arrest the growth of Tu159 and MDA1986 cells. The inhibitory concentration (50%), calculated from regression lines on the linear portion of the growth inhibition graphs, was 0.18 micro M (R = 0.98) for Tu159 cells and 0.23 micro M (R = 0.97) for MDA1986 cells. Nude mice were inoculated subcutaneously with 1 x 10(6) Tu159 tumor cells and observed for 7 days. Next, daily doses of PKI166 (0, 10, or 50 mg/kg) were delivered by orogastric lavage for 28 days and the animals were observed for tumor growth. PKI166 significantly reduced tumor growth in mice treated for 1 month with oral PKI166 in a dose-dependent fashion. CONCLUSIONS: Targeted molecular therapy with EGF-R blockade arrests the growth of oral cancer in vitro and reduces its proliferation in an experimental xenograft animal model.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/therapeutic use , Gene Targeting , Genetic Therapy , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , ErbB Receptors/genetics , Genes, erbB-1/genetics , Humans , In Vitro Techniques , Male , Mice , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured/drug effects
11.
Clin Cancer Res ; 8(7): 2413-22, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12114447

ABSTRACT

The purpose of this study was to optimize the antitumor andantiangiogenic activities of pegylated IFN-alpha (PEG-IFN-alpha)alone or in combination with paclitaxel against SKOV3ip1 human ovarian cancer cells growing orthotopically in female nude mice. Seven days after the i.p. implantation of tumor cells, groups of mice (n = 10) were injected s.c. once per week (for 4 weeks) with different doses of PEG-IFN-alpha (3,500, 7,000, 35,000, and 350,000 units). PEG-IFN-alpha at 7,000 units significantly decreased tumor incidence and volume. At doses exceeding 7,000 units, PEG-IFN-alpha was less efficacious. In another set of studies conducted 7 days after the i.p. implantation of SKOV3ip1 cells, groups of mice (n = 10) received (once per week for 4 weeks) either s.c. administrations of PEG-IFN-alpha (7,000 units), i.p. injections of paclitaxel (100 microg/wk), or a combination of PEG-IFN-alpha and paclitaxel. The mice were killed 7 days after the last treatment, and tumor burden was assessed. Administration of PEG-IFN-alpha at the optimal biological dose (7,000 units) in combination with paclitaxel significantly decreased angiogenesis and progressive growth of human ovarian carcinoma cells in a synergistic fashion. The combination therapy produced the most significant inhibition in expression of the proangiogenic molecules basic fibroblast growth factor and matrix metalloproteinase-9. Decreased microvessel density, decreased proliferating cell nuclear antigen staining, and increased endothelial cell apoptosis also correlated with therapeutic success. Collectively, the data suggest that combining the optimal biological dose of PEG-IFN-alpha with paclitaxel may provide a novel and effective approach to the treatment of human ovarian carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon-alpha , Ovarian Neoplasms/drug therapy , Polyethylene Glycols , Animals , Apoptosis/drug effects , Blotting, Northern , Cell Division/drug effects , Drug Synergism , Endothelial Growth Factors/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins/metabolism , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interleukin-8/metabolism , Lymphokines/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Recombinant Proteins , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...