Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Transl Oncol ; 5(1): 39-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22348175

ABSTRACT

Endothelin receptors (ETRs) are often overexpressed in ovarian tumors, which can be resistant to conventional therapies. Thus, we investigated whether blockage of the ETR pathways using the dual ETR antagonist macitentan combined with taxol or cisplatinum can produce therapy for orthotopically growing multidrug-resistant (MDR) human ovarian carcinoma. In several studies, nude mice were injected in the peritoneal cavity with HeyA8-MDR human ovarian cancer cells. Ten days later, mice were randomized to receive vehicle (saline), macitentan (oral, daily), taxol (intraperitoneal, weekly), cisplatinum (intraperitoneal, weekly), macitentan plus taxol, or macitentan plus cisplatinum. Moribund mice were killed, and tumors were collected, weighed, and prepared for immunohistochemical analysis. The HeyA8-MDR tumors did not respond to taxol, cisplatinum, or macitentan administered as single agents. In contrast, combination therapy with macitentan and taxol or macitentan and cisplatinum significantly decreased the tumor incidence and weight and significantly increased the survival of mice and their general condition. Multiple immunohistochemical analyses revealed that treatment with macitentan and macitentan plus taxol or cisplatinum inhibited the phosphorylation of ETRs, decreased the levels of pVEGFR2, pAkt, and pMAPK in tumor cells after 2 weeks of treatment and induced a first wave of apoptosis in tumor-associated endothelial cells followed by apoptosis in surrounding tumor cells. Our study shows that ovarian cancer cells, which express the endothelin axis and are multidrug resistant, are exquisitely sensitive to treatment with a dual ET antagonist and can be resensitized to both taxol and cisplatinum. This combined therapy led to a significant reduction in tumor weight.

2.
Clin Cancer Res ; 17(13): 4331-40, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622717

ABSTRACT

PURPOSE: There is growing evidence implicating that neutrophil gelatinase-associated lipocalin (NGAL) plays a role in the development and progression of cancers. However, the effect of NGAL in colorectal carcinoma (CRC) has not been clearly elucidated. In this study, we investigated the role of NGAL in the tumorigenesis and progression of CRC and evaluated the clinical value of NGAL expression. EXPERIMENTAL DESIGN: We examined NGAL expression in 526 colorectal tissue samples, including 53 sets of matched specimens (histologically normal mucosa, adenomas, and carcinomas) using immunohistochemical analysis. In CRCs, correlations between NGAL expression and clinicopathologic parameters were analyzed, and survival analysis was conducted. The role of NGAL was further tested using mouse xenograft models. RESULTS: NGAL expression was elevated during the colorectal adenoma-carcinoma sequence both among the 526 cases (r(s) = 0.66, P < 0.001) and in the 53 sets of matched specimens (r(s) = 0.60, P < 0.001). In CRCs, NGAL expression was associated with cancer stage (P = 0.041) and tumor recurrence in stage II patients (P = 0.037). Survival analysis revealed that NGAL expression was an independent prognostic factor for overall survival (HR = 1.84, P = 0.004) and for disease-free survival of stage II patients (HR = 5.88, P = 0.021). In mouse models, the xenografts in cecum and spleen were heavier and more numerous in the group injected with NGAL-overexpressing CRC cells (P < 0.05). CONCLUSIONS: NGAL overexpression may promote the tumorigenesis and progression of CRC. Detecting NGAL expression in tumor tissues may be useful for evaluating prognosis of patients with CRC.


Subject(s)
Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Adenoma/genetics , Adenoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Lipocalins/genetics , Lipocalins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Adenoma/diagnosis , Adenoma/mortality , Animals , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/mortality , Disease Progression , Gene Expression , Humans , Lipocalin-2 , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Staging , Prognosis , Recurrence , Survival Analysis , Xenograft Model Antitumor Assays
3.
Transl Oncol ; 4(2): 83-91, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21461171

ABSTRACT

There is an urgent need to develop novel markers of pancreatic cancer to facilitate early diagnosis. Pancreatic carcinoma is characterized by marked stroma formation with a high number of infiltrating tumor-associated macrophages (TAMs) that originate from circulating mononuclear cells (MNCs). We hypothesized that differential analysis of protein expression and phosphorylation in circulating MNCs from healthy nude mice and nude mice bearing orthotopic human pancreatic cancer would identify a surrogate marker of pancreatic cancer. These differences were analyzed by two-dimensional gel electrophoresis followed by Western blot analysis using antibody against phosphorylated tyrosine proteins (pY). Protein and phosphorylated protein spots of interest were identified by mass spectrometry and validated by Western blot analysis as candidate markers for pancreatic cancer. We found that the expression and phosphorylation of Src family proteins were significantly higher in circulating MNCs from mice bearing pancreatic cancer than in circulating MNCs from healthy mice. TAMs in mice with pancreatic tumors also had higher Src family protein expression and phosphorylation than resident macrophages in the pancreas of healthy mice. The expression and phosphorylation of Src family proteins were correlated with tumor weight; however, increased Src expression and phosphorylation also occurred in MNCs from mice with chronic pancreatitis. This is the first report to explore novel pancreatic tumor markers in circulating MNCs. Although the specificity of the marker for pancreatic cancer was low, it could be used to monitor the disease or to select high-risk patients with chronic pancreatitis.

4.
Am J Pathol ; 178(5): 2357-66, 2011 May.
Article in English | MEDLINE | ID: mdl-21514446

ABSTRACT

An approach that facilitates rapid isolation and characterization of tumor cells with enhanced metastatic potential is highly desirable. Here, we demonstrate that plating GI-101A human breast cancer cells on hard (0.9%) agar selects for the subpopulation of metastasis-initiating cells. The agar-selected cells, designated GI-AGR, were homogeneous for CD44(+) and CD133(+) and five times more invasive than the parental GI-101A cells. Moreover, mice injected with GI-AGR cells had significantly more experimental brain metastases and shorter overall survival than did mice injected with GI-101A cells. Comparative gene expression analysis revealed that GI-AGR cells were markedly distinct from the parental cells but shared an overlapping pattern of gene expression with the GI-101A subline GI-BRN, which was generated by repeated in vivo recycling of GI-101A cells in an experimental brain metastasis model. Data mining on 216 genes shared between GI-AGR and GI-BRN breast cancer cells suggested that the molecular phenotype of these cells is consistent with that of cancer stem cells and the aggressive basal subtype of breast cancer. Collectively, these results demonstrate that analysis of cell growth in a hard agar assay is a powerful tool for selecting metastasis-initiating cells in a heterogeneous population of breast cancer cells, and that such selected cells have properties similar to those of tumor cells that are selected based on their potential to form metastases in mice.


Subject(s)
Breast Neoplasms/pathology , Cell Culture Techniques/methods , Neoplasm Metastasis/pathology , Neoplastic Stem Cells , Tumor Cells, Cultured/pathology , Agar , Animals , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Cell Proliferation , Female , Gene Expression , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Neoplasia ; 13(3): 286-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21390191

ABSTRACT

In the United States, more than 40% of cancer patients develop brain metastasis. The median survival for untreated patients is 1 to 2 months, which may be extended to 6 months with conventional radiotherapy and chemotherapy. The growth and survival of metastasis depend on the interaction of tumor cells with host factors in the organ microenvironment. Brain metastases are surrounded and infiltrated by activated astrocytes and are highly resistant to chemotherapy. We report here that coculture of human breast cancer cells or lung cancer cells with murine astrocytes (but not murine fibroblasts) led to the up-regulation of survival genes, including GSTA5, BCL2L1, and TWIST1, in the tumor cells. The degree of up-regulation directly correlated with increased resistance to all tested chemotherapeutic agents. We further show that the up-regulation of the survival genes and consequent resistance are dependent on the direct contact between the astrocytes and tumor cells through gap junctions and are therefore transient. Knocking down these genes with specific small interfering RNA rendered the tumor cells sensitive to chemotherapeutic agents. These data clearly demonstrate that host cells in the microenvironment influence the biologic behavior of tumor cells and reinforce the contention that the organ microenvironment must be taken into consideration during the design of therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytes/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Astrocytes/pathology , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Profiling , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Up-Regulation , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
6.
Cancer ; 117(17): 4092-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21365635

ABSTRACT

BACKGROUND: Large multinucleated cells (MNCs) commonly exist in tumorigenic cancer cell lines that are used widely in research. However, the contributions of MNCs to tumorigenesis are unknown. METHODS: In this study, MNCs were characterized in the murine fibrosarcoma cell line UV-2237 in vitro and in vivo at the single-cell level. RESULTS: The authors observed that MNCs originated from a rare subpopulation of mononuclear cells and were positive for a senescent marker, ß-galactosidase. In addition, MNCs were responsible for the majority of clonogenic activity when cultured in hard agar; they were more resistant to chemotherapeutic agents than mononuclear cells; they could undergo asymmetric division (producing mononuclear cells) and self-renewal in vitro and in vivo; and, most important; a single MNC produced orthotopic, subcutaneous tumors (composed mainly of mononuclear cells) that gave rise to spontaneous lung metastases in nude mice. CONCLUSIONS: The current results indicated that the growth of MNCs may be arrested under stress and that MNCs are highly resistant to chemotherapy and can generate clonal, orthotopic, metastatic tumors.


Subject(s)
Fibrosarcoma/pathology , Giant Cells/pathology , Neoplastic Stem Cells/pathology , Skin Neoplasms/etiology , Animals , Cell Line, Tumor , Cellular Senescence , Clone Cells , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasms/pathology , beta-Galactosidase/metabolism
7.
Int Immunopharmacol ; 11(7): 862-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21251905

ABSTRACT

To test the hypothesis that tumor-associated macrophages (TAMs) enhance the growth and metastasis of human prostate cancer in the bone, we evaluated the effects of decreasing interleukin-6 (IL-6) production by tumor cells and TAMs in a mouse model of bone metastasis. Human PC-3MM2 cells that produce IL-6 were transfected with lentivirus containing IL-6 small hairpin RNA (shRNA) or nonspecific RNA and injected into the tibias of nude mice treated intraperitoneally every 5days for 5weeks with phosphate-buffered saline (PBS), liposomes containing PBS, or liposomes containing clodronate (to decrease the number of macrophages). Transfection of PC-3MM2 cells with IL-6 shRNA significantly decreased cellular expression of IL-6 and the number of TAMs and osteoclasts in bone tumors, which correlated with significant decreases in tumor size, bone lysis, and incidence of lymph node metastasis. Treatment of mice with clodronate liposomes significantly decreased the number of TAMs and osteoclasts in the bone tumors, the expression of IL-6 in the PC3-MM2 cells, and the production of tumor necrosis factor (TNF)-α by TAMs. These findings correlated with a significant decrease in tumor size, bone lysis, and lymph node metastasis. Knocking down IL-6 in tumor cells and decreasing TAMs was associated with the lowest incidences of bone tumors and lymph node metastasis. These results suggest that TAMs enhance the growth of prostate cancer cells in the bone.


Subject(s)
Bone Neoplasms/immunology , Carcinoma/immunology , Macrophages/metabolism , Osteoclasts/metabolism , Prostatic Neoplasms/immunology , Animals , Bone Neoplasms/secondary , Carcinoma/secondary , Cell Communication/drug effects , Cell Communication/genetics , Cell Communication/immunology , Cell Line, Tumor , Clodronic Acid/administration & dosage , Clodronic Acid/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Liposomes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Nude , Osteoclasts/immunology , Osteoclasts/pathology , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
Transl Oncol ; 3(6): 380-8, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21151477

ABSTRACT

Experimental metastases in the brain of mice are infiltrated by microglia, and parabiosis experiments of green fluorescent protein (GFP(+)) and GFP(-) mice revealed that these microglia are derived from circulating monocytes (GFP(+), F4/80(+), and CD68(+)). These findings raised the question as to whether microglia (specialized macrophages) possess tumoricidal activity. C8-B4 murine microglia cells were incubated in vitro in medium (control) or in medium containing both lipopolysaccharide and interferon-γ. Control microglia were not tumoricidal against a number of murine and human tumor cells, whereas lipopolysaccharide/interferon-γ-activated microglia lysed murine and human tumor cells by release of nitric oxide. Parallel experiments with murine peritoneal macrophages produced identical results. Neither activated microglia nor activated macrophages lysed nontumorigenic murine or human cells. Collectively, these data demonstrate that brain metastasis-associated microglia are derived from circulating mononuclear cells and exhibit selective and specific tumoricidal activity.

9.
Neoplasia ; 12(9): 748-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20824051

ABSTRACT

Brain metastases are highly resistant to chemotherapy. Metastatic tumor cells are known to exploit the host microenvironment for their growth and survival. We report here that melanoma brain metastases are surrounded and infiltrated by activated astrocytes, and we hypothesized that these astrocytes can play a role similar to their established ability to protect neurons from apoptosis. In coculture experiments, astrocytes, but not fibroblasts, reduced apoptosis in human melanoma cells treated with various chemotherapeutic drugs. This chemoprotective effect was dependent on physical contact and gap junctional communication between astrocytes and tumor cells. Moreover, the protective effect of astrocytes resulted from their sequestering calcium from the cytoplasm of tumor cells. These data suggest that brain tumors can, in principle, harness the neuroprotective effects of reactive astrocytes for their own survival and implicate a heretofore unrecognized mechanism for resistance in brain metastasis that might be of relevance in the clinic.


Subject(s)
Antineoplastic Agents/therapeutic use , Astrocytes/physiology , Calcium/metabolism , Connexins/physiology , Drug Resistance, Neoplasm , Animals , Antineoplastic Agents/pharmacology , Astrocytes/metabolism , Cell Communication/physiology , Cell Line, Tumor , Cells, Cultured , Connexins/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/physiology , Humans , Intracellular Fluid/metabolism , Male , Mice , Mice, Nude , Mice, Transgenic , Models, Biological , NIH 3T3 Cells
10.
Neoplasia ; 12(2): 173-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20126475

ABSTRACT

The lymphatic system plays a critical role in melanoma metastasis, and yet, virtually no information exists regarding the cellular and molecular mechanisms that take place between melanoma cells and the lymphatic vasculature. Here, we generated B16-F1 melanoma cells that expressed high (B16alpha(4)+) and negligible (B16alpha(4)-) levels of alpha(4) integrin to determine how the expression of alpha(4) integrins affects tumor cell interactions with lymphatic endothelial cells in vitro and how it impacts lymphatic metastasis in vivo. We found a direct correlation between alpha(4) integrin expression on B16-F1 melanoma cells and their ability to form adhesive interactions with monolayers of lymphatic endothelial cells. Adhesion of B16-F1 melanoma cells to lymphatic endothelial cells was mediated by the melanoma cell alpha(4) integrin binding to its counterreceptor, vascular cell adhesion molecule 1 (VCAM-1), that was constitutively expressed on the lymphatic endothelial cells. VCAM-1 was also expressed on the tumor-associated lymphatic vessels of B16-F1 and B16alpha(4)+ tumors growing in the subcutaneous space of C57BL/6J mice. B16-F1 tumors metastasized to lymph nodes in 30% of mice, whereas B16alpha(4)+ tumors generated lymph node metastases in 80% of mice. B16-F1 melanoma cells that were deficient in alpha(4) integrins (B16alpha(4)-) were nontumorigenic. Collectively, these data show that the alpha(4) integrin expressed by melanoma cells contributes to tumorigenesis and may also facilitate metastasis to regional lymph nodes by promoting stable adhesion of melanoma cells to the lymphatic vasculature.


Subject(s)
Endothelial Cells/metabolism , Integrin alpha4/biosynthesis , Lymphatic Metastasis/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Animals , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Fluorescent Antibody Technique , Lymphatic Metastasis/genetics , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Vascular Cell Adhesion Molecule-1/metabolism
11.
Cancer ; 116(1): 184-92, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19885836

ABSTRACT

BACKGROUND: The authors sought to determine the incidence and severity of cardiovascular toxicity caused by imatinib mesylate in gastrointestinal stromal tumor (GIST) and other sarcoma patients, and to explore cardiotoxicity caused by imatinib mesylate using cell culture and in vitro models. METHODS: To determine the incidence and significance of serious cardiac adverse events in GIST and other sarcoma patients receiving imatinib mesylate, the authors performed a retrospective analysis of 219 consecutive patients treated with imatinib mesylate. In vitro studies of imatinib mesylate on cultured cardiomyocytes and biochemical studies of cardiac lysates from mice treated with imatinib mesylate were performed to define the potential cardiotoxic effects of imatinib mesylate. RESULTS: Grade 3 or 4 potentially cardiotoxic adverse events (mostly edema or effusions) occurred in 8.2% of patients, were manageable with medical therapy, and infrequently required dose reduction or discontinuation of imatinib mesylate. Arrhythmias, acute coronary syndromes, or heart failure were uncommon, occurring in <1% of treated patients. However, administration of imatinib in a mouse model system resulted in inhibition of activation of protein kinases that are known to be important in the cardiac stress response. CONCLUSIONS: The authors concluded that imatinib is an uncommon cause of cardiotoxicity, and that the cardiovascular adverse events that occur are manageable when recognized and treated. Nevertheless, our preclinical findings suggest that imatinib remains a potential cardiotoxin. Furthermore, the cardiac consequences of long-term imatinib therapy remain unknown. We therefore recommend treatment of risk factors for cardiovascular disease in imatinib-treated patients in accord with the American Heart Association guidelines for the prevention and treatment of heart failure.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Heart Failure/chemically induced , Piperazines/adverse effects , Pyrimidines/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Benzamides , Cells, Cultured , Echocardiography , Female , Humans , Imatinib Mesylate , Incidence , Male , Middle Aged , Myocytes, Cardiac/drug effects , Young Adult
12.
Int J Oncol ; 35(4): 665-72, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19724901

ABSTRACT

Astrocytes play a critical role in maintaining cerebral homeostasis and their dysregulation is thought to contribute to the pathogenesis of several diseases, including brain cancer and metastasis. Similar to the human disease, we found that lung and melanoma metastases in the mouse brain are accompanied by a reactive gliosis. To begin to study the biology of astrocytes and examine how these cells might contribute to metastasis formation and progression in the brain, we generated a conditionally immortal astrocyte cell line from H-2Kb-tsA58 mice. Astrocytes grown in culture expressed glial fibrillary acid protein (GFAP), glutamate receptor 1, and the N-methyl-D-aspartate (NMDA) receptor. Astrocytes also expressed the glial-specific transporters excitatory amino acid transporter 1 (EAAT1) and EAAT2. Astrocytes grown under permissive conditions (33 degrees C) expressed SV40 large T antigen and had a doubling time of 36 h, whereas expression of SV40 large T antigen was negligible in astrocytes grown at 37 degrees C for 72 h, which coincided with a plateau in cell division. In a co-culture assay with human lung adenocarcinoma cells (PC14-PE6), astrocytes activated programs in the tumor cells that signal for cell division and survival. Hence, the immortalized cell line will be useful for studying the role of astrocytes in disease processes in the brain, such as metastasis.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Astrocytes/pathology , Brain Neoplasms/secondary , Cell Transformation, Viral/genetics , H-2 Antigens/genetics , Adenocarcinoma/pathology , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Astrocytes/metabolism , Brain Neoplasms/metabolism , Cell Communication , Cell Line , Cell Proliferation , Cell Survival , Coculture Techniques , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Lung Neoplasms/pathology , Melanoma/pathology , Mice , Mice, Inbred C3H , Mice, Nude , Mice, Transgenic , Receptors, N-Methyl-D-Aspartate/metabolism , Temperature , Time Factors
13.
Am J Pathol ; 174(5): 1972-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19349357

ABSTRACT

To identify the roles of various circulating cells (eg, endothelial and/or stem and progenitor cells) in angiogenesis, we parabiosed a wild-type syngeneic mouse with a transgenic syngeneic green fluorescent protein mouse. Following the establishment of a common circulation between these parabionts, we investigated acute (7 to 10 days), subacute (2 to 3 weeks), and chronic (4 to 6 weeks) phases of angiogenesis in wild-type mice using wound healing, implanted gel foam fragments, and subcutaneous tumor assays, respectively. We found that under in vitro conditions, circulating murine monocytes expressed F4/80, CD31, and vascular endothelial growth factor receptor 2, but neither CD133 nor von Willebrand factor, whereas murine endothelial cells expressed CD31, vascular endothelial growth factor receptor 2, and von Willebrand factor, but neither CD133 nor F4/80. Immunofluorescence analysis revealed that green fluorescent protein-positive cells in the walls of new vessels in wounds, gel foam blocks, and tumors expressed both F4/80 and CD31, that is, macrophages. Pericytes, cells that express both CD31 and desmin, were found both in the walls of tumor-associated vessels and within tumors. Collectively, these data demonstrate that monocytes (ie, cells that express both CD31 and F4/80) may be recruited to the site of tissue injury and directly contribute to angiogenesis, reaffirming the close relationships between various cell types within the reticuloendothelial system and suggesting possible targets for anticancer treatments.


Subject(s)
Endothelium, Vascular/physiology , Monocyte-Macrophage Precursor Cells/physiology , Monocytes/metabolism , Neoplasms/blood supply , Neoplastic Cells, Circulating/metabolism , Neovascularization, Pathologic/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , AC133 Antigen , Acute Disease , Animals , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Chronic Disease , Female , Fluorescent Antibody Technique , Glycoproteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing , von Willebrand Factor/metabolism
14.
J Exp Ther Oncol ; 8(2): 85-94, 2009.
Article in English | MEDLINE | ID: mdl-20192115

ABSTRACT

SV40-transformed 3T3 cells (SV3T3) treated with the antitubulin chemotherapeutic agent vinblastine exhibited ultrastructural alterations in their cellular microtubular scaffolding by electron microscopy. Apparent disappearance of the subcellular microtubules occurred after the cells were incubated with vinblastine at 10 microg/ml at 37 degrees C for 8 hours. Typical cytoplasmic microtubular crystals were found with additional smooth membrane-limited vesicles. These vesicles mimic the differentiation cellular organelle called annulated lamellae. Microtubules were frequently seen associated with the Golgi apparatus and rough endoplasmic reticulum in the SV3T3 cells. These microtubules may contribute to transport of products between the Golgi and the rough endoplasmic reticulum. When treated with vinblastine, microtubular crystals were also observed between the Golgi and the rough endoplasmic reticulum. The size and numbers of cytoplasmic inclusions were increased in vinblastine-treated cells. In the SV3T3 cells, microtubules are determined by image analysis to be 230 A degrees in diameter with a subunit wall of 45 A degrees thick. These microtubules have a center-to-center space of 55.6 A degrees between the protofilaments. The skewed heterodimeric microtubular subunits are composed of disk-like structures of 45 A degrees in length, 30 A degrees in width, and 20 A degrees in thickness. The subunit center-to-center skewed angle is 40 degree. After the cells were treated with vinblastine, the microtubules undergo a transitional dissociation, and reassemble into ordered crystals. These transitional microtubules have an increased diameter of 350 A degrees, and a larger protofilament center-to-center space of 85 A degrees. These morphometric measurements indicated that there is a critical microtubule dissociation distance of 30 A degrees, exceeded which the typical microtubular ultrastructure may no longer exist.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Microtubules/ultrastructure , Simian virus 40/physiology , Vinblastine/pharmacology , 3T3 Cells , Animals , Cell Transformation, Neoplastic/drug effects , Crystallization , Culture Media , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Mice , Microscopy, Electron , Microtubules/drug effects , Ribosomes/drug effects , Ribosomes/ultrastructure
15.
Neoplasia ; 10(5): 489-500, 2008 May.
Article in English | MEDLINE | ID: mdl-18472966

ABSTRACT

We determined whether phosphorylated epidermal growth factor receptor (EGFR) expressed on tumor-associated endothelial cells is a primary target for therapy with EGFR tyrosine kinase inhibitors (TKIs). Human colon cancer cells SW620CE2 (parental) that do not express EGFR or human epidermal growth factor receptor 2 (HER2) but express transforming growth factor alpha (TGF-alpha) were transduced with a lentivirus carrying nontargeting small hairpin RNA (shRNA) or TGF-alpha shRNA. The cell lines were implanted into the cecum of nude mice. Two weeks later, treatment began with saline, 4-[R]-phenethylamino-6-[hydroxyl] phenyl-7H-pyrrolo [2,3-D]-pyrimidine (PKI166), or irinotecan. Endothelial cells in parental and nontargeting shRNA tumors expressed phosphorylated EGFR. Therapy with PKI166 alone or with irinotecan produced apoptosis of these endothelial cells and necrosis of the EGFR-negative tumors. Endothelial cells in tumors that did not express TGF-alpha did not express EGFR, and these tumors were resistant to treatment with PKI166. The response of neoplasms to EGFR antagonists has been correlated with EGFR mutations, HER2 expression, Akt activation, and EGFR gene copy number. Our present data using colon cancer cells that do not express EGFR or HER2 suggest that the expression of TGF-alpha by tumor cells leading to the activation of EGFR in tumor-associated endothelial cells is a major determinant for the susceptibility of neoplasms to therapy by specific EGFR-TKI.


Subject(s)
Colonic Neoplasms/drug therapy , ErbB Receptors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cecum/metabolism , Cecum/pathology , Cell Proliferation , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Irinotecan , Lymphatic Metastasis , Male , Mice , Mice, Nude , Necrosis , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Topoisomerase I Inhibitors , Transforming Growth Factor alpha/antagonists & inhibitors , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
16.
Clin Exp Metastasis ; 25(4): 477-89, 2008.
Article in English | MEDLINE | ID: mdl-18324358

ABSTRACT

Immunohistochemical analysis of human colon cancers growing in the cecal walls of nude mice revealed that epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor 2 (VEGFR2) were expressed by different tumor cells and tumor-associated endothelial cells, whereas platelet-derived growth factor receptor (PDGFR)beta was expressed by tumor-associated endothelial cells and pericytes. We hypothesized that treatment of nude mice with AEE788 (an inhibitor of EGFR and VEGFR phosphorylation) and STI571 (an inhibitor of PDGFRbeta phosphorylation) combined with irinotecan would overcome the intratumoral heterogeneity of these growth factors and efficiently inhibit colon cancer growth and metastasis. We implanted HT29 and KM12SM cells into the cecal walls of nude mice. Two weeks later, the mice were treated with oral vehicle solution; oral AEE788, oral STI571, or intraperitoneal injection of irinotecan as single agents; or the various combinations of these agents. We then assessed the mice for tumor growth and metastasis. Immunohistochemical analyses revealed that oral AEE788 suppressed proliferation and increased apoptosis of tumor cells and tumor-associated endothelial cells. Oral STI571 increased apoptosis of tumor-associated endothelial cells and pericytes. The combination of AEE788, STI571, and irinotecan produced the greatest inhibition of primary tumor growth and metastasis. Collectively, these data demonstrate that only targeting multiple tyrosine kinase receptors on colon cancer cells and tumor-associated stromal cells can overcome the effects of biologic heterogeneity for resistance to treatment and has the potential to improve therapeutic outcome for patients with this disease.


Subject(s)
Colonic Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Apoptosis , Benzamides , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/chemistry , Colonic Neoplasms/pathology , ErbB Receptors/analysis , Humans , Imatinib Mesylate , Immunohistochemistry , Ki-67 Antigen/analysis , Lymphatic Metastasis , Male , Mice , Mice, Nude , Piperazines/therapeutic use , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Purines/therapeutic use , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/analysis , Receptors, Vascular Endothelial Growth Factor/analysis , Stromal Cells/chemistry , Stromal Cells/drug effects
17.
Am J Pathol ; 172(2): 358-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202197

ABSTRACT

The design of targeted therapy, particularly patient-specific targeted therapy, requires knowledge of the presence and intratumoral distribution of tyrosine kinase receptors. To determine whether the expression of such receptors is constant or varies between and within individual colon cancer neoplasms, we examined the pattern of expression of the ligands, epidermal growth factor, vascular endothelial growth factor, and platelet-derived growth factor-B as well as their respective receptors in human colon cancer surgical specimens and orthotopic human colon cancers growing in the cecal wall of nude mice. The expression of the epidermal growth factor receptor and the vascular endothelial growth factor receptor on tumor cells and stromal cells, including tumor-associated endothelial cells, was heterogeneous in surgical specimens and orthotopic tumors. In some tumors, the receptor was expressed on both tumor cells and stromal cells, and in other tumors the receptor was expressed only on tumor cells or only on stromal cells. In contrast, the platelet-derived growth factor receptor was expressed only on stromal cells in both surgical specimens and orthotopic tumors. Examination of receptor expression in both individual surgical specimens and orthotopic tumors revealed that the platelet-derived growth factor receptor was expressed only on stromal cells and that the patterns of epidermal growth factor receptor and vascular endothelial growth factor receptor 2 expression differed between tumor cells. This heterogeneity in receptor expression among different tumor cells suggests that targeting a single tyrosine kinase may not yield eradication of the disease.


Subject(s)
Colonic Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Animals , Cell Line, Tumor , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Microscopy, Confocal , Neoplasms, Experimental/metabolism , Stromal Cells/metabolism
18.
Neoplasia ; 9(12): 1066-77, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18084614

ABSTRACT

The purpose of our study was to determine whether the dual inhibition of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) signaling pathways in tumor-associated endothelial cells can inhibit the progressive growth of human colon carcinoma in the cecum of nude mice. SW620CE2 human colon cancer cells growing in culture and orthotopically in the cecum of nude mice expressed a high level of transforming growth factor alpha (TGF-alpha) and vascular endothelial growth factor (VEGF) but were negative for EGFR, human epidermal growth factor receptor 2 (HER2), and VEGFR. Double immunofluorescence staining revealed that tumor-associated endothelial cells expressed EGFR, VEGFR2, phosphorylated EGFR (pEGFR), and phosphorylated VEGFR (pVEGFR). Treatment of mice with either 7H-pyrrolo [2,3-d]-pyrimidine lead scaffold (AEE788; an inhibitor of EGFR and VEGFR tyrosine kinase) or CPT-11 as single agents significantly inhibited the growth of cecal tumors (P < .01); this decrease was even more pronounced with AEE788 combined with CPT-11 (P < .001). AEE788 alone or combined with CPT-11 also inhibited the expression of pEGFR and pVEGFR on tumor-associated endothelial cells, significantly decreased vascularization and tumor cell proliferation, and increased the level of apoptosis in both tumor-associated endothelial cells and tumor cells. These data demonstrate that targeting EGFR and VEGFR signaling on tumor-associated endothelial cells provides a viable approach for the treatment of colon cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Endothelial Cells/drug effects , ErbB Receptors/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Purines/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cecal Neoplasms/drug therapy , Cecal Neoplasms/enzymology , Cecal Neoplasms/pathology , Cell Line, Tumor/transplantation , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Irinotecan , Male , Mice , Mice, Nude , Neoplasm Proteins/analysis , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Purines/administration & dosage , Purines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Neoplasia ; 9(11): 979-86, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18030366

ABSTRACT

We determined whether host matrix metalloproteinase (MMP) 9 is essential to angiogenesis and to the growth of L3.6pl human pancreatic cancer cells implanted into the pancreas of wild-type (MMP-9(+/+)) and knockout (MMP-9(-/-)) nude mice. Four weeks after tumor cell injection, pancreatic tumors in MMP-9(+/+) mice were large, had many blood vessels, and contained many macrophages expressing MMP-9. In contrast, pancreatic tumors in MMP-9(-/-) mice were significantly smaller, had few blood vessels, and had few macrophages. Next, we parabiosed MMP-9(+/+) mice with MMP-9(+/+) mice, MMP-9(-/-) mice with MMP-9(-/-) mice, and MMP-9(+/+) mice with MMP-9(-/-) mice. Two weeks after parabiosis, we implanted L3.6pl cells into the pancreas of the recipient mouse in each pair. Four weeks later, the mice were necropsied. The parabiosis experiment revealed a direct correlation between intratumoral MMP-9(+/+) expressing macrophages, angiogenesis, and progressive tumor growth. Because the expression of MMP-9 by L3.6pl tumor cells was similar in all parabionts, the data clearly demonstrate a major role for host-derived MMP-9 in angiogenesis and in the growth of human pancreatic cancer in the pancreas of nude mice.


Subject(s)
Matrix Metalloproteinase 9/physiology , Neovascularization, Pathologic/etiology , Pancreatic Neoplasms/enzymology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Humans , Macrophages/enzymology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Transplantation, Heterologous
20.
Cancer Res ; 67(16): 7597-604, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699763

ABSTRACT

Using Affymetrix HG-U133 Plus 2.0 array and laser capture microdissection techniques, we determined whether different zones of the same pancreatic tumor exhibited differential expression of genes. Human L3.6pl pancreatic cancer cells were implanted into the pancreas of nude mice. Three weeks later when tumors were 7 to 9 mm in diameter, gene expression patterns in tumor cells within the central and peripheral zones were compared, and 1,222 genes showed statistically significant differences. Bioinformatic functional analysis revealed that 346 up-regulated genes in the peripheral zone were related to cytoskeleton organization and biogenesis, cell cycle, cell adhesion, cell motility, DNA replication, localization, integrin-mediated signaling pathway, development, morphogenesis, and IkappaB kinase/nuclear factor-kappaB cascade; 876 up-regulated genes in the central zone were related to regulation of cell proliferation, regulation of transcription, transmembrane receptor protein tyrosine kinase signaling pathways, response to stress, small GTPase-mediated signal transduction, hexose metabolism, cell death, response to external stimulus, carbohydrate metabolism, and response to wounding. The reliability of the microarray results were confirmed by in situ hybridization analysis of the expression of two genes. Collectively, the data showed zonal heterogeneity for gene expression profiles in tumors and suggest that characterization of zonal gene expression profiles is essential if microarray analyses of genetic profiles are to produce reproducible data, predict disease prognosis, and allow design of specific therapeutics.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...