Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncotarget ; 10(38): 3654-3666, 2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31217900

ABSTRACT

The feasibility and required sensitivity of circulating free DNA (cfDNA)-based detection methods in second-line epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) treatment are not well elucidated. We examined T790M and other activating mutations of EGFR by cfDNA to assess the clinical usability. In 45 non-small cell lung cancer (NSCLC) patients harboring activating EGFR mutations, cfDNAs were prepared from the plasma samples. EGFR mutations in cfDNA were detected using highly sensitive methods and originally developed assays and these results were compared to tissue-based definitive diagnoses. The specificity of each cfDNA-based method ranged 96-100% whereas the sensitivity ranged 56-67%, indicating its low pseudo-positive rate. In EGFR-TKI failure cohort, 41-46% samples were positive for T790M by each cfDNA-based method, which was comparable to re-biopsy tissue-based T790M positive rates in literature. The concordance of the results for each EGFR mutation ranged from 83-95%. In eight patients, the results of the cfDNA-based assays and re-biopsy-derived tissue-based test were compared. The observed overall agreement ranged in 50-63% in T790M, and in 63-100% in activating EGFR mutations. In this study, we have newly developed three types of assay which have enough sensitivity to detect cfDNA. We also detected T790M in 44% of patients who failed prior EGFR-TKI treatment, indicating that cfDNA-based assay has clinical relevance for detecting acquired mutations of EGFR.

2.
Life Sci ; 228: 251-257, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31078545

ABSTRACT

AIMS: In addition to potentially progressing to either cirrhosis or hepatocellular carcinoma, non-alcoholic steatohepatitis (NASH) is currently the leading indication for liver transplantation. Nintedanib has been clinically used to treat idiopathic pulmonary fibrosis for many years, but its effects in an animal model of NASH have not been tested. The purpose of this study was to evaluate the effects of nintendanib on NASH in choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD)-fed mice. MAIN METHODS: Male C57BL/6 mice were fed a CDAHFD for 6 weeks to induce NASH with liver fibrosis, and they were administered nintedanib (60 mg/kg/day) or distilled water orally in the last 2 weeks of the feeding period. Serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), triglyceride, and non-esterified fatty acids concentrations were measured. Serum cytokeratin 18 fragment (CK18) was detected using ELISA. Liver tissue sections from mice were stained with hematoxylin-eosin and Masson's trichrome to assess the level of steatohepatitis and fibrosis. KEY FINDINGS: CDAHFD-fed mice exhibited higher serum ALT, AST, and ALP levels compared with Control mice. A significant increase in the serum CK18 level was observed in the NASH group compared with the Control group. CDAHFD feeding also enhanced steatohepatitis and hepatic fibrosis pathological features, which were reduced after nintedanib treatment. SIGNIFICANCE: Nintedanib exerted anti-inflammatory and anti-fibrotic effects in CDAHFD-induced NASH mice.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Indoles/therapeutic use , Liver Cirrhosis/drug therapy , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Diet, High-Fat/adverse effects , Disease Models, Animal , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/blood
3.
Sci Rep ; 8(1): 6760, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29713062

ABSTRACT

Thymidine phosphorylase (TP) is a rate-limiting enzyme in the thymidine catabolic pathway. TP is identical to platelet-derived endothelial cell growth factor and contributes to tumour angiogenesis. TP induces the generation of reactive oxygen species (ROS) and enhances the expression of oxidative stress-responsive genes, such as interleukin (IL)-8. However, the mechanism underlying ROS induction by TP remains unclear. In the present study, we demonstrated that TP promotes NADPH oxidase-derived ROS signalling in cancer cells. NADPH oxidase inhibition using apocynin or small interfering RNAs (siRNAs) abrogated the induction of IL-8 and ROS in TP-expressing cancer cells. Meanwhile, thymidine catabolism induced by TP increased the levels of NADPH and intermediates of the pentose phosphate pathway (PPP). Both siRNA knockdown of glucose 6-phosphate dehydrogenase (G6PD), a rate-limiting enzyme in PPP, and a G6PD inhibitor, dihydroepiandrosterone, reduced TP-induced ROS production. siRNA downregulation of 2-deoxy-D-ribose 5-phosphate (DR5P) aldolase, which is needed for DR5P to enter glycolysis, also suppressed the induction of NADPH and IL-8 in TP-expressing cells. These results suggested that TP-mediated thymidine catabolism increases the intracellular NADPH level via the PPP, which enhances the production of ROS by NADPH oxidase and activates its downstream signalling.


Subject(s)
Glucosephosphate Dehydrogenase/genetics , NADPH Oxidases/metabolism , Neoplasms/metabolism , Thymidine Phosphorylase/genetics , Thymidine/metabolism , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Gene Knockout Techniques , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Humans , Interleukin-8/genetics , Metabolism/genetics , NADPH Oxidases/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Pentose Phosphate Pathway/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Thymidine Phosphorylase/metabolism
4.
Cell Rep ; 19(7): 1313-1321, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514652

ABSTRACT

Thymidine phosphorylase (TP), a rate-limiting enzyme in thymidine catabolism, plays a pivotal role in tumor progression; however, the mechanisms underlying this role are not fully understood. Here, we found that TP-mediated thymidine catabolism could supply the carbon source in the glycolytic pathway and thus contribute to cell survival under conditions of nutrient deprivation. In TP-expressing cells, thymidine was converted to metabolites, including glucose 6-phosphate, lactate, 5-phospho-α-D-ribose 1-diphosphate, and serine, via the glycolytic pathway both in vitro and in vivo. These thymidine-derived metabolites were required for the survival of cells under low-glucose conditions. Furthermore, activation of thymidine catabolism was observed in human gastric cancer. These findings demonstrate that thymidine can serve as a glycolytic pathway substrate in human cancer cells.


Subject(s)
Stomach Neoplasms/metabolism , Thymidine Phosphorylase/metabolism , Thymidine/metabolism , Animals , Carbon/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxyribose/pharmacology , Glycolysis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Nutritional Status/drug effects , Phosphorylation/drug effects , Stomach Neoplasms/pathology , Survival Analysis , Thymidine/chemistry
5.
Nat Commun ; 6: 8792, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26635184

ABSTRACT

Bevacizumab exerts anti-angiogenic effects in cancer patients by inhibiting vascular endothelial growth factor (VEGF). However, its use is still limited due to the development of resistance to the treatment. Such resistance can be regulated by various factors, although the underlying mechanisms remain incompletely understood. Here we show that bone marrow-derived fibrocyte-like cells, defined as alpha-1 type I collagen-positive and CXCR4-positive cells, contribute to the acquired resistance to bevacizumab. In mouse models of malignant pleural mesothelioma and lung cancer, fibrocyte-like cells mediate the resistance to bevacizumab as the main producer of fibroblast growth factor 2. In clinical specimens of lung cancer, the number of fibrocyte-like cells is significantly increased in bevacizumab-treated tumours, and correlates with the number of treatment cycles, as well as CD31-positive vessels. Our results identify fibrocyte-like cells as a promising cell biomarker and a potential therapeutic target to overcome resistance to anti-VEGF therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Bevacizumab/administration & dosage , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Lung Neoplasms/drug therapy , Animals , Cell Line, Tumor , Fibroblast Growth Factor 2/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/antagonists & inhibitors
6.
Arthritis Rheumatol ; 66(3): 560-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24574215

ABSTRACT

OBJECTIVE: Thymidine phosphorylase (TP) in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) is induced by tumor necrosis factor α (TNFα) and other cytokines that have been reported to be major inflammation mediators in RA. We previously demonstrated that TP plays an important role in angiogenesis and tumor growth, invasion, and metastasis. The aim of this study was to investigate whether the role of TP in the pathogenesis of RA is similar to its role in tumors. METHODS: In FLS obtained from 2 patients with RA, the expression of TP, interferon-γ (IFNγ)-inducible protein 10 (CXCL10), and other cytokines was measured by quantitative real-time polymerase chain reaction, immunoblotting, and enzyme-linked immunosorbent assays. Microarray analysis was performed using FLS transfected with TYMP complementary DNA and treated with a TP inhibitor. RESULTS: The expression of TP in FLS was up-regulated by TNFα, interleukin-1ß (IL-1ß), IL-17, IFNγ, and lipopolysaccharide. Microarray analysis of FLS overexpressing TP identified CXCL10 as a thymidine phosphorylase-related gene. The expression of CXCL10 was induced by TNFα, and this induction was suppressed by TYMP small interfering RNA and TP inhibitor. Furthermore, the combination of TNFα and IFNγ synergistically augmented the expression of TP and CXCL10. TP-induced CXCL10 expression was suppressed by the antioxidant EUK-8. In the synovial tissue of patients with RA, TP levels were significantly correlated with CXCL10 expression. CONCLUSION: The combination of TNFα and IFNγ strongly induced the expression of thymidine phosphorylase in RA FLS. The induction of thymidine phosphorylase enhanced the expression of CXCL10, which may contribute to the Th1 phenotype and bone destruction observed in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokine CXCL10/metabolism , Interferon-gamma/metabolism , Synovial Membrane/metabolism , Thymidine Phosphorylase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokine CXCL10/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Interferon-gamma/genetics , Synovial Membrane/pathology , Thymidine Phosphorylase/genetics , Tumor Necrosis Factor-alpha/genetics
7.
Am J Pathol ; 182(5): 1843-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23499372

ABSTRACT

Surfactant protein A (SP-A) is a large multimeric protein found in the lungs. In addition to its immunoregulatory function in infectious respiratory diseases, SP-A is also used as a marker of lung adenocarcinoma. Despite the finding that SP-A expression levels in cancer cells has a relationship with patient prognosis, the function of SP-A in lung cancer progression is unknown. We investigated the role of SP-A in lung cancer progression by introducing the SP-A gene into human lung adenocarcinoma cell lines. SP-A gene transduction suppressed the progression of tumor in subcutaneous xenograft or lung metastasis mouse models. Immunohistochemical analysis showed that the number of M1 antitumor tumor-associated macrophages (TAMs) was increased and the number of M2 tumor-promoting TAMs was not changed in the tumor tissue produced by SP-A-expressing cells. In addition, natural killer (NK) cells were also increased and activated in the SP-A-expressing tumor. Moreover, SP-A did not inhibit tumor progression in mice depleted of NK cells. Taking into account that SP-A did not directly activate NK cells, these results suggest that SP-A inhibited lung cancer progression by recruiting and activating NK cells via controlling the polarization of TAMs.


Subject(s)
Cell Polarity , Disease Progression , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophages/pathology , Pulmonary Surfactant-Associated Protein A/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Killer Cells, Natural/pathology , Macrophages/metabolism , Male , Mice , Mice, Nude , Neoplasm Metastasis , Subcutaneous Tissue/pathology , Xenograft Model Antitumor Assays
8.
Clin Exp Metastasis ; 30(3): 333-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23011677

ABSTRACT

The organ microenvironment significantly affects the processes of cancer metastasis. Elucidating the molecular mechanisms of interaction between tumor cells and the organ microenvironment is crucial for the development of effective therapeutic strategies to eradicate cancer metastases. Macrophage stimulating protein (MSP), an activator of macrophages, regulates a pleiotropic array of effects, including proliferation, cellular motility, invasiveness, angiogenesis, and resistance to anoikis. However, the role of MSP in cancer metastasis is still largely unknown. In this study, the action of MSP on the production of metastases was determined in a multiple-organ metastasis model. The murine MSP gene was transfected into two human SCLC cell lines, SBC-5 and H1048, to establish transfectants secreting biologically active MSP. MSP gene transduction did not affect cell proliferation and motility in vitro. Intravenously inoculated MSP transfectants produced significantly larger numbers of liver metastases than parental cells or vector control clones, while there were no significant differences in bone or lung metastases among them. Immunohistochemical analyses of liver metastases revealed that tumor-associated microvessel density and tumor-infiltrating macrophages were significantly increased in lesions produced by MSP transfectants. MSP could stimulate the migration of murine macrophages and endothelial cells in vitro. Consequently, MSP may be one of the major determinants that affects the properties of tumor stroma and that produces a permissive microenvironment to promote cancer metastasis.


Subject(s)
Hepatocyte Growth Factor/physiology , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Tumor Microenvironment , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Proliferation , DNA Primers , Hepatocyte Growth Factor/genetics , Humans , Liver Neoplasms/pathology , Mice , Mice, SCID , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic
9.
Mol Cancer Ther ; 11(12): 2578-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22989420

ABSTRACT

Notch signaling regulates cell-fate decisions during development and postnatal life. Little is known, however, about the role of Delta-like-4 (Dll4)-Notch signaling between cancer cells, or how this signaling affects cancer metastasis. We, therefore, assessed the role of Dll4-Notch signaling in cancer metastasis. We generated a soluble Dll4 fused to the IgG1 constant region (Dll4-Fc) that acts as a blocker of Dll4-Notch signaling and introduced it into human small cell lung cancer (SCLC) cell lines expressing either high levels (SBC-3 and H1048) or low levels (SBC-5) of Dll4. The effects of Dll4-Fc on metastasis of SCLC were evaluated using a mouse model. Although Dll4-Fc had no effect on the liver metastasis of SBC-5, the number of liver metastasis inoculated with SBC-3 and H1048 cells expressing Dll4-Fc was significantly lower than that injected with control cells. To study the molecular mechanisms of the effects of Dll4-Fc on liver metastasis, a PCR array analysis was conducted. Because the expression of NF-κB target genes was affected by Dll4-Fc, we conducted an electrophoretic mobility shift assay and observed that NF-κB activities, both with and without stimulation by TNF-α, were downregulated in Dll4-Fc-overexpressing SBC-3 and H1048 cells compared with control cells. Moreover, Dll4-Fc attenuates, at least in part, the classical and alternative NF-κB activation pathway by reducing Notch1 signaling. These results suggest that Dll4-Notch signaling in cancer cells plays a critical role in liver metastasis of SCLC by regulating NF-κB signaling.


Subject(s)
Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/secondary , Lung Neoplasms/drug therapy , NF-kappa B/metabolism , Receptors, Notch/metabolism , Recombinant Fusion Proteins/administration & dosage , Small Cell Lung Carcinoma/drug therapy , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Movement/drug effects , Down-Regulation/drug effects , Humans , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , NF-kappa B/genetics , Receptors, Notch/genetics , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transfection , Xenograft Model Antitumor Assays
10.
Oncol Rep ; 28(3): 895-902, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22751949

ABSTRACT

Thymidine phosphorylase (TP) is an angiogenic factor that plays a pivotal role in tumor angiogenesis. Various kinds of solid tumors express TP and high TP activity is correlated with microvessel density. We have previously reported that TP enhances interleukin-8 (IL-8) expression in KB human epidermoid carcinoma cells. In this study, TP was shown to be involved in enhanced expression of IL-8 in EJ human bladder cancer cells and Yumoto human cervical cancer cells as well as KB human epidermoid carcinoma cells. The enzymatic activity of TP was required for the enhanced expression of IL-8. A degradation product of thymidine was implicated in the enhanced expression of IL-8. TP augmented reactive oxygen species (ROS) generation in KB and Yumoto cells, and the enzymatic activity of TP was again required for the generation of ROS. An antioxidant, N-acetylcysteine (NAC), attenuated the generation of ROS and IL-8 mRNA expression in KB and Yumoto cells, and H2O2 increased IL-8 mRNA expression in Yumoto cells, suggesting that ROS generated by TP caused the increased expression of IL-8 mRNA. Since TP also reduced cellular glutathione levels and transcription of γ-GCS in KB cells, the TP-induced augmentation of ROS may be partially attributed to the decreased glutathione. Our findings suggest that thymidine-derived sugars enhanced ROS generation and consequently increased IL-8 expression.


Subject(s)
Gene Expression , Interleukin-8/metabolism , Reactive Oxygen Species/metabolism , Thymidine Phosphorylase/metabolism , Acetylcysteine/pharmacology , Cell Line, Tumor , Free Radical Scavengers/pharmacology , Gene Expression Regulation, Neoplastic , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-8/genetics , Real-Time Polymerase Chain Reaction
11.
Respirology ; 17(6): 984-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22574723

ABSTRACT

BACKGROUND AND OBJECTIVE: Malignant pleural mesothelioma (MPM) is an aggressive neoplasm of the mesothelium with high chemotherapeutic resistance. In this study, the preclinical therapeutic activity of the multiple tyrosine kinase inhibitor, SU6668, against MPM was examined. METHODS: Two human MPM cell lines with different pro-angiogenic cytokine expression, Y-MESO-14 cells that express high levels of vascular endothelial growth factor (VEGF) and MSTO-211H cells that express high levels of basic fibroblast growth factor (bFGF), were orthotopically inoculated into the thoracic cavities of mice with severe combined immunodeficiency. The mice with MPM were treated or not treated with SU6668 (200 mg/kg/day). RESULTS: SU6668 abrogated the proliferation of endothelial cells stimulated by VEGF or bFGF, but did not directly affect the growth of human MPM cells in vitro. In this orthotopic implantation model, treatment with SU6668 effectively reduced tumour weight and pleural effusion volumes, in association with inhibition of the growth of tumour vasculature. More importantly, treatment with SU6668 significantly prolonged survival time in mice with MPM. CONCLUSIONS: These findings suggest that SU6668 has a promising therapeutic effect on the progression of MPM in vivo through its anti-angiogenic effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Mesothelioma/drug therapy , Pleural Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Male , Mesothelioma/mortality , Mice , Mice, SCID , Neovascularization, Pathologic/drug therapy , Oxindoles , Pleural Effusion, Malignant/drug therapy , Pleural Neoplasms/mortality , Propionates , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
12.
Int J Oncol ; 40(5): 1455-69, 2012 May.
Article in English | MEDLINE | ID: mdl-22294041

ABSTRACT

Lung cancer is commonly associated with multi-organ metastasis, and the bone is a frequent metastatic site for lung cancer. However, the molecular mechanism of organ-specific metastasis remains poorly understood. To elucidate this issue, we analyzed in this study genome-wide gene expression profiles of 15 metastatic lesions from three organs (bone, lung and liver) in a mouse model with multi-organ metastasis properties of human non-small cell lung cancer cells (ACC-LC319/bone2), using a combination of laser-microbeam microdissection and DNA microarrays. We identified 299 genes that could potentially be involved in the organ-selective nature of lung cancer metastasis. Among them, 77 were bone-specifically expressed elements, including genes involved in cell adhesion, cytoskeleton/cell motility, extracellular matrix remodeling and cell-cell signaling as well as genes already known to be involved in the bone metastasis of breast cancers. Quantitative RT-PCR confirmed the specific upregulation of eight genes in bone metastasis tumors, suggesting that these genes may be involved in bone metastasis. Our findings should be helpful for a better understanding of the molecular aspects of the metastatic process in different organs, and could lead to molecular target-based anticancer drugs and prevention of metastasis, especially bone metastasis.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Animals , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Laser Capture Microdissection , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Phenotype , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
Clin Exp Metastasis ; 29(3): 207-16, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22170031

ABSTRACT

Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) show dramatic antitumor activity in a subset of patients with non-small cell lung cancer who have an active mutation in the epidermal growth factor receptor (EGFR) gene. On the other hand, some lung cancer patients with wild type EGFR also respond to EGFR-TKIs, suggesting that EGFR-TKIs have an effect on host cells as well as tumor cells. However, the effect of EGFR-TKIs on host microenvironments is largely unknown. A multiple organ metastasis model was previously established in natural killer cell-depleted severe combined immunodeficient mice using human lung cancer cells. This model was used to investigate the therapeutic efficacy of erlotinib, an EGFR-TKI, on multiple organ metastases induced by human small cell lung cancer cells (SBC-5 cells) that did not express EGFR. Although erlotinib did not have any effect on the proliferation of SBC-5 cells in vitro, it significantly suppressed bone and lung metastases in vivo, but not liver metastases. An immunohistochemical analysis revealed that, erlotinib significantly suppressed the number of osteoclasts in bone metastases, whereas no difference was seen in microvessel density. Moreover, erlotinib inhibited EGF-induced receptor activator of nuclear factor kappa-B expression in an osteoblastic cell line (MC3T3-E1 cells). These results strongly suggested that erlotinib prevented bone metastases by affecting host microenvironments irrespective of its direct effect on tumor cells.


Subject(s)
Carcinoma, Small Cell/pathology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/pathology , Neoplasm Metastasis/prevention & control , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Animals , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Carcinoma, Small Cell/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/analysis , Erlotinib Hydrochloride , Humans , Lung Neoplasms/chemistry , Male , Mice , Neovascularization, Pathologic/prevention & control , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , RANK Ligand/analysis
14.
Cancer Chemother Pharmacol ; 68(2): 497-504, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21079960

ABSTRACT

PURPOSE: Malignant pleural mesothelioma (MPM) is a highly lethal neoplasm. S-1 has been developed as a novel oral antineoplastic agent based on the modulation of 5-fluorouracil (5-FU) bioactivity. This study was conducted to investigate the preclinical therapeutic effect of S-1 on MPM. METHODS: We used three human MPM cell lines, Y-MESO-14, NCI-H290 and MSTO-211H. In vitro proliferation of human MPM cells was determined by MTT assay. Human MPM cells were orthotopically implanted into thoracic cavity of SCID mice. Tumor-bearing mice were treated with S-1 or vehicle. RESULTS: The combination of 5-FU and 5-chloro-2,4-dihydroxypyridine (CDHP) was more effective than 5-FU alone in inhibiting MPM cell proliferation in vitro. This combination was most effective in Y-MESO-14 cells, which co-expressed high protein level of dihydropyrimidine dehydrogenase (DPD) and thymidine phosphorylase (TP). In vivo data showed that treatment with S-1 significantly reduced thoracic tumors and pleural effusion produced by Y-MESO-14 cells. Moreover, treatment with S-1 prolonged the survival of Y-MESO-14 cell-bearing SCID mice. CONCLUSIONS: We demonstrated that S-1 was effective for inhibiting the proliferation of MPM cells, particularly with both DPD and TP expressions, suggesting that S-1 might be therapeutically effective for control of MPM.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mesothelioma/drug therapy , Oxonic Acid/therapeutic use , Pleural Neoplasms/drug therapy , Pyridines/therapeutic use , Tegafur/therapeutic use , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Dihydrouracil Dehydrogenase (NADP)/antagonists & inhibitors , Dihydrouracil Dehydrogenase (NADP)/metabolism , Drug Combinations , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Humans , Male , Mesothelioma/metabolism , Mesothelioma/pathology , Mice , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxonic Acid/administration & dosage , Pentosyltransferases/antagonists & inhibitors , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , Pyridines/administration & dosage , Random Allocation , Survival Analysis , Tegafur/administration & dosage , Thymidine Phosphorylase/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Nihon Rinsho ; 68(6): 997-1006, 2010 Jun.
Article in Japanese | MEDLINE | ID: mdl-20535947

ABSTRACT

Recent insights into the molecular mechanism of cancer progression have given rise to specific target-directed therapies, including monoclonal antibodies and small molecular compounds, and the advent of target-specific therapeutics has remarkably improved the outcomes of patients with various malignancies. Recent advance also lead to the identification of prognostic biomarkers as predictive factors in determining response to molecular targeted drugs. Future studies also need to develop biomarkers to further increase the power of patient selection for molecular targeted therapy. Here we review the recent progress in developing new molecular targeted drugs and the resistance to treatments as well as the importance of measuring the QOL by patient-reported outcome, for personalized therapy.


Subject(s)
Antineoplastic Agents , Drug Design , Neoplasm Metastasis , Neoplasms/drug therapy , Antibodies, Monoclonal , Biomarkers, Tumor , Humans , Neoplasm Proteins , Neoplasms/blood supply , Neoplasms/diagnosis , Neoplasms/pathology , Pharmacogenetics , Quality of Life
16.
Genes Cells ; 13(4): 295-312, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18363962

ABSTRACT

Microtubules (MTs) play crucial roles in a variety of cell functions, such as mitosis, vesicle transport and cell motility. MTs also compose specialized structures, such as centrosomes, spindles and cilia. However, molecular mechanisms of these MT-based functions and structures are not fully understood. Here, we analyzed MT co-sedimented proteins from rat brain by tandem mass spectrometry (MS) upon ion exchange column chromatography. We identified a total of 391 proteins. These proteins were grouped into 12 categories: 57 MT cytoskeletal proteins, including MT-associated proteins (MAPs) and motor proteins; 66 other cytoskeletal proteins; 4 centrosomal proteins; 10 chaperons; 5 Golgi proteins; 7 mitochondrial proteins; 62 nucleic acid-binding proteins; 14 nuclear proteins; 13 ribosomal proteins; 28 vesicle transport proteins; 83 proteins with diverse function and/or localization; and 42 uncharacterized proteins. Of these uncharacterized proteins, six proteins were expressed in cultured cells, resulting in the identification of three novel components of centrosomes and cilia. Our present method is not specific for MAPs, but is useful for identifying low abundant novel MAPs and components of MT-based structures. Our analysis provides an extensive list of potential candidates for future study of the molecular mechanisms of MT-based functions and structures.


Subject(s)
Brain Chemistry , Microtubule Proteins/analysis , Nerve Tissue Proteins/analysis , Animals , Base Sequence , Cell Line , Centrosome/chemistry , Cilia/chemistry , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/isolation & purification , DNA Primers/genetics , DNA, Complementary/genetics , Dogs , Humans , Microtubule Proteins/classification , Microtubule Proteins/genetics , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/classification , Microtubule-Associated Proteins/genetics , Mitochondrial Proteins/analysis , Mitochondrial Proteins/isolation & purification , Molecular Chaperones/analysis , Molecular Chaperones/isolation & purification , Molecular Motor Proteins/analysis , Molecular Motor Proteins/isolation & purification , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/genetics , Nuclear Proteins/analysis , Nuclear Proteins/isolation & purification , Rats , Recombinant Proteins/genetics , Ribosomal Proteins/analysis , Ribosomal Proteins/isolation & purification , Tandem Mass Spectrometry , Transfection
17.
J Biochem ; 137(1): 41-50, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15713882

ABSTRACT

We found that CEL-I was a potent cytotoxic lectin. MDCK, HeLa, and XC cells were highly sensitive to CEL-I cytotoxicity and killed in a dose-dependent manner, whereas CHO, L929, and RAW264.7 cells were relatively resistant to CEL-I, and no significant toxicity was observed up to 10 microg/ml. Among these cell lines, MDCK cells showed the highest susceptibility to CEL-I cytotoxicity. A binding study using FITC-labeled CEL-I (F-CEL-I) revealed that the amounts of bound F-CEL-I on the sensitive cell lines were evidently greater than those on the resistant cell lines, suggesting that the different susceptibility of the cell lines to CEL-I cytotoxicity is partly explained by different efficiencies of binding of CEL-I to these cell lines. Interestingly, the cytotoxicity of CEL-I toward MDCK cells was more potent than those of other lectins such as WGA, PHA-L, and Con A, even though these lectins were capable of binding to MDCK cells at comparable levels to CEL-I. Since the cytotoxicity of CEL-I was strongly inhibited by GalNAc, the binding to cell surface specific carbohydrates is essential for the CEL-I cytotoxicity. The trypan blue dye exclusion test indicated that CEL-I caused a disorder of plasma membrane integrity as a relatively early event. CEL-I failed to induce the release of carboxyfluorescein (CF) from CF-loaded MDCK cells as seen for pore-forming hemolytic isolectin CEL-III, suggesting that the primary cellular target of CEL-I may be the plasma membrane, but its action mechanism differs from that of CEL-III. Although CEL-I induced dramatic cellular morphological changes in MDCK cells, neither typical apoptotic nuclear morphological changes nor DNA fragmentation was observed in CEL-I-treated MDCK cells even after such cellular changes. Our results demonstrated that CEL-I showed a potent cytotoxic effect, especially on MDCK cells, by causing plasma membrane disorder without induction of apoptosis.


Subject(s)
Cucumaria/chemistry , Lectins, C-Type/metabolism , Animals , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Humans , Lectins, C-Type/isolation & purification , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...