Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Gan To Kagaku Ryoho ; 46(6): 974-980, 2019 Jun.
Article in Japanese | MEDLINE | ID: mdl-31273159

ABSTRACT

Lung cancer as progressive disease is often associated with a poor quality of life(QOL)which is related to a poor prognosis. We review the impact of patient-reported outcome(PRO)as an indicator of health-related QOL on the management of lung cancer patients. Cancer-specific PRO measures, which are primarily applied in scientific research for the past 30 years to compare the therapy outcomes and in drug development with adverse events. Among several PRO measures developed, the EORTC QLQ-C30 and its lung cancer-specific module QLQ-LC13 are the most frequently used instruments in clinical trials with lung cancer patients. Interestingly, cancer-specific PRO measures have been also increasingly used as daily practice, which may provide positive effects on the communication with the patient, mutual decision making and the monitoring and managing of the patient. Moreover, PRO measures have an independent prognostic value to predict survival in lung cancer patients. PRO measures have also the potential to improve the quality of care. Electronic platforms are expected to simplify the implementation of PRO measure in the daily clinic. Nevertheless, the PRO measures have not been properly implemented in Japan.


Subject(s)
Lung Neoplasms , Patient Reported Outcome Measures , Humans , Japan , Quality of Life , Surveys and Questionnaires
2.
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
3.
Respir Investig ; 56(1): 80-86, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29325686

ABSTRACT

BACKGROUND: S-1 is an oral fluoropyrimidine that is active in the treatment of non-small cell lung cancer (NSCLC); however, an optimal treatment schedule and appropriate dose adjustments of S-1 in elderly patients have not yet been established. METHODS: We conducted a phase II trial to evaluate the efficacy and safety of a 2-week S-1 monotherapy treatment followed by a 1-week interval as a first-line treatment of elderly NSCLC patients, by adjusting the dose based on the individual creatinine clearance (Ccr) and body surface area (BSA). The primary endpoint was the disease control rate. RESULTS: Forty patients were enrolled. The disease control and response rates were 89.5% (95% confidence interval [CI] = 79.8-99.2) and 7.9% (95% CI = 0.0-16.4), respectively. The median progression-free survival and overall survival times were 4.4 months (95% CI = 4.2-8.5) and 17.0 months (95% CI = 11.2-18.7), respectively. Neutropenia, anorexia, hyponatremia, hypokalemia, and pneumonia of grade ≥ 3 occurred in 5.0%, 7.5%, 5.0%, 2.5%, and 2.5% of patients, respectively. Among the patient-reported outcomes, most of the individual factors in the patients' quality of life, including upper intestine-related symptoms improved with the treatment, except for dyspnea, which slightly albeit continuously worsened throughout the study. CONCLUSIONS: In elderly patients with previously untreated advanced NSCLC, a 2-week S-1 monotherapy treatment, tailored to both the Ccr and BSA, with a 1-week interval was well tolerated and demonstrated promising efficacy. This study was registered at the University Hospital Medical Information Network (UMIN) Center (ID: UMIN000002035), Japan.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Oxonic Acid/administration & dosage , Precision Medicine , Tegafur/administration & dosage , Administration, Oral , Aged , Aged, 80 and over , Body Surface Area , Carcinoma, Non-Small-Cell Lung/mortality , Creatinine , Drug Administration Schedule , Drug Combinations , Female , Humans , Lung Neoplasms/mortality , Male , Metabolic Clearance Rate , Survival Rate , Treatment Outcome
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.
Gan To Kagaku Ryoho ; 42(8): 893-902, 2015 Aug.
Article in Japanese | MEDLINE | ID: mdl-26321701

ABSTRACT

In Japan, investigator-initiated clinical phase IV trials must follow the Ethical Guidelines for Medical and Health Research Involving Human Subjects, issued in December 2014. In addition, researchers must follow the Helsinki Declaration. In these clinical trials, academia-industry collaborations involving funding and technical support are required to develop better evidence- based medicine. Nevertheless, instances of publications reporting biases or misconduct in the research process occur frequently, which may lead to other problems due to lack of transparency. To address this issue, an institutional framework must be developed and maintained in which investigators maintain a high level of ethical adherence to protect the welfare of research subjects while carrying out research scientifically and appropriately under a conflict of interest (COI) disclosure. All authors must be seriously committed to greater responsibility, accountability, and transparency in collaborating with the industry.


Subject(s)
Biomedical Research/ethics , Guidelines as Topic , Drug Discovery , Drug Industry/ethics , Humans , Peer Review, Research/ethics
6.
Nat Commun ; 6: 7035, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25923845

ABSTRACT

The p16(INK4a) tumour suppressor has an established role in the implementation of cellular senescence in stem/progenitor cells, which is thought to contribute to organismal ageing. However, since p16(INK4a) knockout mice die prematurely from cancer, whether p16(INK4a) reduces longevity remains unclear. Here we show that, in mutant mice homozygous for a hypomorphic allele of the α-klotho ageing-suppressor gene (kl(kl/kl)), accelerated ageing phenotypes are rescued by p16(INK4a) ablation. Surprisingly, this is due to the restoration of α-klotho expression in kl(kl/kl) mice and does not occur when p16(INK4a) is ablated in α-klotho knockout mice (kl(-/-)), suggesting that p16(INK4a) is an upstream regulator of α-klotho expression. Indeed, p16(INK4a) represses α-klotho promoter activity by blocking the functions of E2Fs. These results, together with the observation that the expression levels of p16(INK4a) are inversely correlated with those of α-klotho throughout ageing, indicate that p16(INK4a) plays a previously unrecognized role in downregulating α-klotho expression during ageing.


Subject(s)
Aging/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Genes, p16 , Glucuronidase/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Glucuronidase/metabolism , Humans , Klotho Proteins , Male , Mice , Mice, Inbred C57BL , Phenotype , Promoter Regions, Genetic
7.
PLoS One ; 9(11): e113606, 2014.
Article in English | MEDLINE | ID: mdl-25411851

ABSTRACT

Small cell lung cancer (SCLC) is aggressive, with rapid growth and frequent bone metastasis; however, its detailed molecular mechanism remains poorly understood. Here, we report the critical role of early growth factor 4 (EGR4), a DNA-binding, zinc-finger transcription factor, in cell proliferation of SCLC. EGR4 overexpression in HEK293T cells conferred significant upregulation of specific splice variants of the parathyroid hormone-related protein (PTHrP) gene, resulting in enhancement of the secretion of PTHrP protein, a known mediator of osteolytic bone metastasis. More importantly, depletion of EGR4 expression by siRNA significantly suppressed growth of the SCLC cell lines, SBC-5, SBC-3 and NCI-H1048. On the other hand, introduction of EGR4 into NIH3T3 cells significantly enhanced cell growth. We identified four EGR4 target genes, SAMD5, RAB15, SYNPO and DLX5, which were the most significantly downregulated genes upon depletion of EGR4 expression in all of the SCLC cells examined, and demonstrated the direct recruitment of EGR4 to their promoters by ChIP and luciferase reporter analysis. Notably, knockdown of the expression of these genes by siRNA remarkably suppressed the growth of all the SCLC cells. Taken together, our findings suggest that EGR4 likely regulates the bone metastasis and proliferation of SCLC cells via transcriptional regulation of several target genes, and may therefore be a promising target for the development of anticancer drugs for SCLC patients.


Subject(s)
Early Growth Response Transcription Factors/metabolism , Parathyroid Hormone-Related Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Early Growth Response Transcription Factors/antagonists & inhibitors , Early Growth Response Transcription Factors/genetics , HEK293 Cells , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NIH 3T3 Cells , Paracrine Communication , Parathyroid Hormone-Related Protein/genetics , RANK Ligand/genetics , RANK Ligand/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
8.
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
9.
Respir Investig ; 51(4): 241-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24238232

ABSTRACT

BACKGROUND: Bronchial asthma is characterized by allergic airway inflammation involving C-C chemokine receptor type 4 (CCR4)-positive Th2 cells. As such, we hypothesize that the disease can be alleviated by targeted-elimination of CCR4⁺ cells. Thymus and activation-regulated chemokine (TARC)-PE38, a TARC fused the exotoxin fragment PE38 from Pseudomonas aeruginosa, has been shown to efficiently kill CCR4⁺ cells by delivering the exotoxin fragment PE38 into CCR4⁺ cells. To test our hypothesis, we examined whether TARC-PE38 could suppress allergic airway inflammation in a mouse model of house dust mite (HDM)-induced allergic airway inflammation. METHODS: We evaluated the effect of TARC-PE38 on the major characteristics of HDM-induced allergic airway inflammation. Airway hyperresponsiveness, lung histopathology, lung Th1/Th2 cell populations, and concentrations of Th1/Th2 cytokines in the lungs were assessed in HDM-sensitized and challenged mice in the presence and absence of TARC-PE38. RESULTS: TARC-PE38 efficiently suppressed allergic airway inflammation by significantly reducing airway hyperresponsiveness, the overall area of inflammation, and goblet cell hyperplasia. In HDM-sensitized and challenged mice, TARC-PE38 specifically reduced the numbers of CCR4⁺ cells. This reduction was associated with a significant decrease in the production of Th2 cytokines in the airway,and a decrease in the number of leukocytes, including macrophages, eosinophils and lymphocytes, within the subepithelial area of the lungs and airway lumen. TARC-PE38 had noeffect on Th1 cells. CONCLUSION: Our data suggest that the elimination of CCR4⁺ cells via TARC-PE38 treatment is sufficient to control allergic airway inflammation and airway hyperresponsiveness.


Subject(s)
Asthma/drug therapy , Asthma/immunology , Chemokine CCL17/therapeutic use , Exotoxins/therapeutic use , Molecular Targeted Therapy , Receptors, CCR4 , Recombinant Fusion Proteins/therapeutic use , Th2 Cells/immunology , Airway Resistance/drug effects , Animals , Asthma/etiology , Asthma/genetics , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/etiology , Chemokine CCL17/pharmacology , Disease Models, Animal , Exotoxins/pharmacology , Female , Immunoglobulin E/blood , Lung/cytology , Lung/immunology , Mice , Mice, Inbred BALB C , Pyroglyphidae/immunology , Recombinant Fusion Proteins/pharmacology , Th1 Cells/immunology
10.
Am J Respir Cell Mol Biol ; 49(4): 536-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23642017

ABSTRACT

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in various biological functions, including cell survival, proliferation, migration, and adhesion. FAK is an essential factor for transforming growth factor ß to induce myofibroblast differentiation. In the present study, we investigated whether the targeted inhibition of FAK by using a specific inhibitor, TAE226, has the potential to regulate pulmonary fibrosis. TAE226 showed inhibitory activity of autophosphorylation of FAK at tyrosine 397 in lung fibroblasts. The addition of TAE226 inhibited the proliferation of lung fibroblasts in response to various growth factors, including platelet-derived growth factor and insulin-like growth factor I, in vitro. TAE226 strongly suppressed the production of type I collagen by lung fibroblasts. Furthermore, treatment of fibroblasts with TAE226 reduced the expression of α-smooth muscle actin induced by transforming growth factor ß, indicating the inhibition of differentiation of fibroblasts to myofibroblasts. Administration of TAE226 ameliorated the pulmonary fibrosis induced by bleomycin in mice even when used late in the treatment. The number of proliferating mesenchymal cells was reduced in the lungs of TAE226-treated mice. These data suggest that FAK signal plays a significant role in the progression of pulmonary fibrosis and that it can become a promising target for therapeutic approaches to pulmonary fibrosis.


Subject(s)
Focal Adhesion Kinase 1/antagonists & inhibitors , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Actins/genetics , Actins/metabolism , Animals , Bleomycin , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , DNA/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tyrosine/genetics , Tyrosine/metabolism
11.
J Immunol ; 190(12): 6239-49, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23690472

ABSTRACT

Podoplanin (Aggrus), which is a type I transmembrane sialomucin-like glycoprotein, is highly expressed in malignant pleural mesothelioma (MPM). We previously reported the generation of a rat anti-human podoplanin Ab, NZ-1, which inhibited podoplanin-induced platelet aggregation and hematogenous metastasis. In this study, we examined the antitumor effector functions of NZ-1 and NZ-8, a novel rat-human chimeric Ab generated from NZ-1 including Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity against MPM in vitro and in vivo. Immunostaining with NZ-1 showed the expression of podoplanin in 73% (11 out of 15) of MPM cell lines and 92% (33 out of 36) of malignant mesothelioma tissues. NZ-1 could induce potent ADCC against podoplanin-positive MPM cells mediated by rat NK (CD161a(+)) cells, but not murine splenocytes or human mononuclear cells. Treatment with NZ-1 significantly reduced the growth of s.c. established tumors of MPM cells (ACC-MESO-4 or podoplanin-transfected MSTO-211H) in SCID mice, only when NZ-1 was administered with rat NK cells. In in vivo imaging, NZ-1 efficiently accumulated to xenograft of MPM, and its accumulation continued for 3 wk after systemic administration. Furthermore, NZ-8 preferentially recognized podoplanin expressing in MPM, but not in normal tissues. NZ-8 could induce higher ADCC mediated by human NK cells and complement-dependent cytotoxicity as compared with NZ-1. Treatment with NZ-8 and human NK cells significantly inhibited the growth of MPM cells in vivo. These results strongly suggest that targeting therapy to podoplanin with therapeutic Abs (i.e., NZ-8) derived from NZ-1 might be useful as a novel immunotherapy against MPM.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotherapy/methods , Membrane Glycoproteins/immunology , Mesothelioma/immunology , Pleural Neoplasms/immunology , Animals , Antibodies, Monoclonal/pharmacology , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Rats , Rats, Wistar , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Xenograft Model Antitumor Assays
12.
J Med Invest ; 60(1-2): 127-37, 2013.
Article in English | MEDLINE | ID: mdl-23614921

ABSTRACT

Circulating fibrocytes had been reported to migrate into the injured lungs, and contribute to fibrogenesis via chemokine-chemokine receptor systems including CXCL12-CXCR4 axis. Here we hypothesized that blockade of CXCR4 might inhibit the migration of fibrocytes to the injured lungs and the subsequent pulmonary fibrosis. To explore the antifibrotic effects of blockade of CXCR4, we used a specific antagonist for CXCR4, AMD3100, in bleomycin-induced pulmonary fibrosis model in mice. Administration of AMD3100 significantly improved the loss of body weight of mice treated with bleomycin, and inhibited the fibrotic lesion in subpleural areas of the lungs. The quantitative analysis demonstrated that treatment with AMD3100 reduced the collagen content and fibrotic score (Aschcroft score) in the lungs. Although AMD3100 did not affect cell classification in bronchoalveolar lavage fluid on day 7, the percentage of lymphocytes was reduced by AMD3100 on day 14. AMD3100 directly inhibited the migration of human fibrocytes in response to CXCL12 in vitro, and reduced the trafficking of fibrocytes into the lungs treated with bleocmycin in vivo. These results suggest that the blockade of CXCR4 might be useful strategy for therapy of patients with pulmonary fibrosis via inhibiting the migration of circulating fibrocytes.


Subject(s)
Bleomycin/toxicity , Heterocyclic Compounds/therapeutic use , Pulmonary Fibrosis/prevention & control , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Cell Movement/drug effects , Chemokine CXCL12/analysis , Chemotaxis/drug effects , Cyclams , Female , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced
13.
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
14.
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
15.
Int J Rheum Dis ; 15(5): 486-95, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23083039

ABSTRACT

AIM: Rheumatoid arthritis (RA) is a chronic inflammatory disorder of the synovium resulting in the destruction of affected joint cartilage and bone structures. Etanercept is a biological agent that blocks the tumor necrosis factor-α (TNF-α)-mediated inflammatory processes in RA patients, and has a regenerative effect on cartilage. In order to identify novel disease-related proteins and candidate biomarkers, we performed proteomic profiling of the serum in patients with RA who were treated with etanercept. METHOD: Serum samples were obtained from eight RA patients before and after etanercept treatment. The low molecular weight proteins in the serum were concentrated and analyzed by liquid chromatography-tandem mass spectrometry. The results before and after etanercept treatment were compared by the spectrum count method. RESULTS: Among a total of 477 proteins identified, 12 were found to be decreased and five were increased by etanercept treatment. Some of the changed proteins were known to be related to RA, and most of the other changed proteins may play possible roles in the TNF-α signaling pathway or the state of cartilage and extracellular matrix. CONCLUSION: The present proteomic study identified several proteins that could be involved in the pathogenesis of RA. These findings could thus lead to the identification of novel candidate disease-related protein biomarkers for RA, or indicate new targets for therapy.


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Immunoglobulin G/therapeutic use , Proteome/metabolism , Receptors, Tumor Necrosis Factor/therapeutic use , Adult , Complement Factor D/metabolism , Enkephalins/blood , Etanercept , Extracellular Matrix Proteins/blood , Female , Humans , Intramolecular Oxidoreductases/blood , Kininogens/blood , Lipocalins/blood , Middle Aged , Protein Precursors/blood , Proteoglycans/blood
16.
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
17.
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
18.
Am J Pathol ; 181(3): 1034-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22789825

ABSTRACT

Acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) is a serious problem in the management of EGFR mutant lung cancer. We recently reported that hepatocyte growth factor (HGF) induces resistance to EGFR-TKIs by activating the Met/PI3K pathway. HGF is also known to induce angiogenesis in cooperation with vascular endothelial growth factor (VEGF), which is an important therapeutic target in lung cancer. Therefore, we hypothesized that dual inhibition of HGF and VEGF may be therapeutically useful for controlling HGF-induced EGFR-TKI-resistant lung cancer. We found that a dual Met/VEGF receptor 2 kinase inhibitor, E7050, circumvented HGF-induced EGFR-TKI resistance in EGFR mutant lung cancer cell lines by inhibiting the Met/Gab1/PI3K/Akt pathway in vitro. HGF stimulated VEGF production by activation of the Met/Gab1 signaling pathway in EGFR mutant lung cancer cell lines, and E7050 showed an inhibitory effect. In a xenograft model, tumors produced by HGF-transfected Ma-1 (Ma-1/HGF) cells were more angiogenic than vector control tumors and showed resistance to gefitinib. E7050 alone inhibited angiogenesis and retarded growth of Ma-1/HGF tumors. E7050 combined with gefitinib induced marked regression of tumor growth. Moreover, dual inhibition of HGF and VEGF by neutralizing antibodies combined with gefitinib also markedly regressed tumor growth. These results indicate the therapeutic rationale of dual targeting of HGF-Met and VEGF-VEGF receptor 2 for overcoming HGF-induced EGFR-TKI resistance in EGFR mutant lung cancer.


Subject(s)
Aminopyridines/pharmacology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Hepatocyte Growth Factor/pharmacology , Lung Neoplasms/blood supply , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Aminopyridines/therapeutic use , Animals , Antibodies/pharmacology , Antibody Specificity/drug effects , Cell Line, Tumor , ErbB Receptors/metabolism , Gefitinib , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Models, Biological , Mutation/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
19.
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
20.
J Med Invest ; 59(1-2): 174-85, 2012.
Article in English | MEDLINE | ID: mdl-22450006

ABSTRACT

BACKGROUND: Gefitinib, an inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase, has been reported to be associated with interstitial lung disorders, and their high incidence and mortality have become a matter of great concern, especially in Japan. In this study, we investigated the effect of gefitinib on different phases of radiation-induced lung disorders in an experimental model. METHODS: The thoraxes of Wistar rats were irradiated on day 1 with a single X-ray dose of 20 Gy, and gefitinib (50 mg/kg/day) was orally administered from day 1 to 14. The rat lungs were harvested on days 15 and 57 and the bronchoalveolar lavage (BAL) was performed. RESULTS: Gefitinib treatment increased the infiltration of inflammatory cells, which produced more pro-inflammatory cytokines (IL-6, IL-1ß), in the lungs of the irradiated rats on days 15 and 57, while gefitinib treatment reduced collagen content of the lungs in irradiated rats and decreased proliferation and EGFR expression in the lung fibroblasts from irradiated rats on day 57. CONCLUSIONS: In irradiated rats, gefitinib treatment augmented lung inflammation, including inflammatory cell infiltration and pro-inflammatory cytokine expression, while gefitinib treatment attenuated fibrotic lung remodeling due to the inhibition of lung fibroblast proliferation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Quinazolines/pharmacology , Radiation Pneumonitis/drug therapy , Animals , Disease Models, Animal , Gefitinib , Male , Protein Kinase Inhibitors/pharmacology , Radiation Pneumonitis/pathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...