Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Oncology ; 100(5): 267-277, 2022.
Article in English | MEDLINE | ID: mdl-35134807

ABSTRACT

INTRODUCTION: Antidiabetic drug metformin exerts various antitumor effects on different cancers. Esophageal squamous cell carcinoma (ESCC) is an intractable digestive organ cancer and new treatment strategy is required. In this study, we performed a comprehensive gene expression analysis of ESCC cell lines treated with metformin, which provided helpful information on the antitumor effects of metformin in ESCC. Next, we selected a promising gene among them and examined its effects on ESCC properties. METHODS: We examined metformin-induced mRNA expression changes in two human ESCC cell lines by performing next-generation sequencing (NGS) and pathway analysis. Heat shock protein family A (Hsp70) member 6 (HSPA6) expression in surgical specimens obtained from 83 ESCC patients who underwent curative operations was evaluated immunohistochemically and analyzed. RESULTS: Metformin upregulated mRNA expression of the many genes, including HSPA6, a cancer immune-related gene, and inhibited mRNA expression of the other many genes. Pathway analysis indicated major canonical pathways and upstream regulators related to metformin. The result indicated HSPA6 as a promising biomarker. HSPA6 expression correlated with disease-free survival (DFS) of the patients with all stage ESCC (p = 0.021), especially with stage I/II ESCC (p < 0.001). With stage III, low HSPA6 expression was not associated with poor DFS (p = 0.918). Multivariate analysis indicated that independent low HSPA6 expression was an independent poor prognostic factor of stage I/II ESCC (p < 0.001). However, HSPA6 expression did not correlate with the clinicopathological characteristics, including age, sex, tumor depth, lymph node metastasis, tumor stage, and tumor markers of the patients with stage I/II ESCC. CONCLUSIONS: This NGS analysis detected prospective candidate genes, including HSPA6. Our results indicate that HSPA6 is a promising biomarker of the recurrence risk of stage I/II ESCC. Further studies on HSPA6 would lead to better treatment.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , HSP70 Heat-Shock Proteins/metabolism , Metformin , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Heat-Shock Proteins/genetics , Humans , Metformin/pharmacology , Metformin/therapeutic use , Prognosis , Prospective Studies , RNA, Messenger
2.
Oncol Lett ; 18(1): 872-881, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31289565

ABSTRACT

Expression of genes is controlled by histone modification, histone acetylation and methylation, but abnormalities of these modifications have been observed in carcinogenesis and cancer development. The effect of the lysine-specific histone demethylase 1 (LSD1) inhibitor, a demethylating enzyme of histones, is thought to be caused by controlling the expression of genes. The aim of the present study is to elucidate the efficacies of the LSD1 inhibitor on the gene expression of esophageal cancer cell lines using chromatin immunoprecipitation (ChIP)-Seq. A comprehensive analysis of gene expression changes in esophageal squamous cell carcinoma (ESCC) cell lines induced by the LSD1 inhibitor NCL1 was clarified via analysis using microarray. In addition, ChIP-seq analysis was conducted using a SimpleChIP plus Enzymatic Chromatin IP kit. NCL1 strongly suppressed the proliferation of T.Tn and TE2 cells, which are ESCC cell lines, and further induced apoptosis. According to the combinatory analysis of ChIP-seq and microarray, 17 genes were upregulated, and 16 genes were downregulated in both cell lines. The comprehensive gene expression study performed in the present study is considered to be useful for analyzing the mechanism of the antitumor effect of the LSD1 inhibitor in patients with ESCC.

3.
Oncology ; 94(3): 142-148, 2018.
Article in English | MEDLINE | ID: mdl-29216641

ABSTRACT

OBJECTIVE: ZNF750, a transcriptional regulator of epidermal differentiation, has been identified as a tumor suppressor in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to investigate the clinical and prognostic significance of ZNF750 expression and to evaluate the effect of ZNF750 knockdown on cell proliferation, migration, and invasion in ESCC. METHODS: A total of 124 patients with ESCC who underwent curative esophagectomy were evaluated in this study. The expression of ZNF750 in surgical specimens was immunohistochemically assessed and used in the analysis of clinicopathological features and overall survival (OS). The molecular role of ZNF750 was investigated by ZNF750 knockdown using small interfering RNA (siRNA) in ESCC cell lines. RESULTS: Low ZNF750 expression had a significant correlation with positive lymph node metastasis (p = 0.028). Furthermore, there was a significant relationship between low expression of ZNF750 in ESCC and a poor OS, and a multivariate analysis showed that low ZNF750 expression was an independent prognostic factor (p = 0.020). The cell growth, migration, and invasion were significantly increased by downregulation of ZNF750. CONCLUSIONS: The low expression of ZNF750 was significantly associated with a poor prognosis, and ZNF750 expression may, therefore, be a reliable prognostic biomarker in ESCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation/physiology , Esophageal Squamous Cell Carcinoma , Female , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Prognosis , Tumor Suppressor Proteins
4.
Ann Surg Oncol ; 24(11): 3446-3455, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27882471

ABSTRACT

BACKGROUND: The expression of Fos-related antigen 1 (Fra-1) affects tumor progression, migration, and invasion. In this study, we identified the genes regulated by Fra-1 in esophageal squamous cell carcinoma (ESCC). METHODS: We constructed Fra-1 knockdown models via the transfection of small interfering RNA (siRNA) into ESCC cell lines (TE10, TE11). The expression levels of the genes in the knockdown models were analyzed using a microarray and a Biobase Upstream Analysis, while the expression levels of the candidate genes in the primary tumors of surgical specimens obtained from ESCC patients were determined using real-time polymerase chain reaction (PCR) and immunohistochemical staining. The clinicopathological features were then analyzed. RESULTS: The Biobase Upstream Analysis showed the high-mobility-group protein-1 (HMGA1) to be a significant gene regulated by Fra-1. Actual binding of Fra-1 to the promotor region of HMGA1 was revealed in subsequent chromatin immunoprecipitation PCR experiments. Patients with a positive HMGA1 expression had a poor prognosis, and a multivariate analysis demonstrated a positive HMGA1 expression to be a significant independent prognostic factor. CONCLUSION: HMGA1 is regulated by Fra-1 in ESCC, and the HMGA1 expression is significantly associated with a poor prognosis in ESCC patients. Downregulation of the HMGA1 expression may become a practical treatment strategy against ESCC in the future.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/secondary , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HMGA1a Protein/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/surgery , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/surgery , HMGA1a Protein/genetics , Humans , Lymphatic Metastasis , Neoplasm Invasiveness , Prognosis , Proto-Oncogene Proteins c-fos/genetics , Tumor Cells, Cultured
5.
Oncol Rep ; 36(5): 2535-2543, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27599779

ABSTRACT

Exosomes play important roles in cancer progression. Although its contents (e.g., proteins and microRNAs) have been focused on in cancer research, particularly as potential diagnostic markers, the exosome behavior and methods for exosome quantification remain unclear. In the present study, we analyzed the tumor-derived exosome behavior and assessed the quantification of exosomes in patient plasma as a biomarker for esophageal squamous cell carcinoma (ESCC). A CD63-GFP expressing human ESCC cell line (TE2-CD63-GFP) was made by transfection, and mouse subcutaneous tumor models were established. Fluorescence imaging was performed on tumors and plasma exosomes harvested from mice. GFP-positive small vesicles were confirmed in the plasma obtained from TE2-CD63-GFP tumor-bearing mice. Patient plasma was collected in Chiba University Hospital (n=86). Exosomes were extracted from 100 µl of the plasma and quantified by acetylcholinesterase (AChE) activity. The relationship between exosome quantification and the patient clinical characteristics was assessed. The quantification of exosomes isolated from the patient plasma revealed that esophageal cancer patients (n=66) expressed higher exosome levels than non-malignant patients (n=20) (P=0.0002). Although there was no correlation between the tumor progression and the exosome levels, exosome number was the independent prognostic marker and low levels of exosome predicted a poor prognosis (P=0.03). In conclusion, exosome levels may be useful as an independent prognostic factor for ESCC patients.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , Esophageal Neoplasms/blood , Exosomes/metabolism , Prognosis , Aged , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Exosomes/genetics , Exosomes/pathology , Female , Green Fluorescent Proteins/blood , Humans , Male , Mice , MicroRNAs/blood , Middle Aged , Neoplasm Proteins/blood , Tetraspanin 30/blood
6.
Ann Surg Oncol ; 23(1): 312-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25791791

ABSTRACT

BACKGROUND: The expression of genes can be influenced by the balance of histone acetylation and/or histone demethylation, with an imbalance of these processes possibly observed in many cancers. The histone demethylase LSD1 inhibitor activity is associated with selective transcriptional regulation and alterations in the gene expression. However, the exact mechanisms underlying the antitumor effects of LSD1 inhibitors are not fully understood. METHODS: The antitumor effects of NCL1, an LSD1 inhibitor, in esophageal squamous cell cancer (ESCC) cell lines were evaluated. A comprehensive analysis of the changes in the gene expression in ESCC cell lines induced by NCL1 was carried out using a microarray analysis. A loss-of-function assay using a siRNA analysis was performed to examine the oncogenic function of the gene. RESULTS: NCL1 strongly inhibited the cell growth of T.Tn and TE2 ESCC cells and induced apoptosis. According to the microarray analysis, 81 genes in the T.Tn cells and 149 genes in the TE2 cells were up- or down-regulated 2-fold or more by NCL1 exposure. Among these genes, 27 were contained in both cell lines and exhibited similar expression patterns. PHLDB2, one of the genes down-regulated by NCL1, was overexpressed in the ESCC tumor tissues. Moreover, a high expression level of PHLDB2 was found to be significantly correlated with poor prognosis. CONCLUSIONS: The present observations of the comprehensive analysis of the gene expression levels provide insight into the mechanisms underlying the antitumor effects of LSD1 inhibitors in ESCC patients.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/prevention & control , Cell Proliferation/drug effects , Esophageal Neoplasms/prevention & control , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Movement/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Gene Expression Profiling , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Immunoenzyme Techniques , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Anticancer Res ; 35(3): 1353-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25750285

ABSTRACT

BACKGROUND/AIM: Although conventional staging laparoscopy (SL) has improved the diagnostic accuracy of peritoneal dissemination, novel technology is needed to increase the sensitivity of SL. We herein describe a new imaging method employing near-infrared (NIR) fluorescence imaging using a liposomal synthesized indocyanine green (ICG) liposomal derivative, LP-ICG-C18. METHODS AND RESULTS: LP-ICG-C18 is a NIR-photoactivating probe in which an ICG fluorophore is covalently conjugated with a phospholipid moiety. Nude mice were intraperitoneally injected with gastric cancer cells. Twelve days later, the mice were given intravenous injections of LP-ICG-C18 at a dose of 0.15 mg/kg. A NIR imaging system was used to identify the disseminated tumors. The disseminated nodules in mice were detected without any difficulties. Disseminated tumor nodules were collected from mice with or without injections of liposomal formulation and were transferred into the swine peritoneal cavity. The nodules in the swine peritoneal cavity were clearly and promptly defined by the NIR imaging system. CONCLUSION: NIR-fluorescing liposomal probes can effectively target peritoneal disseminated tumors and can be easily detected by a NIR imaging system. These results warrant future clinical trials of our imaging system and may contribute to a more precise diagnosis and therapeutic approach for gastric cancer patients.


Subject(s)
Indocyanine Green/administration & dosage , Laparoscopy/methods , Peritoneal Neoplasms/diagnosis , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Humans , Liposomes , Mice , Mice, Inbred BALB C , Optical Imaging , Swine
8.
Ann Surg Oncol ; 18(3): 832-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20839070

ABSTRACT

BACKGROUND: Heat-shock protein gp96 plays an important role in antitumor immunoreactions. Gp96 has a close relationship with antitumor immunity. This study evaluated the correlation between gp96 expression and the prognosis in esophageal squamous cell carcinoma. METHODS: Seventy-eight patients with primarily resected esophageal squamous cell carcinoma were enrolled onto this study, and gp96 expression was evaluated by immunohistochemical staining. The association of clinicopathological factors and patients' survival was calculated by univariate (log rank test) and multivariate (Cox proportional hazard regression method) analyses. RESULTS: Fifty-seven (73%) of 78 cases were gp96 positive, and 21 were negative (27%). The survival of patients with gp96-negative disease was significantly shorter (5-year survival, 22.9 months) than with gp96-positive disease (45.8 months; P = 0.049), and the multivariate analysis showed that gp96 negativity is an independent risk factor for poor survival (hazard ratio, 2.577; P = 0.040). Gp96-negative cases had more metastatic lymph nodes than did negative cases, especially in T1 cases (4.8 in gp96-negative cases vs. 0.84 in gp96-positive cases; P = 0.064) CONCLUSIONS: The downregulation of gp96 expression is closely correlated with poor survival in esophageal squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophagectomy , Membrane Glycoproteins/metabolism , Adult , Aged , Carcinoma, Squamous Cell/surgery , Esophageal Neoplasms/surgery , Female , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Retrospective Studies , Survival Rate , Treatment Outcome
9.
Oncology ; 75(1-2): 113-9, 2008.
Article in English | MEDLINE | ID: mdl-18784437

ABSTRACT

BACKGROUND: p53 gene therapy has been examined in several clinical trials, however, the results of those trials have mostly been unsatisfactory due to the low efficacy of this therapy. Leptomycin B (LMB) is an antibiotic originally isolated from Streptomyces that has the ability to inhibit the export of proteins containing a nuclear export signal from the nucleus to the cytoplasm. Currently, it has been shown that p53 protein has a nuclear export signal. In this study, we assessed whether LMB augments the transduced p53 gene effect. METHODS: Antiproliferative effect of LMB was assessed in human esophageal squamous cancer cell lines. Accumulation of p53 protein into the nucleus by LMB was observed by fluorescence microscopy. The combined effect of p53 and LMB was evaluated in in vitro experiments. RESULTS: LMB induced cell death in a dose-dependent manner and p53 drastically accumulated in the nucleus after LMB treatment. The combinatory treatment of p53 gene and LMB significantly increases the efficiency compared to either agent alone. CONCLUSIONS: Our findings suggest that LMB has a potent ability to augment the effect of the tumor suppressor p53 in esophageal squamous cancer cell lines and that it is a promising component in p53 gene therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Genes, p53 , Genetic Therapy , Cell Line, Tumor , Combined Modality Therapy , Fatty Acids, Unsaturated/therapeutic use , Humans
10.
Anticancer Res ; 28(2A): 665-71, 2008.
Article in English | MEDLINE | ID: mdl-18507005

ABSTRACT

BACKGROUND: Recently, the use of histone deacetylase inhibitors (HDACI) with gene therapy has been shown to improve the effect of this therapy. The effectiveness of one of the novel HDACIs, FK228, was examined in adenovirus-mediated p53 gene therapy of esophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS: The expression levels of coxsackie and adenovirus receptor (CAR) in ESCC patients were examined immunohistochemically. CAR induction by FK228 in ESCC cells was analyzed by real-time PCR and Western blotting. The efficiencies of adenoviral transduction treated with FK228 were determined using AV1.0CMV-betagal. The acetylation of p53 protein was detected by Western blotting. RESULTS: CAR expression was reduced in some tumor specimens compared to that in normal specimens. CAR expression was increased by FK228 in both in vitro and in vivo experiments. FK228 improved the efficiency of adenovirus infection. Acetylated p53 protein was increased in a dose- and a time-dependent manner. CONCLUSION: Our findings suggest that FK228 has a potent ability to augment the effect of adenovirus-mediated p53 gene therapy in ESCC cells.


Subject(s)
Carcinoma, Squamous Cell/therapy , Depsipeptides/pharmacology , Esophageal Neoplasms/therapy , Genes, p53 , Genetic Therapy , Histone Deacetylase Inhibitors , Acetylation , Adenoviridae/genetics , Animals , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enzyme Inhibitors/pharmacology , Female , Gene Transfer Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Virus/metabolism , Xenograft Model Antitumor Assays
11.
Int J Oncol ; 31(3): 509-15, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671676

ABSTRACT

We tested a new therapeutic modality for head and neck and esophageal cancers, a combination of direct intratumoral (i.t.) administration of dendritic cells (DCs) and radiation therapy (RT) in mouse squamous cell carcinoma (SCC). We also evaluated the functions of gp96, which can enhance systemic antitumor activity, and the mechanism of the abscopal effect. Mouse SCC cells (1 x 10(5)), SCCVII, were inoculated into the left femur of C3H/He mice subcutaneously, and also similarly inoculated into chest subcutaneous tissue. Only the left femur tumor was exposed to 4 or 10 Gy of ionizing radiation, and then 1 x 10(6) DCs i.t. was injected only into the femur tumor. Following this procedure, tumor volumes of the femur and chest were measured. We evaluated whether gp96 could enhance the antitumor effect. With DCs i.t. and RT, tumor growth was markedly suppressed. Tumor growth of non-treated tumors were also suppressed, indicating that the combination therapy of DCs and RT evoked systemic antitumor activity. In vitro, the enhancement of gp96 expression was strongly detected by immunostaining after irradiation, DCs with gp96 induced strong cytotoxic activity in vitro, and tumor growth inhibition was observed by direct i.t. injection of gp96. A combination of DCs i.t. and RT can induce a strong antitumor effect not only against treated local tumor but also against non-treated distant tumor, indicating that this treatment can evoke a strong systemic antitumor effect. Gp96 is thought to be one of the target molecules to explain the abscopal effect.


Subject(s)
Antigens, Neoplasm/chemistry , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Dendritic Cells/cytology , Immunotherapy, Adoptive/methods , Membrane Glycoproteins/chemistry , Animals , CD3 Complex/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Female , Mice , Mice, Inbred C3H , Neoplasms, Experimental , Phenotype , Prognosis
12.
Oncol Rep ; 18(3): 585-92, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671705

ABSTRACT

Histone deacetylase inhibitors (HDACIs) are promising therapeutic agents with the potential for regulating cell cycle, differentiation and apoptosis in cancer cells. HDACI activity is associated with selective transcriptional regulation and altering gene expression. However, the exact mechanisms leading to the antitumor effect of HDACIs are not fully understood. FK228, one of the powerful HDACIs, strongly inhibited cell growth of T.Tn and TE2 cells and induced apoptosis. Therefore, comprehensive analysis of the changes in gene expression in human esophageal cancer cell lines by the HDACI FK228 was carried out by microarray analysis. This analysis was used to clarify the expression profiles of genes after exposure to FK228. Of the 4,608 genes analyzed, 93 genes in T.Tn and 65 genes in TE2 were up- or down-regulated 2-fold or more at least at one time point during FK228 exposure and they were classified into four clusters based on their expression patterns. Among them, 15 genes were contained in both cell lines and their expression patterns were similar. Except p21, Prdx1 (reported by us) and IGFBP3, the behaviour/expression of 12 highly responsive genes has still not been reported in esophageal cancer cells. These observations of the expression patterns of functionally classified genes provided insights into the mechanism of the antitumor effect of FK228 in esophageal squamous carcinoma.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Depsipeptides/pharmacology , Gene Expression Profiling , Histone Deacetylase Inhibitors , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Humans , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
13.
Cancer Sci ; 97(1): 57-63, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16367922

ABSTRACT

Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients, although the precise mechanism is controversial. From an immunological point of view, HLA-DR antigen, induced by interferon (IFN)-gamma, is required for tumor-associated antigen recognition by CD4(+) T cells. For instance, as reported previously, the expression of HLA-DR antigen in normal colorectal epithelium immediately adjacent to cancer coincided significantly with the existence of IFN-gamma mRNA in the tissue. From another aspect, IFN-gamma has been revealed to suppress c-myc expression in vivo through a stat1-dependent mechanism, which is important for cell growth, cell cycle and chromosome instability. In the present study, strong HLA-DR-positive expression on cancer cells was significantly related to better prognosis for colorectal cancer patients. High IFN-gamma mRNA expression in situ indicated significantly less activation of c-myc mRNA expression. Further, HLA-DR antigen expression in cancer cells, as well as Dukes stages, was an independent factor for better long-term survival by multivariate analysis. Taken together, IFN-gamma, which induces HLA-DR antigens on the cell surface, also suppresses c-myc expression in situ, and is a possible non-immunological mechanism involved in the better long-term survival of colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , HLA-DR Antigens/metabolism , Interferon-gamma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic , HLA-DR Antigens/genetics , Humans , Interferon-gamma/genetics , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Survival Rate , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...