Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Oncologist ; 29(3): e330-e336, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-37950903

ABSTRACT

BACKGROUND: Trifluridine/tipiracil (FTD/TPI) plus bevacizumab has shown clinical benefit for metastatic colorectal cancer (mCRC) refractory to standard therapy. However, few data have been available for patients with pretreated mCRC who are intolerant of intensive therapy (vulnerable). METHODS: We performed a multicenter retrospective study (WJOG14520G; TWILIGHT) of FTD/TPI plus bevacizumab for vulnerable patients with pretreated mCRC. Eligibility criteria included previous chemotherapy (although patients treated with all key cytotoxic agents, a fluoropyrimidine, oxaliplatin, and irinotecan, were excluded) and intolerance of full-dose combination therapy with oxaliplatin or irinotecan at the start of FTD/TPI plus bevacizumab. RESULTS: The median age of 93 evaluable patients was 79 years (range, 21-90). Intolerance of intensive therapy was attributable to an older age in 60 (65%) patients, serious concomitant disease in 24 (26%) patients, and a poor performance status in 19 (20%) patients. FTD/TPI plus bevacizumab was administered as second-line treatment in 74 (80%) patients and as third- or fourth-line treatment in 19 (20%) patients. The objective response rate was 4.9% (95% confidence interval [CI], 1.4%-12.2%), and the disease control rate was 67.9% (95% CI, 56.6%-77.8%). With a median follow-up time of 21.6 months, median overall survival and progression-free survival were 18.6 months (95% CI, 12.1-23.2) and 6.3 months (95% CI, 5.0-8.3), respectively. Neutropenia of grade ≥3 developed in 50 (54%) patients, whereas 2 (2%) patients experienced febrile neutropenia, and no treatment-related death was observed. CONCLUSION: Our data show the potential efficacy and acceptable safety profile of FTD/TPI plus bevacizumab for vulnerable patients with pretreated mCRC.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Frontotemporal Dementia , Pyrrolidines , Rectal Neoplasms , Thymine , Humans , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Bevacizumab/adverse effects , Colorectal Neoplasms/pathology , Retrospective Studies , Uracil , Oxaliplatin/therapeutic use , Trifluridine/adverse effects , Irinotecan/therapeutic use , Frontotemporal Dementia/chemically induced , Frontotemporal Dementia/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colonic Neoplasms/drug therapy , Rectal Neoplasms/drug therapy , Drug Combinations
2.
J Gen Fam Med ; 18(5): 288-290, 2017 10.
Article in English | MEDLINE | ID: mdl-29264045

ABSTRACT

A 74-year-old man with silicosis was admitted to the hospital because of prolonged fever. After referral to internal medicine for persistent fever and renal dysfunction, workup revealed antineutrophil cytoplasmic antibodies (ANCA) positivity. He was diagnosed with microscopic polyangiitis (MPA). After treatment with immunosuppressive therapy, his condition improved. Herein, we discuss silica exposure and the risk of ANCA-associated vasculitis (AAV), particularly in terms of work-related diseases. Silica exposure is a notorious risk factor for developing AAV, which is potentially lethal when not identified. When we see a silicosis patient with new-onset prolonged fever and generalized fatigue, AAV should be taken into consideration. This case report provides beneficial information to reliably assess patients at high risk of developing AAV in primary care settings.

3.
Invest New Drugs ; 33(5): 1058-67, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26259955

ABSTRACT

BACKGROUND: Trametinib is an inhibitor of MEK1/MEK2 activation and kinase activity. In order to evaluate the safety, tolerability, pharmacokinetics, and preliminary efficacy of single-agent trametinib (part 1) and trametinib in combination with gemcitabine (part 2), we undertook the first clinical study of this combination in Japanese patients with cancer and herein report our results. METHODS: In part 1, 13 patients with advanced solid tumors were enrolled into 3 dose cohorts, receiving trametinib once daily at a dose of 1.0, 2.0, or 3.0 mg. In part 2, 5 patients with pancreatic cancer received trametinib (2.0 mg once daily) in combination with gemcitabine (1000 mg/m(2)). RESULTS: In part 1, a dose-limiting toxicity was observed in a patient in the 2.0-mg dose cohort, but the maximum tolerated dose was not reached at doses up to 3.0 mg daily. The best overall response was a PR in 1 patient, and 6 patients had SD. In part 2, the combination of trametinib and gemcitabine was tolerated for a short period of time. However, serious interstitial lung disease (ILD) was observed in 3 of 5 patients 4 weeks or more after the start of the treatment, including 1 fatal case. Three patients achieved a PR, and 2 patients had SD. The most common adverse event was rash (85 % in part 1 and 100 % in part 2). CONCLUSIONS: Trametinib monotherapy was tolerable in Japanese patients with cancer. However, the combination of trametinib plus gemcitabine carried a higher risk as compared with monotherapy, during which no ILD was observed. (ClinicalTrials.gov number, NCT01324258.).


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Asian People , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Humans , Japan , Male , Maximum Tolerated Dose , Middle Aged , Pancreatic Neoplasms/drug therapy , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Gemcitabine
4.
Cancer Chemother Pharmacol ; 76(2): 279-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050210

ABSTRACT

PURPOSE: The aims of this dose-escalating phase I study were to determine the maximum tolerable dose (MTD) and recommended dose (RD) of 5-fluorouracil (5-FU), docetaxel, and nedaplatin (UDON) combination therapy for future phase II studies, and to evaluate the safety and efficacy of this regimen in patients with untreated recurrent or metastatic esophageal cancer. METHODS: Patients were administered 5-FU on days 1-5, docetaxel on days 1 and 15, and nedaplatin on day 1 at 4-week intervals. The dose levels of 5-FU/docetaxel/nedaplatin were escalated as follows (mg/m(2)): level 1, 800/30/80; level 2, 800/30/90; and level 3, 800/35/90. Toxicity was evaluated using Common Terminology Criteria for Adverse Events version 4.0. RESULTS: Overall, nine patients were enrolled in this study. All patients had an Eastern Cooperative Oncology Group performance status of 0 or 1 and were diagnosed with squamous cell carcinoma. No dose-limiting toxicity was observed at any level, and planned dose escalation was completed without reaching the MTD. No grade 4 or higher toxicity was observed in this study. The observed grade 3 hematological toxicities included neutropenia in five patients (55.6 %) and leukopenia in three patients (33.3 %). None of the patients developed febrile neutropenia, and no grade 3 or 4 non-hematological toxicities were observed. The overall response rate was 77.8 %, including two complete responses, and the disease control rate was 100 %. CONCLUSION: The RD of UDON was identified as level 3. The good tolerability and high antitumor efficacy of this regimen warrant further evaluation in this setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Adult , Aged , Docetaxel , Esophageal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local , Organoplatinum Compounds/administration & dosage , Taxoids/administration & dosage , Young Adult
5.
BMC Res Notes ; 8: 100, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25885256

ABSTRACT

BACKGROUND: Trastuzumab + chemotherapy is considered the standard therapy for advanced human epidermal growth factor receptor 2 (HER2)-positive gastric cancer with mild manageable toxicity, on the basis of the results of a pivotal phase-III trial. Cerebrovascular events are not recognized as expected adverse effects of such therapy. CASE PRESENTATION: We report the case of a 67-year-old, current-smoking male with stage-IV HER2-positive gastric cancer who suffered right middle cerebral artery (MCA) embolism after trastuzumab + chemotherapy. He received trastuzumab and cisplatin on Day 1, followed by a continuous 5-fluorouracil infusion for 5 days as a first-line treatment. Four days after chemotherapy initiation, he presented with left hemiplegia, and brain magnetic resonance imaging and magnetic resonance angiography revealed a right MCA occlusion. No further chemotherapy was administered because of worsening performance status. CONCLUSION: The present case, possibly the first such reported case, suggests the risk of development of embolism after trastuzumab + chemotherapy in HER2-positive advanced gastric cancer, although other factors should be considered.


Subject(s)
Antineoplastic Agents/adverse effects , Intracranial Embolism/pathology , Liver Neoplasms/secondary , Stomach Neoplasms/pathology , Trastuzumab/adverse effects , Aged , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Cisplatin/administration & dosage , Cisplatin/adverse effects , Fatal Outcome , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Gene Expression , Humans , Intracranial Embolism/diagnostic imaging , Intracranial Embolism/drug therapy , Intracranial Embolism/etiology , Liver Neoplasms/complications , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/drug therapy , Lymphatic Metastasis , Male , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/pathology , Neoplasm Staging , Radiography , Receptor, ErbB-2/genetics , Stomach Neoplasms/complications , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Trastuzumab/administration & dosage
6.
Cancer Sci ; 106(6): 734-739, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25786335

ABSTRACT

We conducted a phase I study of a weekly nab-paclitaxel and S-1 combination therapy in patients with human epidermal growth factor receptor type 2-negative metastatic breast cancer. The primary objective was to estimate the maximum tolerated and recommended doses. Each treatment was repeated every 21 days. Levels 1, 2a, 2b, and 3 were set depending on the S-1 dose (65 or 80 mg/m(2) ) and nab-paclitaxel infusion schedule (days 1 and 8 or days 1, 8, and 15). Fifteen patients were enrolled. Dose-limiting toxicity was observed in one patient at Level 3 (100 mg/m(2) nab-paclitaxel on days 1, 8, and 15 with 80 mg/m(2) S-1 daily for 14 days, followed by 7 days of rest). Although the maximum tolerated dose was not reached, the recommended dose was determined to be Level 3. Neutropenia was the most frequent grade 3-4 treatment-related adverse event. For patients with measurable lesions, the response rate was 50.0% and the median time to treatment failure and median progression-free survival was 13.2 and 21.0 months, respectively. The present results show the feasibility and potential for long-term administration of this combination therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/analysis , Adult , Aged , Albumins/administration & dosage , Albumins/adverse effects , Albumins/pharmacokinetics , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Drug Administration Schedule , Drug Combinations , Female , Humans , Middle Aged , Oxonic Acid/administration & dosage , Oxonic Acid/adverse effects , Oxonic Acid/pharmacokinetics , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Tegafur/administration & dosage , Tegafur/adverse effects , Tegafur/pharmacokinetics
7.
PLoS One ; 9(7): e101902, 2014.
Article in English | MEDLINE | ID: mdl-25020203

ABSTRACT

BACKGROUND: Nephrotoxicity remains a problem for patients who receive cisplatin chemotherapy. We retrospectively evaluated potential risk factors for cisplatin-induced nephrotoxicity as well as the potential impact of intravenous magnesium supplementation on such toxicity. PATIENTS AND METHODS: We reviewed clinical data for 401 patients who underwent chemotherapy including a high dose (≥60 mg/m2) of cisplatin in the first-line setting. Nephrotoxicity was defined as an increase in the serum creatinine concentration of at least grade 2 during the first course of cisplatin chemotherapy, as assessed on the basis of National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. The severity of nephrotoxicity was evaluated on the basis of the mean change in the serum creatinine level. Magnesium was administered intravenously to 67 patients (17%). RESULTS: Cisplatin-induced nephrotoxicity was observed in 127 patients (32%). Multivariable analysis revealed that an Eastern Cooperative Oncology Group performance status of 2 (risk ratio, 1.876; P = 0.004) and the regular use of nonsteroidal anti-inflammatory drugs (NSAIDs) (risk ratio, 1.357; P = 0.047) were significantly associated with an increased risk for cisplatin nephrotoxicity, whereas intravenous magnesium supplementation was associated with a significantly reduced risk for such toxicity (risk ratio, 0.175; P = 0.0004). The development of hypomagnesemia during cisplatin treatment was significantly associated with a greater increase in serum creatinine level (P = 0.0025). Magnesium supplementation therapy was also associated with a significantly reduced severity of renal toxicity (P = 0.012). CONCLUSIONS: A relatively poor performance status and the regular use of NSAIDs were significantly associated with cisplatin-induced nephrotoxicity, although the latter association was marginal. Our findings also suggest that the ability of magnesium supplementation to protect against the renal toxicity of cisplatin warrants further investigation in a prospective trial.


Subject(s)
Cisplatin/adverse effects , Dietary Supplements , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Magnesium/pharmacology , Neoplasms/drug therapy , Cisplatin/therapeutic use , Creatinine/blood , DNA Mutational Analysis , Humans , Multivariate Analysis , Odds Ratio , Retrospective Studies , Risk Factors
8.
Anticancer Res ; 33(12): 5699-705, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24324120

ABSTRACT

AIM: We performed a pharmacokinetic phase I trial of the combination of S-1 granules and nedaplatin for head and neck squamous cell carcinoma (HNSCC). PATIENTS AND METHODS: Patients were treated with both nedaplatin on day 1 at a dose starting at 80 mg/m(2) (level 1) escalating up to 90 mg/m(2) (level 2), and S-1 granules at a daily dose of 80 mg/m(2) on days 1 to 14 every three weeks. The primary end-point was determination of the recommended dose. RESULTS: Twenty patients were enrolled. Dose-limiting toxicities occurred in one out of six patients at dose level 1 (neutropenia) and in all three patients at level 2 (neutropenia and thrombocytopenia). The recommended dose was determined as level 1. Pharmacokinetic parameters of S-1 granule did not differ from the capsula formulation. The response rate was 42.1%. CONCLUSION: This combination was well-tolerated and manifested a promising activity against HNSCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Organoplatinum Compounds/pharmacokinetics , Oxonic Acid/pharmacokinetics , Tegafur/pharmacokinetics , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Combinations , Female , Humans , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Oxonic Acid/administration & dosage , Oxonic Acid/therapeutic use , Tegafur/administration & dosage , Tegafur/therapeutic use
9.
Mol Oncol ; 7(4): 840-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23664167

ABSTRACT

BACKGROUND & AIM: Runt-related transcription factor 3 (RUNX3) is a tumor suppressor gene that is expressed in gastric and other cancers including pancreatic cancer. However, the precise function of RUNX3 in pancreatic cancer has not been fully elucidated. In this study, we aimed to determine the effect of decreased RUNX3 expression in pancreatic cancer. METHODS: This study included 36 patients with primary pancreatic cancer, who had undergone pancreaticoduodenectomy. All patients were treated with 1000 mg/m2 gemcitabine after the surgery. The pancreatic cancer cell lines PANC-1, MIAPaCa-2, BxPC-3, SUIT-2, and KLM-1 were used for immunoblotting analysis of RUNX3 and multidrug resistance protein (MRP) expressions. Ectopic RUNX3 expression was achieved by cDNA transfection of the cells, and small interfering RNA (siRNA) against RUNX3 was used to knock down endogenous RUNX3. Cell growth in the presence of gemcitabine was assessed using the MTT assay. RESULTS: Patients with RUNX3-positive and RUNX3-negative pancreatic cancer had a median survival of 1006 and 643 days, respectively. Exogenous RUNX3 expression reduced the expression of MRP1, MRP2, and MRP5 in endogenous RUNX3-negative cells, whereas RUNX3 siRNA increased the expressions of these genes in endogenous RUNX3-positive cells. Exogenous RUNX3 expression decreased gemcitabine IC50 in RUNX3-negative cells. CONCLUSION: Loss of RUNX3 expression contributes to gemcitabine resistance by inducing MRP expression, thereby resulting in poor patient survival.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/genetics , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Immunoblotting , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , RNA, Small Interfering , Gemcitabine , Pancreatic Neoplasms
10.
Dig Dis ; 30(6): 541-6, 2012.
Article in English | MEDLINE | ID: mdl-23258092

ABSTRACT

OBJECTIVE: A number of studies have reported reactivation of hepatitis B during intensive immunosuppressive therapy such as cases of hematological malignancy, whereas little has been reported for characteristics of reactivation triggered by chemotherapy for solid cancer. METHODS: A total of 130 patients underwent chemotherapy for treatments of common solid cancer between May 2011 and May 2012 at Kinki University Hospital. Among them, 27 patients were suspected for a past infection of hepatitis B virus (HBV), showing positive for hepatitis B core antibody or surface antibody but negative for hepatitis B surface antigen, and were eligible for this study. RESULTS: Hepatitis B reactivation was observed in 2 of 27 cases (7.4%). The duration between the start of chemotherapy and increase of serum HBV load was 30 days in both cases. CONCLUSIONS: We reported the 2 cases of hepatitis B reactivation receiving chemotherapy for solid cancer in terms of patterns and characteristics of reactivation. Accumulation of such cases will help in clarifying the clinical importance of hepatitis B reactivation during treatment of solid malignancies.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/virology , Neoplasms/drug therapy , Virus Activation , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , DNA, Viral/blood , Female , Hepatitis B/complications , Hepatitis B virus/drug effects , Humans , Male , Middle Aged , Neoplasms/complications , Viral Load
11.
Nihon Rinsho ; 70(12): 2093-7, 2012 Dec.
Article in Japanese | MEDLINE | ID: mdl-23259379

ABSTRACT

Advances in technology of antibody modification, such as chimeric antibody or humanized antibody against cancer specific proteins, allows us to apply them into the clinic efficiently. Most therapeutic antibodies inhibit proliferation of cancer cells by blockade of their receptors or ligands as well as related signal transductions. In addition to cancer cells, therapeutic antibodies are able to target vascular endothelial cells and immune cells, exerting their anti-tumor effects by inhibition of angiogenesis and enhancement of immune system. Recently, new therapeutic antibodies have been developed, which are able to induce cell death by binding to a cytotoxin or a radioisotope. In this review, we summarize the mechanisms of therapeutic antibodies by classifying their actions in regard to blocking, targeting, and signaling.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Humans , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Neoplasms/blood supply , Neoplasms/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Signal Transduction/immunology
13.
J Gastroenterol Hepatol ; 27(10): 1602-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22554292

ABSTRACT

BACKGROUND AND AIM: Hepatocellular carcinoma (HCC) is a hypervascular tumor, and angiogenesis plays an important role in its development. Previously, we demonstrated that des-γ-carboxyl prothrombin (DCP) promotes both cell proliferation and migration of human umbilical vein endothelial cells (HUVECs) by inducing the autophosphorylation of kinase insert domain receptor (KDR). In the present study, DCP-associated tumor angiogenesis was assessed by comparing hypovascular and common hypervascular HCC. METHODS: The solitary HCCs of 827 patients were classified into two groups according to the tumor density at the arterial phase of a dynamic computed tomography scan; the initial clinical data of patients with the hyper- and hypovascular types were compared. The HCC tissues from 95 tumors were analyzed by immunohistochemical staining for DCP and phosphorylated KDR, and intratumoral microvessel density (MVD) was analyzed to evaluate microvessel angiogenesis. RESULTS: The serum DCP levels (320 ± 3532 mAU/mL) and tumor size (18.4 ± 9.0 mm) of patients with hypervascular HCC were significantly greater than those with hypovascular HCC (38.7 ± 80 mAU/mL and 14.6 ± 5.2 mm, P < 0.001). Immunohistochemical analysis revealed that the expressions of DCP and phospho-KDR were significantly greater in hypervascular HCC (71.4% and 31.0%, respectively) than in hypovascular HCC (7.6% and 5.7%, respectively). Intratumoral MVD was significantly correlated with DCP (r = 0.48, P < 0.0001). CONCLUSIONS: des-γ-carboxyl prothrombin production is associated with tumor angiogenesis in HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Biomarkers/metabolism , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Neovascularization, Pathologic , Protein Precursors/metabolism , Prothrombin/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Biomarkers, Tumor/blood , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/blood , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Phosphorylation , Protein Precursors/blood , Tomography, X-Ray Computed , Transfection , Tumor Burden , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Int J Cancer ; 131(11): 2537-46, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22488108

ABSTRACT

Loss or decreased expression of runt-related transcription factor 3 (RUNX3), a tumor suppressor gene involved in gastric and other cancers, has been frequently observed in hepatocellular carcinoma (HCC). The objective of this study was to identify the regulatory mechanism of the epithelial-mesenchymal transition (EMT) by RUNX3 in HCC. Human HCC cell lines, Hep3B, Huh7, HLF and SK-Hep1, were divided into low- and high-EMT lines, based on their expression of TWIST1 and SNAI2, and were used in this in vitro study. Ectopic RUNX3 expression had an anti-EMT effect in low-EMT HCC cell lines characterized by increased E-cadherin expression and decreased N-cadherin and vimentin expression. RUNX3 expression has previously been reported to reduce jagged-1 (JAG1) expression; therefore, JAG1 ligand peptide was used to reinduce EMT in RUNX3-expressing low-EMT HCC cells. Immunohistochemical analyses were performed for RUNX3, E-cadherin, N-cadherin and TWIST1 in 33 human HCC tissues, also divided into low- and high-EMT HCC, based on TWIST1 expression. E-cadherin expression was correlated positively and N-cadherin expression was correlated negatively with RUNX3 expression in low-EMT HCC tissues. Correlations between EMT markers and RUNX3 mRNA expression were analyzed using Oncomine datasets. Similarly, mRNA expression of E-cadherin was also significantly correlated with that of RUNX3 in low-EMT HCC, while mRNA expression of JAG1 was negatively correlated with that of RUNX3. These results suggest a novel mechanism by which loss or decreased expression of RUNX3 induces EMT via induction of JAG1 expression in low-EMT HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Core Binding Factor Alpha 3 Subunit/biosynthesis , Epithelial-Mesenchymal Transition , Liver Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/genetics , Serrate-Jagged Proteins , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Vimentin/genetics , Vimentin/metabolism
15.
Oncol Rep ; 26(3): 523-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21637926

ABSTRACT

Runt-related transcription factor 3 (RUNX3) is a candidate tumor suppressor gene that is downregulated in various cancers. In the present study, we analyzed the regulatory function of RUNX3 on Jagged-1 (JAG1) expression and cancer stem cell (CSC) signaling in hepatocellular carcinoma (HCC). Eleven HCC cell lines and 30 human HCC tissues were used. RUNX3 and JAG1 expression levels were analyzed by immunoblotting and immunohistochemistry. Ectopic RUNX3 expression was induced by introducing RUNX3 cDNA into the RUNX3-negative HCC cell line Hep3B and Huh7 cells. Furthermore endogenous RUNX3 expression was knocked down by RUNX3 siRNA in SK-Hep-1 cells. In order to analyze JAG1 transcriptional regulation, we conducted reporter assays, chromatin immunoprecipitation (ChIP) assays and electrophoretic mobility shift assays (EMSAs). Tumorigenicity was analyzed using a SCID mouse liver injection model. An inverse correlation was observed between RUNX3 expression and JAG1 expression in most HCC cell lines and tissues. Restoring RUNX3 expression decreased the expression of JAG1 in Hep3B and Huh7 cells, whereas JAG1 expression was upregulated in RUNX3 siRNA-treated SK-Hep-1 cells. Reporter assays, ChIP assays and EMSAs revealed that RUNX3 directly bound to the transcriptional regulatory region of JAG1 and suppressed JAG1 transcription. Moreover, RUNX3 restoration downregulated CSCs by suppressing JAG1-mediated Notch signaling. The tumorigenic capacity of RUNX3-expressing Hep3B cells was lower compared to that of control Hep3B cells. RUNX3 expression suppressed JAG1 expression and resulted in downregulation of tumorigenesis by suppression of JAG1-mediated CSCs.


Subject(s)
Calcium-Binding Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Recombinant Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Liver Neoplasms/pathology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Male , Membrane Proteins/genetics , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Recombinant Proteins/metabolism , Regulatory Elements, Transcriptional , Serrate-Jagged Proteins , Signal Transduction , Transcription, Genetic , Transplantation, Heterologous , Tumor Burden , Young Adult
16.
J Cell Biochem ; 112(3): 745-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21328447

ABSTRACT

Runt-related transcription factor 3 (RUNX3) is reported as a tumor suppressor gene for gastric cancer, and may be important in the development of hepatocellular carcinoma (HCC). RUNX3 expression is frequently lost or decreased by hemizygous deletion or hypermethylation of its promoter lesion in HCC. The significance of decreased expression of RUNX3 in HCC has not been fully elucidated, but is likely related to dysfunction of cell cycle regulation, decrement of apoptosis, enhancement of angiogenesis, and development of epithelial-mesenchymal transition. RUNX3 is a promising candidate as a tumor suppressor gene for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Core Binding Factor Alpha 3 Subunit/genetics , Liver Neoplasms/pathology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Core Binding Factor Alpha 3 Subunit/physiology , DNA Methylation , Down-Regulation , Epithelial-Mesenchymal Transition , Genes, Neoplasm , Humans , Liver Neoplasms/metabolism , Loss of Heterozygosity , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic
17.
BMC Cancer ; 11: 3, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21205319

ABSTRACT

BACKGROUND: Runt-related transcription factor 3 (RUNX3) is known as a tumor suppressor gene for gastric cancer and other cancers, this gene may be involved in the development of hepatocellular carcinoma (HCC). METHODS: RUNX3 expression was analyzed by immunoblot and immunohistochemistry in HCC cells and tissues, respectively. Hep3B cells, lacking endogenous RUNX3, were introduced with RUNX3 constructs. Cell proliferation was measured using the MTT assay and apoptosis was evaluated using DAPI staining. Apoptosis signaling was assessed by immunoblot analysis. RESULTS: RUNX3 protein expression was frequently inactivated in the HCC cell lines (91%) and tissues (90%). RUNX3 expression inhibited 90±8% of cell growth at 72 h in serum starved Hep3B cells. Forty-eight hour serum starvation-induced apoptosis and the percentage of apoptotic cells reached 31±4% and 4±1% in RUNX3-expressing Hep3B and control cells, respectively. Apoptotic activity was increased by Bim expression and caspase-3 and caspase-9 activation. CONCLUSION: RUNX3 expression enhanced serum starvation-induced apoptosis in HCC cell lines. RUNX3 is deleted or weakly expressed in HCC, which leads to tumorigenesis by escaping apoptosis.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Liver Neoplasms/metabolism , Adolescent , Adult , Aged , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Core Binding Factor Alpha 3 Subunit/genetics , Culture Media, Serum-Free/pharmacology , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Immunoblotting , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
18.
BMC Cancer ; 9: 240, 2009 Jul 18.
Article in English | MEDLINE | ID: mdl-19615090

ABSTRACT

BACKGROUND: Twist, a transcription factor of the basic helix-loop-helix class, is reported to regulate cancer metastasis. It is known to induce epithelial-mesenchymal transition (EMT). In this study, we evaluated the expression of twist and its effect on cell migration in hepatocellular carcinoma (HCC). METHODS: We examined twist expression using immunohistochemistry in 20 tissue samples of hepatocellular carcinoma, and assessed twist expression in HCC cell lines by RT-PCR and Western blot analysis. Ectopic twist expression was created by introducing a twist construct in the twist-negative HCC cell lines. Endogenous twist expression was blocked by twist siRNA in the twist-positive HCC cell lines. We studied EMT related markers, E-cadherin, Vimentin, and N-cadherin by Western blot analysis. Cell proliferation was measured by MTT assay, and cell migration was measured by in vitro wound healing assay. We used immunofluorescent vinculin staining to visualize focal adhesion. RESULTS: We detected strong and intermediate twist expression in 7 of 20 tumor samples, and no significant twist expression was found in the tumor-free resection margins. In addition, we detected twist expression in HLE, HLF, and SK-Hep1 cells, but not in PLC/RPF/5, HepG2, and Huh7 cells. Ectopic twist-expressing cells demonstrated enhanced cell motility, but twist expression did not affect cell proliferation. Twist expression induced epithelial-mesenchymal transition together with related morphologic changes. Focal adhesion contact was reduced significantly in ectopic twist-expressing cells. Twist-siRNA-treated HLE, HLF, and SK-Hep1 cells demonstrated a reduction in cell migration by 50, 40 and 18%, respectively. CONCLUSION: Twist induces migratory effect on hepatocellular carcinoma by causing epithelial-mesenchymal transition.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Movement/physiology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Nuclear Proteins/biosynthesis , Twist-Related Protein 1/biosynthesis , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/pathology , Cell Adhesion/physiology , Cell Line, Tumor , Female , Gene Silencing , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Twist-Related Protein 1/genetics
19.
Mol Oncol ; 2(3): 241-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19383345

ABSTRACT

Using GGCX gene-specific real-time PCR, exon 2 deletion splice variant of vitamin K-dependent gamma-glutamyl carboxylase (GGCX) mRNA was identified in HCC cell lines. Expressions of wild type and exon 2 deletion variant of GGCX were analyzed with relevance to DCP production in HCC cell lines. Hep3B, HepG2, HuH1, HuH7, and PLC/PRF/5 produced DCP, while SK-Hep-1, HLE, HLF, and JHH1 produced no detectable level of DCP. DCP-producing cells expressed exon 2 deletion variant of GGCX mRNA and protein, while DCP-negative cells expressed no detectable level of exon 2 deletion variant of GGCX. These results suggest that exon 2 deletion splice variant of GGCX causes dysfunction of GGCX enzyme activity resulting in DCP production in HCC cell lines.


Subject(s)
Carbon-Carbon Ligases/physiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Protein Precursors/biosynthesis , Prothrombin/biosynthesis , Biomarkers , Carbon-Carbon Ligases/genetics , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Exons , Genetic Variation , Humans , Isoenzymes/genetics , Polymerase Chain Reaction , RNA, Messenger/analysis
20.
J Biol Chem ; 282(12): 8741-8, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17255102

ABSTRACT

Des-gamma-carboxyl prothrombin (DCP) is a well recognized tumor marker for hepatocellular carcinoma. Previously, we have demonstrated that DCP stimulates cell proliferation in hepatocellular carcinoma cell lines through Met-Janus kinase 1 signal transducer and activator of transcription 3 signaling pathway. In the present study, we demonstrated that DCP induces both cell proliferation and migration in human umbilical vein endothelial cells. DCP was found to bind with the kinase insert domain receptor (KDR), alternatively referred to as vascular endothelial growth factor receptor-2. Furthermore, DCP induced autophosphorylation of KDR and its downstream effector phospholipase C-gamma and mitogen-activated protein kinase (MAPK). To support these results, we showed that DCP-induced cell proliferation and cell migration were inhibited by KDR short interfering RNA, KDR kinase inhibitor, or MAPK inhibitor. In conclusion, these results indicate that DCP is a novel type of vascular endothelial growth factor that possesses potent mitogenic and migrative activities.


Subject(s)
Biomarkers/chemistry , Endothelium, Vascular/cytology , Protein Precursors/chemistry , Prothrombin/chemistry , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Silencing , Humans , Janus Kinase 1/metabolism , MAP Kinase Signaling System , Phospholipase C gamma/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...