Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Taro Matsuzaki is active.

Publication


Featured researches published by Taro Matsuzaki.


Cancer Science | 2009

DNA methyltransferase inhibitor 5-aza-CdR enhances the radiosensitivity of gastric cancer cells

Hong Qiu; Masakazu Yashiro; Osamu Shinto; Taro Matsuzaki; Kosei Hirakawa

The National Comprehensive Cancer Network guidelines recommend radiotherapy as a standard treatment for patients with a high risk of recurrence in gastric cancer. Because radiation is harmful to the surrounding organs, a radiation sensitizer might therefore be useful to decrease the side effects of patients with advanced gastric carcinoma. The aim of the current study was to clarify the effect of a DNA methyltransferase inhibitor, 5‐aza‐2′‐deoxycytidine (CdR), on radiation sensitivity in gastric cancer cells. Five gastric cancer cell lines, OCUM‐2M, OCUM‐12, KATO‐III, MKN‐45, and MKN‐74, were used. The effects of 5‐aza‐CdR with irradiation on the growth activity, cell‐cycle distribution, apoptosis, and apoptosis‐associated gene expression were examined. 5‐aza‐CdR sensitized three of five gastric cancer cell lines to radiation. A combination of irradiation and 5‐aza‐CdR significantly (P < 0.05) decreased the growth activity compared with irradiation alone in OCUM‐2M, OCUM‐12, and MKN‐45 cells, but not in KATO‐III and MKN‐74 cells. The percentage of cells in G2–M phase and the apoptotic rate with irradiation in combination with 5‐aza‐CdR were increased in OCUM‐2M, OCUM‐12, and MKN‐45 cells compared with irradiation alone, but not in KATO‐III and MKN‐74 cells. 5‐aza‐CdR increased the expression of p53, RASSF1, and death‐associated protein kinases (DAPK) genes compared with the control or irradiation alone. These findings suggest that 5‐aza‐CdR might therefore be useful as a radiation sensitizer to treat some types of gastric carcinoma. The arrest at G2–M phase and increased apoptotic rate might be partly mediated by enhanced expression of the p53, RASSF1, or DAPK gene families by 5‐aza‐CdR. (Cancer Sci 2009; 100: 181–188)


International Journal of Cancer | 2009

Synergistic antitumor effects of FGFR2 inhibitor with 5-fluorouracil on scirrhous gastric carcinoma

Masakazu Yashiro; Osamu Shinto; Kazunori Nakamura; Masashige Tendo; Tasuku Matsuoka; Taro Matsuzaki; Ryoji Kaizaki; Atsushi Miwa; Kosei Hirakawa

Scirrhous gastric carcinoma (SGC) carries the highest mortality because of a frequent metastasis to lymph node (LN). S1, a 5‐fluorouracil (5‐FU) analog, is clinically available for gastric cancer at an advanced stage. Fibroblast growth factor receptor 2 (FGFR2) is required for the proliferation of SGC. The objective of this study is to clarify the benefit of a combination of S1 and kinase inhibitors including FGFR2 inhibitor Ki23057 in gastric cancer. OCUM‐2MLN and KATO‐III were derived from SGC. MKN‐7 and MKN‐74 were derived from non‐SGC. MTT assay was used to examine the growth‐inhibitory activity of 5 small‐synthetic molecules including Ki23057, Sunitinib, Glivec, Lapatinib or SU11274, in cells cultured with 5‐FU. Combination effects of 5‐FU with Ki23057 on proliferation, apoptosis and mRNA expression were examined. S1 and/or Ki23057 were administered to murine models of SGC created by the orthotopic inoculation of OCUM‐2MLN cells. Ki23057 at 100 nM significantly (p < 0.01) inhibited the proliferation and decreased the phosphorylation of FGFR2 in SGC cells, but not in non‐SGC. Ki23057 showed synergistic antitumor effects for SGC cells in combination with 5‐FU using CalcuSyn analysis, but Sunitinib, Glivec, Lapatinib and SU11274 did not. The combination of Ki23057 and 5‐FU decreased DPD expression and increased apoptosis rates and p21 expression level of SGC cells. The combined administration of S1 and Ki23057 significantly (p < 0.05) decreased orthotopic tumors as well as LN metastasis more effectively than S1 alone. These findings suggested that the combined treatment with 5‐FU and Ki23057 produced synergistic antitumor effects and is therapeutically promising for SGC treatment.


Cancer Science | 2009

Synergistic antiproliferative effect of mTOR inhibitors in combination with 5-fluorouracil in scirrhous gastric cancer.

Taro Matsuzaki; Masakazu Yashiro; Ryoji Kaizaki; Koichi Yasuda; Yosuke Doi; Tetsuji Sawada; Masaichi Ohira; Kosei Hirakawa

The aim of this study is to clarify the benefit of combination chemotherapy in gastric cancer based on a cell‐signal inhibitor and an anticancer drug. Two scirrhous gastric cancer cell lines and two non‐scirrhous gastric cancer cell lines were used. Five anticancer drugs (5‐fluorouracil [5FU], paclitaxel, oxaliplatin, irinotecan, and gemcitabine) and four cell‐signal inhibitors, mammalian target of rapamycin (mTOR) inhibitor, glycogen synthase kinase 3β, p38αβMAPK, and cyclin‐dependent kinase, were used. The proliferation of cancer cells was examined by MTT assay and in vivo study. The apoptosis of cancer cells and the expression of apoptosis‐related molecules were examined by flow cytometry, real‐time PCR, and immunostaining. mTOR inhibitors with 5FU showed a synergistic antiproliferative effect in scirrhous gastric cancer, whereas the other signal inhibitors showed no synergistic effect with any anticancer drugs. mTOR inhibitor decreased the IC50 of 5FU and increased the apoptosis rate in scirrhous gastric cancer cells, but not in non‐scirrhous gastric cancer cells. The pan‐caspase inhibitor, zVAD‐fmk, inhibits apoptosis induced in combination with 5FU and mTOR inhibitor. mTOR inhibitor decreased dihydropyrimidine dehydrogenase, thymidylatesynthase, and bcl‐2expression, and increased caspase‐3 and p21 expression of scirrhous gastric cancer cells, but did not affect those of non‐scirrhous gastric cancer cells. In an in vivo study, mTOR inhibitor significantly enhanced the therapeutic efficacy of S1, an analog of 5FU. These findings suggest that mTOR inhibitor interacts with 5FU in a synergistic manner in scirrhous gastric cancer cells by the activation of the apoptosis signal. Therefore, mTOR inhibitor is a promising therapeutic agent in combination with 5FU in scirrhous gastric cancer. (Cancer Sci 2009; 100: 2402–2410)


British Journal of Cancer | 2013

A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan in gastric carcinoma

Masakazu Yashiro; Takafumi Nishii; Tsuyoshi Hasegawa; Taro Matsuzaki; Tamami Morisaki; Tatsunari Fukuoka; Kosei Hirakawa

Background:Cancer stem cells (CSCs) may be postulated mediators of the chemoresistance. This study aimed to determine an effective signal inhibitor with effects on the proliferation of CSCs in combination with anticancer drugs.Methods:We used three gastric cancer cell lines and three side population (SP)-enriched CSC cell lines. We examined the combined effects of inhibitors against stemness signals, including c-Met inhibitor SU11274, and five anticancer drugs on the CSC proliferation and mRNA expression of chemoresistance-associated genes.Results:The IC50 of irinotecan in SP-enriched CSC was 10.5 times higher than parent OCUM-2M cells, whereas that of oxaliplatin, taxol, gemcitabine, and 5-fluorouracil was 2.0, 2.8, 2.0, and 1.2, respectively. The SP cell lines had higher expression levels of UGT1A1, ABCG2, and ABCB1 than their parent cell lines. There was a synergistic antiproliferative effect with a combination of SU11274 and SN38 in SP cells, but not other inhibitors. The SU11274 significantly decreased the expression of UGT1A1, but not ABCG2 and ABCB1. The SN38 plus SU11274 group more effectively suppressed in vivo tumour growth by OCUM-2M/SP cells than either group alone.Conclusion:Cancer stem cells have chemoresistance to irinotecan. The c-Met inhibitor may be a promising target molecule for irinotecan-based chemotherapy of gastric cancer.


BMC Cancer | 2010

Phosphorylated Smad2 in Advanced Stage Gastric Carcinoma

Osamu Shinto; Masakazu Yashiro; Takahiro Toyokawa; Takafumi Nishii; Ryoji Kaizaki; Taro Matsuzaki; Satoru Noda; Naoshi Kubo; Hiroaki Tanaka; Yosuke Doi; Masaichi Ohira; Kazuya Muguruma; Tetsuji Sawada; Kosei Hirakawa

BackgroundTransforming growth factor β (TGFβ) receptor signaling is closely associated with the invasion ability of gastric cancer cells. Although Smad signal is a critical integrator of TGFβ receptor signaling transduction systems, not much is known about the role of Smad2 expression in gastric carcinoma. The aim of the current study is to clarify the role of phosphorylated Smad2 (p-Smad2) in gastric adenocarcinomas at advanced stages.MethodsImmunohistochemical staining with anti-p-Smad2 was performed on paraffin-embedded specimens from 135 patients with advanced gastric adenocarcinomas. We also evaluated the relationship between the expression levels of p-Smad2 and clinicopathologic characteristics of patients with gastric adenocarcinomas.ResultsThe p-Smad2 expression level was high in 63 (47%) of 135 gastric carcinomas. The p-Smad2 expression level was significantly higher in diffuse type carcinoma (p = 0.007), tumours with peritoneal metastasis (p = 0.017), and tumours with lymph node metastasis (p = 0.047). The prognosis for p-Smad2-high patients was significantly (p = 0.035, log-rank) poorer than that of p-Smad2-low patients, while a multivariate analysis revealed that p-Smad2 expression was not an independence prognostic factor.ConclusionThe expression of p-Smad2 is associated with malignant phenotype and poor prognosis in patients with advanced gastric carcinoma.


British Journal of Cancer | 2009

Effects of VEGFR-3 phosphorylation inhibitor on lymph node metastasis in an orthotopic diffuse-type gastric carcinoma model

Masakazu Yashiro; Osamu Shinto; Kazunori Nakamura; Masashige Tendo; Tasuku Matsuoka; Taro Matsuzaki; Ryoji Kaizaki; Masaichi Ohira; Atsushi Miwa; Kosei Hirakawa

Background:Vascular endothelial growth factor receptor-3 (VEGFR-3) signalling mediates lymphangiogenesis and lymphatic invasion; however, the effect of VEGFR-3 inhibition on the lymph node (LN) metastasis remains unclear. The aim of this study is to clarify the benefit of a VEGFR-3 inhibitor Ki23057 for LN metastasis.Methods:Ki23057 was administered orally to gastric cancer models created by orthotopic inoculation of diffuse-type gastric cancer cells, OCUM-2MLN. The effects of Ki23057 on lymphatic vessel invasion, lymphatic vessel density, and VEGFR-3 phosphorylation were examined by immunostaining or immunoblotting.Results:Ki23057 inhibited the autophosphorylation of VEGFR-3, with IC50 values of 4.3 nM in the cell-free kinase assay. Murine gastric cancer models created by the orthotopic inoculation of OCUM-2MLN cells showed the diffusely infiltrating growth and frequently developed LN metastasis. The oral administration of Ki23057 significantly (P<0.01) reduced the size of orthotopic tumours and the number of the metastatic LN in gastric cancer models. The degree of lymphatic invasion and lymphangiogenesis was significantly (P<0.05) lower in the gastric tumours treated by Ki23057. Ki23057 inhibited the phosphorylation of VEGFR-3 of lymphatic endothelial cells in gastric tumours.Conclusion:The inhibition of lymphangiogenesis targeting VEGFR-3 phosphorylation is a therapeutic strategy for inhibiting LN metastasis of diffuse-type gastric cancer.


British Journal of Cancer | 2011

An EGFR inhibitor enhances the efficacy of SN38, an active metabolite of irinotecan, in SN38-refractory gastric carcinoma cells

Masakazu Yashiro; H Qiu; Tsuyoshi Hasegawa; Xiaotian Zhang; Taro Matsuzaki; Kosei Hirakawa

Background:Acquired drug resistance to irinotecan is one of the significant obstacles in the treatment of advanced gastric cancer. This study was performed to clarify the effect of epidermal growth factor receptor (EGFR) inhibitors in combination with SN38, an active metabolite of irinotecan, on the proliferation of irinotecan-refractory gastric cancer.Methods:Two irinotecan-resistant gastric cancer cell lines, OCUM-2M/SN38 and OCUM-8/SN38 were, respectively, established by stepwise exposure to SN38 from the parent gastric cancer cell lines OCUM-2M and OCUM-8. The combination effects of two EGFR inhibitors, gefitinib and lapatinib, with SN38 on proliferation, apoptosis, and cell cycle on gastric cancer cells were examined.Results:Gefitinib or lapatinib showed synergistic anti-tumour effects against OCUM-2M/SN38 and OCUM-8/SN38 cells when used in combination with SN38, but not against OCUM-2M or OCUM-8 cells. SN38 increased the expression of EGFR and HER2 in OCUM-2M/SN38 and OCUM-8/SN38 cells. The combination of an EGFR inhibitor and SN38 significantly increased the levels of apoptosis-related molecules, caspase-6, p53, and DAPK-2, and resulted in the induction of apoptosis of irinotecan-resistant cells. The EGFR inhibitors increased the S-phase and decreased the UGT1A1 and ABCG expression in irinotecan-resistant cells. The SN38 plus Lapatinib group more effectively suppressed in vivo tumour growth by OCUM-2M/SN38 cells than either alone group.Conclusion:The combination treatment with an EGFR inhibitor and irinotecan might produce synergistic anti-tumour effects for irinotecan-refractory gastric cancer cells. The regulation of SN38 metabolism-related genes and cell cycle by EGFR inhibitors might be responsible for the synergism.


Cancer Research | 2013

Abstract 225: A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan active metabolite SN38 in gastric carcinoma.

Masakazu Yashiro; Tatsunari Fukuoka; Haruhito Kinoshita; Takafumi Nishii; Tsuyoshi Hasegawa; Taro Matsuzaki; Tamami Morisaki; Kosei Hirakawa

Purpose: Gastric cancer remains a major global health threat and most patients with advanced stage disease require chemotherapy. The development of drug resistance is a major obstacle in the treatment of gastric cancer and only few effective therapies for combating chemoresistance are currently available. It has been demonstrated that a small subset of cancer cells with stem cell properties, referred to as “cancer stem cells” (CSCs), survive intensive anticancer therapies better than proliferating progenitor cells or differentiated tumor cells. CSCs have been reported to be postulated mediators of chemoresistance, so it might be important to comprehend the drug resistance mechanisms of CSCs to develop a promising therapy to combat chemoresistance. The stemness signal might be associated with the chemosensitivity of CSCs. In the present study, we analyzed the effect of c-Met inhibitors on the chemosensitivity of stem-like cancer cells in gastric cancer. We demonstrated that a c-Met inhibitor synergistically increased the antitumor activity of SN38 in CSCs. To determine mechanisms underlying this observed synergism, we observed that a c-Met inhibitor combined with SN38 also led to a significant increase in UGT1A1 and its subsequent interaction with apoptosis-related genes, i.e., bcl-2 and caspase-6. Experimental Design: We used three gastric cancer cell lines and three side population (SP)-enriched cell lines. We used three signal inhibitors, c-Met inhibitor SU11274, GSK3β inhibitor AR-A014418, and mTOR inhibitor rapamycin, and five anticancer drugs. We examined the combined effects of signal inhibitors and anticancer drugs on proliferation, mRNA expression, and cell cycle. Results: The IC50 of irinotecan, oxaliplatin, taxol, and gemcitabine in SP cells were 10.5, 2.0, 2.8, and 2.0 times higher than their parent cells, respectively. In contrast, the IC50 of 5-fluorouracil did not differ between the two cell lines. There was a synergistic anti-proliferative effect with a combination of c-Met inhibitor and SN38 in SP cells. In contrast, the GSK3β inhibitor and mTOR inhibitor had no synergistic effects in combination with any anticancer drugs. The SP cell lines had higher expression levels of UGT1A1, ABCG2, and ABCB1 than their parent cell lines, while the c-Met inhibitor significantly decreased the expression of UGT1A1, but not ABCG2 and ABCB1. G0-phase of SP cells was higher than their parent cells. c-Met inhibition induced S-phase arrest in SP cells. Conclusions: CSCs are associated with multiresistance during chemotherapy. c-Met inhibitors could be a novel strategy to overcome chemoresistance. c-Met inhibitor may be a promising target molecule for irinotecan-based chemotherapy of gastric cancer. A UGT1A1 alteration might be involved in the irinotecan refractory process in CSCs. Citation Format: Masakazu Yashiro, Tatsunari Fukuoka, Haruhito Kinoshita, Takafumi Nishii, Tsuyoshi Hasegawa, Taro Matsuzaki, Tamami Morisaki, Kosei Hirakawa. A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan active metabolite SN38 in gastric carcinoma. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 225. doi:10.1158/1538-7445.AM2013-225


Cancer Research | 2011

Abstract 2552: Synergistic antitumor effects of FGFR2 inhibitor with 5-fluorouracil on diffuse-type gastric carcinoma

Masakazu Yashiro; Osamu Shinto; Kazunori Nakamura; Masashige Tendo; Tasuku Matsuoka; Taro Matsuzaki; Ryoji Kaizaki; Atsushi Miwa; Kosei Hirakawa

Purpose: Diffuse-type gastric carcinoma carries the highest mortality because of a frequent metastasis to lymph node. S1, a 5-fluorouracil (5-FU) analog, is clinically available for gastric cancer at advanced stage. Fibroblast growth factor receptor 2 (FGFR2) is required for the proliferation of diffuse-type gastric carcinoma. The objective of this study is to clarify the benefit of a combination of S1 and kinase inhibitors including FGFR2 inhibitor Ki23057 in gastric cancer. Experimental Design: OCUM-2MLN and KATO-III were derived from diffuse-type gastric carcinoma. MKN-7 and MKN-74 were derived from intestinal-type gastric carcinoma. MTT assay was used to examine the growth-inhibitory activity of 5 small-synthetic molecules including Ki23057, Sunitinib, Glivec, Lapatinib, or SU11274, in cells cultured with 5-FU. Combination effects of 5-FU with Ki23057 on proliferation, apoptosis, and mRNA expression were examined. S1 and/or Ki23057 were administered to murine models of diffuse-type gastric carcinoma created by the orthotopic inoculation of OCUM-2MLN cells. Results: Ki23057 at 100 nM significantly (p Conclusion: The combined treatment with 5-FU and Ki23057 produced synergistic anti-tumor effects, and is a therapeutically promising for diffuse-type gastric carcinoma treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2552. doi:10.1158/1538-7445.AM2011-2552


Anticancer Research | 2010

Establishment and Characterization of Multidrug-resistant Gastric Cancer Cell Lines

Xiaotian Zhang; Masakazu Yashiro; Hong Qiu; Takafumi Nishii; Taro Matsuzaki; Kosei Hirakawa

Collaboration


Dive into the Taro Matsuzaki's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge