Network


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

Hotspot


Dive into the research topics where Hirofumi Yasui is active.

Publication


Featured researches published by Hirofumi Yasui.


Journal of Clinical Oncology | 2013

Randomized, Open-Label, Phase III Study Comparing Irinotecan With Paclitaxel in Patients With Advanced Gastric Cancer Without Severe Peritoneal Metastasis After Failure of Prior Combination Chemotherapy Using Fluoropyrimidine Plus Platinum: WJOG 4007 Trial

Shuichi Hironaka; Shinya Ueda; Hirofumi Yasui; Tomohiro Nishina; Masahiro Tsuda; Takehiko Tsumura; Naotoshi Sugimoto; Hideki Shimodaira; Shinya Tokunaga; Toshikazu Moriwaki; Taito Esaki; Michitaka Nagase; Kazumasa Fujitani; Kensei Yamaguchi; Takashi Ura; Yasuo Hamamoto; Satoshi Morita; Isamu Okamoto; Narikazu Boku; Ichinosuke Hyodo

PURPOSE This phase III study compared treatment with weekly paclitaxel and biweekly irinotecan in patients with advanced gastric cancer refractory to treatment with fluoropyrimidine plus platinum. PATIENTS AND METHODS Patients were randomly assigned to receive either paclitaxel (80 mg/m(2) on days 1, 8, and 15, every 4 weeks) or irinotecan (150 mg/m(2) on days 1 and 15, every 4 weeks). Primary end point was overall survival (OS), and secondary end points were progression-free survival (PFS), response rate, adverse events, and proportion of patients who received third-line chemotherapy. RESULTS Of 223 patients, 219 were eligible for analysis. Median OS was 9.5 months in 108 patients allocated to the paclitaxel group and 8.4 months in 111 patients allocated to the irinotecan group (hazard ratio [HR], 1.13; 95% CI, 0.86 to 1.49; P = .38). Median PFS was 3.6 months in the paclitaxel group and 2.3 months in the irinotecan group (HR, 1.14; 95% CI, 0.88 to 1.49; P = .33). Response rate was 20.9% in the paclitaxel group and 13.6% in the irinotecan group (P = .24). Common grade 3 to 4 adverse events were neutropenia (paclitaxel group, 28.7%; irinotecan group, 39.1%), anemia (21.3%; 30.0%), and anorexia (7.4%; 17.3%). Treatment-related deaths occurred in two patients (1.8%) in the irinotecan group. Third-line chemotherapy was administered in 97 patients (89.8%) after paclitaxel treatment and in 80 patients (72.1%) after irinotecan treatment (P = .001). CONCLUSION No statistically significant difference was observed between paclitaxel and irinotecan for OS. Both are reasonable second-line treatment options for advanced gastric cancer.


Journal of Gastroenterology | 2003

Matrix metalloproteinase-3 secretion from human colonic subepithelial myofibroblasts: role of interleukin-17

Shigeki Bamba; Akira Andoh; Hirofumi Yasui; Yoshio Araki; Tadao Bamba; Yoshihide Fujiyama

Background. Subepithelial myofibroblasts (SEMFs) play a role in extracellular matrix (ECM) metabolism in the colon. In this study, we investigated the effects of interleukin (IL)-17, IL-1β, and tumor necrosis factor (TNF)-α on matrix metalloproteinase (MMP)-3 secretion in colonic SEMFs. Methods. MMP-3 secretion and MMP-3 mRNA expression were determined by Western and Northern blotting, respectively. The secretion of tissue inhibitor of matrix metalloproteinase (TIMP)-1 was determined by enzyme-linked immunosorbent assay (ELISA). Results. In human colonic SEMFs, MMP-3 secretion and MMP-3 mRNA expression were induced by IL-17, IL-1β, and TNF-α. The effect of IL-17 was observed, but this was weak as compared with those induced by IL-1β or TNF-α. A c-Jun/activating protein-1 (AP-1) inhibitor, curcumin, reduced the IL-17-, IL-1β-, and TNF-α-induced MMP-3 mRNA expression, and mitogen-activated protein (MAP) kinase inhibitors (U0126, PD098059, and SB203580) also blocked MMP-3 secretion. There findings indicate a role for AP-1 and MAP kinases in cytokine-induced MMP-3 secretion. Furthermore, costimulation by IL-17 + IL-1β and by IL-17 + TNF-α induced a marked increase in MMP-3 secretion. The costimulatory effects of these combinations were also observed for TIMP-1 mRNA expression and TIMP-1 secretion. Conclusions. Colonic SEMFs actively secreted MMP-3 in response to IL-17, IL-1β, and TNF-α. This was coupled with TIMP-1 secretion. Colonic SEMFs may play an important role in ECM turnover via MMP secretion.


Journal of Gastroenterology | 2005

Interleukin-17 augments tumor necrosis factor-α-induced granulocyte and granulocyte/macrophage colony-stimulating factor release from human colonic myofibroblasts

Akira Andoh; Hirofumi Yasui; Osamu Inatomi; Zhuobin Zhang; Yasuyuki Deguchi; Kazunori Hata; Yoshio Araki; Tomoyuki Tsujikawa; Katsuyuki Kitoh; Shokei Kim-Mitsuyama; Atsushi Takayanagi; Nobuyoshi Shimizu; Yoshihide Fujiyama

BackgroundInterleukin (IL)-17 is a newly identified T-cell-specific cytokine. In this study, we investigated the effects of IL-17 on colony-stimulating factor (CSF) release in human colonic subepithelial myofibroblasts (SEMFs).MethodsCSF release and mRNA expression were determined by enzyme-linked immunosorbent assay (ELISA) and Northern blotting, respectively. Nuclear factor (NF)-κB- and activating protein (AP-1)-DNA binding activities were evaluated by electrophoretic gel mobility shift assays (EMSAs).ResultsUnstimulated cells secreted a small amount of granulocyte G- and granulocyte/macrophage (GM)-CSF, and a considerable amount of M-CSF. IL-17 weakly enhanced G-CSF release, but did not affect GM- and M-CSF release. IL-17 selectively enhanced tumor necrosis factor (TNF)-α-induced G- and GM-CSF release. The combination of IL-17 plus TNF-α induced a marked increase in NF-κB- and AP-1-DNA binding activities. The adenovirus-mediated transfer of a stable form of IκBα and/or a dominant negative mutant of c-Jun markedly inhibited the IL-17 plus TNF-α-induced G- and GM-CSF mRNA expression. Furthermore, a stability study showed that IL-17 plus TNF-α markedly enhanced the stability of G- and GM-CSF mRNA.ConclusionsIL-17 augments TNF-α-induced G- and GM-CSF release via transcriptional and posttranscriptional mechanisms.


Lancet Oncology | 2017

Nivolumab treatment for oesophageal squamous-cell carcinoma: an open-label, multicentre, phase 2 trial

Toshihiro Kudo; Yasuo Hamamoto; Ken Kato; Takashi Ura; Takashi Kojima; Takahiro Tsushima; Shuichi Hironaka; Hiroki Hara; Taroh Satoh; Satoru Iwasa; Kei Muro; Hirofumi Yasui; Keiko Minashi; Kensei Yamaguchi; Atsushi Ohtsu; Yuichiro Doki; Yuko Kitagawa

BACKGROUND Nivolumab is a human monoclonal IgG4 antibody that inhibits programmed cell death protein 1 (PD-1) expressed on activated T cells. We investigated the safety and activity of nivolumab in patients with treatment-refractory oesophageal cancer. METHODS We did an open-label, single-arm, multicentre phase 2 study. Eligible patients had advanced squamous-cell carcinoma, adenosquamous-cell carcinoma, or adenocarcinoma of the oesophagus refractory or intolerant to fluoropyrimidine-based, platinum-based, and taxane-based chemotherapy. Patients were treated with 3 mg/kg nivolumab given intravenously once every 2 weeks in 6-week cycles. The primary endpoint was centrally assessed objective response (the proportion of patients whose best response was complete or partial response), according to the Response Evaluation Criteria In Solid Tumors, version 1.1. Adverse events and treatment-related adverse events (defined as events for which a causal relation to nivolumab could not be ruled out) were monitored throughout the study. The safety analysis was done in patients who received at least one dose of nivolumab, and drug activity was assessed in patients who received at least one dose of nivolumab and had at least one central assessment of tumour response. This study is registered with clinicaltrials.jp, number ONO-4538-07/JapicCTI-No.142422. Follow-up of patients is ongoing. FINDINGS Between Feb 25 and Nov 14, 2014, 65 patients were enrolled, all with squamous-cell carcinoma. 64 patients were assessable for the primary endpoint as one patient was excluded due to having multiple primary cancers; all patients were assessable for safety. Median follow-up was 10·8 months (IQR 4·9-14·3). 11 (17%, 95% CI 10-28) of 64 patients had a centrally assessed objective response. Of the 65 patients assessed for adverse events, the most common grade 3 or 4 events were grade 4 dyspnoea and hyponatraemia (one [2%) patient each), grade 3 lung infection (five [8%] patients), grade 3 decreased appetite (two [3%] patients), grade 3 increased blood creatinine phosphokinase (two [3%] patients), and grade 3 dehydration (two [3%] patients). Serious adverse events that occurred during the study were lung infection (four [6%] patients), dehydration (two [3%]), interstitial lung disease (two [3%]), and hyponatraemia, dyspnoea, fatigue, abnormal hepatic function, diarrhoea, bile duct stenosis, gastroenteritis, pneumonia, oedema, and back pain (one [2%] patient each). There were no treatment-related deaths. INTERPRETATION Nivolumab showed promising activity with a manageable safety profile. This drug could offer a potential new treatment approach for patients with treatment-refractory advanced squamous-cell carcinoma. FUNDING Ono Pharmaceutical, Bristol-Myers Squibb.


Gastric Cancer | 2009

Weekly paclitaxel for heavily treated advanced or recurrent gastric cancer refractory to fluorouracil, irinotecan, and cisplatin

Rai Shimoyama; Hirofumi Yasui; Narikazu Boku; Yusuke Onozawa; Shuichi Hironaka; Akira Fukutomi; Kentaro Yamazaki; Keisei Taku; Takashi Kojima; Nozomu Machida; Akiko Todaka; Hideharu Tomita; Takeshi Sakamoto; Takahiro Tsushima

BackgroundAlthough triweekly administration of paclitaxel is approved for gastric cancer in Japan, currently, the drug is often delivered with a weekly schedule because of the equivalent efficacy and lesser toxicity of this dosing schedule as compared with the triweekly administration schedule. Weekly administration of paclitaxel as second-line or first-line chemotherapy for gastric cancer has been reported to yield a response rate of about 20%. Because there has been no report of the efficacy of weekly paclitaxel in the third-line setting, this retrospective study investigated the efficacy and toxicities of weekly paclitaxel used in the third-line setting for the treatment of gastric cancer refractory to all three key drugs, fluorouracil, irinotecan, and cisplatin, used in clinical practice.MethodsIn 85 patients with advanced or recurrent histologically confirmed gastric adenocarcinoma who had failed to respond to prior chemotherapy regimens containing fluorouracil, irinotecan, and cisplatin, paclitaxel (80 mg/m2) was administered weekly, three times, for 3 weeks out of 4.ResultsThe median number of courses was 3 (range, 1–38). The overall response rate was 23.2% (19/82) in the patients with measurable lesions, and ascites disappeared in 15 of 48 patients (31.3%). Progression-free survival was 105 days and the median survival time was 201 days from the initiation of paclitaxel administration. Grade 3 or 4 leukopenia, neutropenia, anemia, and thrombocytopenia were observed in 25 (29%), 25 (29%), 37 (44%), and 3 (4%) patients. Other, nonhematological, toxicities were nausea, vomiting, anorexia, sensory neuropathy, fatigue, and febrile neutropenia.ConclusionWeekly paclitaxel administration shows activity against advanced gastric cancer also in the third-line setting.


Cancer Science | 2014

Phase II trial of nanoparticle albumin-bound paclitaxel as second-line chemotherapy for unresectable or recurrent gastric cancer

Yasutsuna Sasaki; Tomohiro Nishina; Hirofumi Yasui; Masahiro Goto; Kei Muro; Akihito Tsuji; Wasaburo Koizumi; Yasushi Toh; Takuo Hara; Yoshinori Miyata

This multicenter phase II study first investigated the efficacy and safety of nanoparticle albumin‐bound paclitaxel (nab‐paclitaxel) when given every 3 weeks to patients with unresectable or recurrent gastric cancer who had received a prior round of fluoropyrimidine‐containing chemotherapy. Patients with unresectable or recurrent gastric cancer who experienced progression despite fluoropyrimidine‐containing treatment were studied. Nab‐paclitaxel was given i.v. at 260 mg/m2 on day 1 of each 21‐day cycle without anti‐allergic premedication until disease progression or study discontinuation. The primary endpoint was the overall response rate. The secondary endpoints were the disease control rate, progression‐free survival, overall survival, and safety. From April 2008 to July 2010, 56 patients were enrolled, 55 patients received the study treatment, and 54 patients were evaluable for responses. According to an independent review committee, the overall response rate was 27.8% (15/54; 95% confidence interval [CI], 16.5–41.6) and the disease control rate was 59.3% (32/54; 95% CI, 45.0–72.4). One patient had a complete response. The median progression‐free survival and overall survival were 2.9 months (95% CI, 2.4–3.6) and 9.2 months (95% CI, 6.9–11.4), respectively. The most common grade 3/4 toxicities were neutropenia (49.1%), leucopenia (20.0%), lymphopenia (10.9%), and peripheral sensory neuropathy (23.6%). There were no treatment‐related deaths. Nab‐paclitaxel, given every 3 weeks, showed promising activity against previously treated unresectable or recurrent gastric cancers, with well‐tolerated toxicities. (Trial registration, ClinicalTrials.gov: NCT00661167).


Annals of Oncology | 2013

The effect of CYP2C19 polymorphism on the safety, tolerability, and pharmacokinetics of tivantinib (ARQ 197): results from a phase I trial in advanced solid tumors

Nobuyuki Yamamoto; Haruyasu Murakami; Tomohiro Nishina; T. Hirashima; Kenji Sugio; Kei Muro; Toshiaki Takahashi; Tateaki Naito; Hirofumi Yasui; Shiro Akinaga; Yasuhiro Koh; Narikazu Boku

BACKGROUND Tivantinib (formerly ARQ 197) is a selective inhibitor of c-Met mainly metabolized by CYP2C19. CYP2C19 is known for genetic polymorphisms, and ~20% of Asians are poor metabolizers (PMs), while others are extensive metabolizers (EMs). In this study, we examined the safety, pharmacokinetics (PK), and preliminary efficacy of tivantinib as a single agent to determine recommended phase II doses (RPIIDs). PATIENTS AND METHODS Forty-seven patients (EMs, 33; PMs, 14) with solid tumors were orally treated with tivantinib, from 70 to 360 mg bid in a 3 + 3 dose-escalation scheme. EMs and PMs were separately enrolled at the doses >120 mg bid. RESULTS Tivantinib was well tolerated up to 360 mg bid for EMs and 240 mg bid for PMs. Neutropenia, leukopenia, anemia, fatigue, and anorexia were the frequent adverse events related to tivantinib and were commonly observed in both EMs and PMs. PMs had 1.9-fold higher AUC(0-12) compared with EMs at 240 mg bid. Regardless of CYP2C19 phenotype, Gr.4 neutropenia occurred in patients with relatively high exposure to tivantinib. A confirmed partial response was achieved in two non-small-cell lung cancer (NSCLC) patients. CONCLUSION Two different settings of RPIIDs, 360 mg bid for EMs and 240 mg bid for PMs, were determined.


Journal of Gastroenterology | 2005

Butyrate blocks interferon-γ-inducible protein-10 release in human intestinal subepithelial myofibroblasts

Osamu Inatomi; Akira Andoh; Ken-ichi Kitamura; Hirofumi Yasui; Zhuobin Zhang; Yoshihide Fujiyama

BackgroundInterferon (IFN)-γ-inducible protein (IP)-10 is a chemoattractant for CXCR 3-expressing T lymphocytes and monocytes. IP-10 has been reported to mediate chronic inflammation such as that in inflammatory bowel disease (IBD). However, the local secretion of IP-10 in the intestine remains unclear. In this study, we investigated IP-10 secretion in human colonic subepithelial myofibroblasts (SEMFs).MethodsIP-10 secretion was determined by enzyme-linked immunosorbent assay (ELISA), and IP-10 mRNA expression was evaluated by Northern blotting.ResultsInterleukin (IL)-10 mRNA was not detected in unstimulated SEMFs. Interferon (IFN)-γ strongly induced IP-10 mRNA expression. Tumor necrosis factor (TNF)-α also stimulated IP-10 mRNA expression, but this was much weaker than that induced by IFN-γ. The effects of IFN-γ and TNF-α were detected in a dose- and time-dependent manner. These responses were also observed at the protein levels. The IFN-γ-induced IP-10 secretion was not affected by acetate or propionate, but was significantly reduced by butyrate. Trichostatin A, a specific inhibitor of histone deacetylase, also blocked the IFN-γ- and TNF-α-induced IP-10 mRNA expression, but the effects of trichostatin A were weaker than those of butyrate. The inhibitory effect of butyrate on IFN-γ-induced IP-10 release was not associated with STAT (signaling transducer and activator of transcription)-1α activation.ConclusionsWe demonstrated that human colonic SEMFs are the local site for the secretion IP-10. The regulation of IP-10 release by IFN-γ and butyrate may play an important role in controlling chronic mucosal inflammation in pathological entities such as IBD.


Digestion | 2004

Role of Fibroblast Growth Factor-2 in the Expression of Matrix Metalloproteinases and Tissue Inhibitors of Metalloproteinases in Human Intestinal Myofibroblasts

Hirofumi Yasui; Akira Andoh; Shigeki Bamba; Osamu Inatomi; Hideaki Ishida; Yoshihide Fujiyama

Background and Aims: The coordinated expression of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) plays a crucial role in tissue remodeling. We investigated the effects of fibroblast growth factor (FGF)-2 on the secretion of MMPs and TIMPs in human intestinal subepithelial myofibroblasts (SEMFs). Methods: The secretion of MMP-s and TIMPs was determined by ELISA or Western blotting. The mRNA expression of MMPs and TIMPs was assessed by Northern blotting. The activating protein (AP)-1-DNA binding activity was evaluated by electrophoretic gel mobility shift assays (EMSA). Results: Unstimulated intestinal SEMFs constitutively secreted MMP-2 and TIMP-2. FGF-2 stimulated MMP-1, MMP-3 and TIMP-1 secretion, but did not affect MMP-2 or TIMP-2 secretion. FGF-2 induced AP-1-DNA binding activity, and the c-Jun/AP-1 inhibitor curcumin attenuated the FGF-2-induced MMP-1, -3 and TIMP-1 mRNA expression. Mitogen-activated protein (MAP) kinase inhibitors (U0126 and PD098059) also blocked the MMP-1, -3 and TIMP-1 secretion. Furthermore, FGF-2 dose-dependently induced FGF-2 mRNA expression in these cells. Conclusions: FGF-2 may be one of important regulatory factors for extracellular matrix turnover via a modulation of MMP and TIMP secretion from SEMFs.


Japanese Journal of Clinical Oncology | 2010

S-1 Monotherapy as Second-line Treatment for Advanced Pancreatic Cancer after Gemcitabine Failure

Akiko Todaka; Akira Fukutomi; Narikazu Boku; Yusuke Onozawa; Shuichi Hironaka; Hirofumi Yasui; Kentaro Yamazaki; Keisei Taku; Nozomu Machida; Takeshi Sakamoto; Hideharu Tomita

OBJECTIVE No standard salvage chemotherapy regimen has been established for patients with advanced pancreatic cancer after failure of gemcitabine-based treatment. Although a Phase II study of S-1 monotherapy was conducted in patients with gemcitabine-refractory advanced pancreatic cancer, the number of patients enrolled was small. METHODS We retrospectively reviewed 84 consecutive patients who received S-1 monotherapy as a second-line treatment after gemcitabine failure at the Shizuoka Cancer Center between May 2004 and April 2008. The selection criteria in this study were age 20-75 years, ECOG performance status <or=2 and preserved organ functions. S-1 was administered orally twice a day at a dose of 40 mg/m(2) for 28 days, followed by 14-day rest. RESULTS Fifty-two patients were selected for the analysis. Out of the 47/52 patients with measurable lesions, only 2 patients (4%) showed a partial response and 15 patients (32%) showed stable disease. The median progression-free survival was 2.1 months and the median overall survival was 5.8 months, with a 1-year survival rate of 12%. The common grade 3/4 toxicities were diarrhea (8%), anorexia (6%), fatigue (6%), anemia (6%) and leucopenia (4%). CONCLUSIONS S-1 monotherapy is marginally effective and well tolerated in the second-line setting in patients with gemcitabine-refractory advanced pancreatic cancer.

Collaboration


Dive into the Hirofumi Yasui's collaboration.

Top Co-Authors

Avatar

Kentaro Yamazaki

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Narikazu Boku

St. Marianna University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kei Muro

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Tomoya Yokota

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kensei Yamaguchi

Japanese Foundation for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Hiroya Taniguchi

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge