Network


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

Hotspot


Dive into the research topics where Hiroyuki Furukawa is active.

Publication


Featured researches published by Hiroyuki Furukawa.


Journal of Pharmacy and Pharmacology | 2004

Disposition kinetics of taxanes after intraperitoneal administration in rats and influence of surfactant vehicles.

Koichi Yokogawa; Mingji Jin; Naho Furui; Masaru Yamazaki; Hiromi Yoshihara; Masaaki Nomura; Junko Ishizaki; Ken-ichi Miyamoto; Hiroyuki Furukawa; Sachio Fushida; Kouichi Miwa

Rats were intraperitoneally administered 40 mg kg−1 of paclitaxel or docetaxel dissolved in various drug solutions. The drug solutions were prepared using 20 mL of saline, adding 4.2% Cremophor EL (crEL) for paclitaxel (TXL), and 1.5% Polysorbate‐80 (PS‐80) (TXT), 7.5% PS‐80 (TXT+PS‐80) or 4.2% crEL (TXT+crEL) for docetaxel. The apparent first‐order absorption rate constant from the peritoneal cavity (ka) of TXL was about one‐twentieth of that of TXT. The ratio of the area under the concentration‐time curve of drug in plasma over that in ascites for TXL was about one‐third of that of TXT. The values of the above ratio and the ka of TXT+PS‐80 and TXT+crEL were similar to those of TXL. After intraperitoneal administration, the values of the blood‐to‐plasma concentration ratio in the four groups were similar and independent of time. In the in‐vitro study, PS‐80 and crEL caused similar, concentration‐dependent decreases of drug permeation into red blood cells after a 15‐min incubation of rat blood with 10 μg mL−1 of TXL. We demonstrated that the disposition kinetics of taxanes after intraperitoneal administration to rats was strongly influenced, in a concentration‐dependent manner, by the surfactant vehicle used, crEL or PS‐80.


Annals of Pharmacotherapy | 2003

Voluntary Medication Error Reporting Program in a Japanese National University Hospital

Hiroyuki Furukawa; Hisashi Bunko; Fumito Tsuchiya; Ken-ichi Miyamoto

BACKGROUND: In Japan, as in other countries, medical accidents arising from human error can seriously damage public confidence in medical services, as well as being intrinsically undesirable. OBJECTIVE: Errors voluntarily reported by the healthcare practitioners in our institution (Kanazawa University Hospital) were considered to assess the contributory factors by using the accumulated error database in the hospital information system. METHODS: Medical errors in our institution during the period from July 1, 2000, to June 30, 2002, were counted using the error reporting system database and were classified. RESULTS: The number of errors reported during the investigation period was 1378, of which 78% were reported by nursing staff. Medication errors involving administration of injectable or oral drugs to inpatients, dispensing, and prescription accounted for about 50% of that number. Among dispensing errors, 53% were detected by patients or their families and 36% by nurses. CONCLUSIONS: The best method of error prevention is to learn from previous errors. For this purpose, the error reporting program is effective. In patient safety management, it is important to take into account the potential risks of future errors, as well as to capture information about errors that have already happened. For safety management, adoption of appropriate information technology (e.g., implementation of a prescription order entry system) is effective in reducing medication errors. However, it is important to note that serious errors can also arise in computer-based systems.


Cancer Science | 2017

Phase II clinical trial of peptide cocktail therapy for patients with advanced pancreatic cancer: VENUS-PC study

Nobuaki Suzuki; Shoichi Hazama; Haruo Iguchi; Kazuhiro Uesugi; Hiroaki Tanaka; Kosei Hirakawa; Atsushi Aruga; Takashi Hatori; Hidenobu Ishizaki; Yuzo Umeda; Toshiyoshi Fujiwara; Tetsuya Ikemoto; Mitsuo Shimada; Kazuhiko Yoshimatsu; Ryoichi Shimizu; Hiroto Hayashi; Koichiro Sakata; Hiroko Takenouchi; Hiroto Matsui; Yoshitaro Shindo; Michihisa Iida; Yasunobu Koki; Hideki Arima; Hiroyuki Furukawa; Tomio Ueno; Shigefumi Yoshino; Yusuke Nakamura; Masaaki Oka; Hiroaki Nagano

We previously conducted a phase I clinical trial combining the HLA‐A*2402‐restricted KIF20A‐derived peptide vaccine with gemcitabine for advanced pancreatic cancer (PC) and confirmed its safety and immunogenicity in cancer patients. In this study, we conducted a multicenter, single‐armed, phase II trial using two antiangiogenic cancer vaccines targeting VEGFR1 and VEGFR2 in addition to the KIF20A peptide. We attempted to evaluate the clinical benefit of the cancer vaccination in combination with gemcitabine. Chemotherapy naïve PC patients were enrolled to evaluate primarily the 1‐year survival rate, and secondarily overall survival (OS), progression free survival (PFS), response rate (RR), disease control rate (DCR) and the peptide‐specific immune responses. All enrolled patients received therapy without the HLA‐A information, and the HLA genotypes were used for classification of the patients. Between June 2012 and May 2013, a total of 68 patients were enrolled. No severe systemic adverse effects of Grade 3 or higher related to these three peptides were observed. The 1‐year survival rates between the HLA‐A*2402‐matched and ‐unmatched groups were not significantly different. In the HLA‐A*2402 matched group, patients showing peptide‐specific CTL induction for KIF20A or VEGFR1 showed a better prognosis compared to those without such induction (P = 0.023, P = 0.009, respectively). In the HLA‐A*2402‐matched group, the patients who showed a strong injection site reaction had a better survival rate (P = 0.017) compared to those with a weak or no injection site reaction. This phase II study demonstrated that this therapeutic peptide cocktail might be effective in patients who demonstrate peptide‐specific immune reactions although predictive biomarkers are needed for patient selection in its further clinical application.


Oncology Reports | 2012

Increased E-selectin in hepatic ischemia-reperfusion injury mediates liver metastasis of pancreatic cancer

Katsuhiro Yoshimoto; Hidehiro Tajima; Tetsuo Ohta; Koichi Okamoto; Seisho Sakai; Jun Kinoshita; Hiroyuki Furukawa; Isamu Makino; Hironori Hayashi; Keishi Nakamura; Katsunobu Oyama; Masafumi Inokuchi; Hisatoshi Nakagawara; Hiroshi Itoh; Hideto Fujita; Hiroyuki Takamura; Itasu Ninomiya; Hirohisa Kitagawa; Sachio Fushida; Takashi Fujimura; Tomohiko Wakayama; Shoichi Iseki; Koichi Shimizu

Several recent studies have reported that selectins are produced during ischemia-reperfusion injury, and that selectin ligands play an important role in cell binding to the endothelium and in liver metastasis. Portal clamping during pancreaticoduodenectomy with vessel resection for pancreatic head cancer causes hepatic ischemia-reperfusion injury, which might promote liver metastasis. We investigated the liver colonization of pancreatic cancer cells under hepatic ischemia-reperfusion and examined the involvement of E-selectin and its ligands. A human pancreatic cancer cell line (Capan-1) was injected into the spleen of mice after hepatic ischemia-reperfusion (I/R group). In addition, to investigate the effect of an anti-E-selectin antibody on liver colonization in the IR group, mice received an intraperitoneal injection of the anti-E-selectin antibody following hepatic ischemia-reperfusion and tumor inoculation (IR+Ab group). Four weeks later, mice were sacrificed and the number of tumor nodules on the liver was compared to mice without hepatic ischemia-reperfusion (control group). The incidence of liver metastasis in the I/R group was significantly higher (16 of 20, 80%) than that in the control group (6 of 20, 30%) (P<0.01). Moreover, mice in the I/R group had significantly more tumor nodules compared to those in the control group (median, 9.9 vs. 2.7 nodules) (P<0.01). In the I/R+Ab group, only 2 of 5 (40%) mice developed liver metastases. RT-PCR and southern blotting of the liver extracts showed that the expression of IL-1 and E-selectin mRNA after hepatic ischemia-reperfusion was significantly higher than the basal levels. Hepatic ischemia-reperfusion increases liver metastases and E-selectin expression in pancreatic cancer. These results suggest that E-selectin produced due to hepatic ischemia-reperfusion is involved in liver metastasis.


Colloids and Surfaces B: Biointerfaces | 2016

pH-sensitive polymeric cisplatin-ion complex with styrene-maleic acid copolymer exhibits tumor-selective drug delivery and antitumor activity as a result of the enhanced permeability and retention effect.

Atsuyuki Saisyo; Hideaki Nakamura; Jun Fang; Kenji Tsukigawa; Khaled Greish; Hiroyuki Furukawa; Hiroshi Maeda

Cisplatin (CDDP) is widely used to treat various cancers. However, its distribution to normal tissues causes serious adverse effects. For this study, we synthesized a complex of styrene-maleic acid copolymer (SMA) and CDDP (SMA-CDDP), which formed polymeric micelles, to achieve tumor-selective drug delivery based on the enhanced permeability and retention (EPR) effect. SMA-CDDP is obtained by regulating the pH of the reaction solution of SMA and CDDP. The mean SMA-CDDP particle size was 102.5 nm in PBS according to electrophoretic light scattering, and the CDDP content was 20.1% (w/w). The release rate of free CDDP derivatives from the SMA-CDDP complex at physiological pH was quite slow (0.75%/day), whereas it was much faster at pH 5.5 (4.4%/day). SMA-CDDP thus had weaker in vitro toxicity at pH 7.4 but higher cytotoxicity at pH 5.5. In vivo pharmacokinetic studies showed a 5-fold higher tumor concentration of SMA-CDDP than of free CDDP. SMA-CDDP had more effective antitumor potential but lower toxicity than did free CDDP in mice after i.v. administration. Administration of parental free CDDP at 4 mg/kg×3 caused a weight loss of more than 5%; SMA-CDDP at 60 mg/kg (CDDP equivalent)×3 caused no significant weight change but markedly suppressed S-180 tumor growth. These findings together suggested using micelles of the SMA-CDDP complex as a cancer chemotherapeutic agent because of beneficial properties-tumor-selective accumulation and relatively rapid drug release at the acidic pH of the tumor-which resulted in superior antitumor effects and fewer side effects compared with free CDDP.


Oncology Letters | 2013

Low‑dose paclitaxel inhibits the induction of epidermal‑mesenchymal transition in the human cholangiocarcinoma CCKS‑1 cell line

Atsushi Hirose; Hidehiro Tajima; Tetsuo Ohta; Tomoya Tsukada; Koichi Okamoto; Shinichi Nakanuma; Seisho Sakai; Jun Kinoshita; Isamu Makino; Hiroyuki Furukawa; Hironori Hayashi; Keishi Nakamura; Katsunobu Oyama; Masafumi Inokuchi; Hisatoshi Nakagawara; Tomoharu Miyashita; Hiroyuki Takamura; Itasu Ninomiya; Hirohisa Kitagawa; Sachio Fushida; Takashi Fujimura; Shinichi Harada

Epidermal-mesenchymal transition (EMT) confers an advantage to cancer cells by improving their invasive capacity and metastatic potential. This phenomenon by which epidermal cells change into mesenchymal cells and therefore acquire a higher ability to automaticity, is considered a key process in cancer development. Transforming growth factor-β (TGF-β) is a significant factor for accelerating EMT through the activation of proteins, including members of the Smad pathway. Furthermore, previous studies have shown that low-dose paclitaxel (PTX) inhibits EMT in certain cell lines, including those of cancer cells. The present study determined whether low-dose PTX was able to inhibit EMT in a human cholangiocarcinoma CCKS-1 cell line that had been treated with TGF-β1. First, the cytotoxic concentration of PTX for the CCKS-1 cells was identified to be ~5 nM by MTT assay and dead cell staining. Therefore, the concentrations of PTX were set as 1 nM, 2.5 nM and 5 nM for the subsequent experiments. In the morphological investigation, the CCKS-1 cells changed into a spindle morphology and became separated by the administration of TGF-β1. However, low-dose PTX inhibited these changes and the morphology resembled the control cells in a dose-dependent manner. Similarly, immunofluorescence and immunoblotting investigations revealed that the CCKS-1 cells expressed mesenchymal markers following the administration of TGF-β1. However, low-dose PTX inhibited the expression of the mesenchymal markers and the CCKS-1 cells expressed the epithelial marker, E-cadherin. In particular, a concentration-dependent effect was observed in the immunoblotting experiments. These results show that PTX may be able to inhibit EMT in cancer cells, depending on the dose concentration.


Academic Radiology | 2011

Triple-negative Breast Cancer: Are the Imaging Findings Different Between Responders and Nonresponders to Neoadjuvant Chemotherapy?

Hiroko Kawashima; Masafumi Inokuchi; Hiroyuki Furukawa; Seiko Kitamura

RATIONALE AND OBJECTIVES The purpose of the present study was to evaluate the imaging findings of triple-negative breast cancer patients receiving neoadjuvant chemotherapy (NAC) and to investigate whether the findings are different between responders and nonresponders, enabling us to predict the final patient response. MATERIALS AND METHODS The subjects included 22 women ages 35-73 years (mean, 50.4 years) with 23 triple-negative breast cancers who underwent NAC. In all cases, a mammography, ultrasound, and magnetic resonance imaging (MRI) were performed a total of three times: before NAC, after the first half of NAC, and after NAC. The mass shape, mass margin, presence of clear intratumoral necrosis, and presence of intratumoral calcification were analyzed. The presence of clear intratumoral necrosis was evaluated on the MRI. If there was a very high signal intensity (similar to that of water) in the tumor on the fat-suppressed T2-weighted MRI scans, we judged it to be clear intratumoral necrosis. RESULTS An irregularly shaped mass (P = .018) and the presence of clear intratumoral necrosis (P = .044) were significantly associated with NAC nonresponse in triple-negative breast cancer patients. The mass margin and the presence of intratumoral calcification were not related to the effects after NAC. CONCLUSIONS In cases of triple-negative breast cancer involving clear intratumoral necrosis with an irregular mass shape, it is predicted that the effects of neoadjuvant chemotherapy will likely be poor, and therefore, the presence of such image findings may be useful for determining the optimal application of neoadjuvant chemotherapy.


Oncology Reports | 2014

Comparative study of the antitumor activity of Nab-paclitaxel and intraperitoneal solvent-based paclitaxel regarding peritoneal metastasis in gastric cancer.

Jun Kinoshita; Sachio Fushida; Tomoya Tsukada; Katsunobu Oyama; Toshihumi Watanabe; Masatoshi Shoji; Koichi Okamoto; Shinichi Nakanuma; Seisho Sakai; Isamu Makino; Hiroyuki Furukawa; Hironori Hayashi; Keishi Nakamura; Masahumi Inokuchi; Hisatoshi Nakagawara; Tomoharu Miyashita; Hidehiro Tajima; Hiroyuki Takamura; Itasu Ninomiya; Takashi Fujimura; Yashiro Masakazu; Kosei Hirakawa; Tetsuo Ohta

Intraperitoneal (i.p.) chemotherapy with paclitaxel (PTX) has been shown to be a promising treatment strategy for peritoneal metastasis. The present study focused on the comparative evaluation of the therapeutic efficacy of nanoparticle albumin-bound PTX (Nab-PTX) and i.p. administration of the conventional solvent-based PTX (Sb-PTX). We also investigated the difference in antitumor activity depending on the route of administration in the Nab-PTX treatment. Nab-PTX was administered i.p. or intravenously (i.v.) and Sb-PTX was administered i.p. at equitoxic and equal doses to nude mice bearing gastric cancer OCUM-2MD3 cell subcutaneous and peritoneal xenografts. Therapeutic efficacy of Sb-PTX and Nab-PTX was evaluated as inhibition of tumor growth using a peritoneal metastatic model with subcutaneous xenografts. The survival rate was also investigated using mouse peritoneal models. For assessment of subcutaneous tumors, the change in tumor volume was measured, and for assessment of peritoneal tumors, the weight of ascitic fluid and the total peritoneal tumor burden were measured for each individual mouse. At equitoxic doses, treatment with Nab-PTX resulted in a greater reduction in the size of subcutaneous tumors and the weight of ascites and peritoneal burden as compared with i.p. Sb-PTX (p<0.05). Treatment with i.p. and i.v. Nab-PTX also achieved greater survival benefit than i.p. Sb-PTX (p<0.05). In contrast, there was no significant difference in the degree of tumor reduction and the survival time between both drugs at equal doses. With regard to the route of administration, the antitumor efficacy of Nab-PTX after i.v. administration was equivalent to the efficacy after i.p. administration. These results suggest that i.v. Nab-PTX may be another encouraging treatment option that can target peritoneal dissemination in gastric cancer.


Molecular and Clinical Oncology | 2013

A phase I study of neoadjuvant chemotherapy with gemcitabine plus oral S-1 for resectable pancreatic cancer

Hidehiro Tajima; Hirohisa Kitagawa; Tomoya Tsukada; Shinich Nakanuma; Koichi Okamoto; Seisho Sakai; Isamu Makino; Hiroyuki Furukawa; Keishi Nakamura; Hironori Hayashi; Katsunobu Oyama; Masafumi Inokuchi; Hisatoshi Nakagawara; Tomoharu Miyashita; Hideto Fujita; Hiroshi Itoh; Hiroyuki Takamura; Itasu Ninomiya; Sachio Fushida; Takashi Fujimura; Tetsuo Ohta

The aim of this study was to determine the maximum-tolerated dose (MTD), the dose-limiting toxicity (DLT) and the recommended dose (RD) of neoadjuvant chemotherapy (NAC) with gemcitabine (GEM) plus oral S-1 in patients with resectable pancreatic cancer. Thirteen patients with radiologically proven resectable pancreatic cancer were included in this study. S-1 was administered orally for 14 consecutive days, and GEM was administered on days 8 and 15 for two pre-operative cycles. The dose of S-1 in this study was planned with fixed doses of GEM (1,000 mg/m2): 20, 30 and 40 mg/day for levels 0, 1 and 2, respectively. Treatment was initiated at level 1 in 3 patients, while adverse events occurred in 2 patients during the second course, leading to a dose reduction to level 0 for the 8 remaining patients. Two of the 10 patients enrolled at level 0 were excluded. Of the remaining 8 patients, GEM administration was terminated due to DLT on day 15, during the first course in 3 patients, while level 0 dosage reached MTD. Surgery was performed for the remaining 11 patients included in the study. Post-operative complications included pancreatic fistulas in 5 patients and Pseudomonas aeruginosa sepsis in 1 patient. Two of the 11 patients exhibited a partial response and 9 patients stable disease. Eight of the 11 tumor specimens showed histopathological evidence of tumor cell injury. In conclusion, NAC with GEM and S-1 was not well-tolerated in this study. However, pre-operative chemotherapy may be effective against pancreatic cancer. Therefore, it is necessary to reconsider NAC regimens for pancreatic cancer.


Journal of Experimental & Clinical Cancer Research | 2017

Predictive biomarkers for the efficacy of peptide vaccine treatment: based on the results of a phase II study on advanced pancreatic cancer

Yoshitaro Shindo; Shoichi Hazama; Nobuaki Suzuki; Haruo Iguchi; Kazuhiro Uesugi; Hiroaki Tanaka; Atsushi Aruga; Takashi Hatori; Hidenobu Ishizaki; Yuzo Umeda; Toshiyoshi Fujiwara; Tetsuya Ikemoto; Mitsuo Shimada; Kazuhiko Yoshimatsu; Hiroko Takenouchi; Hiroto Matsui; Shinsuke Kanekiyo; Michihisa Iida; Yasunobu Koki; Hideki Arima; Hiroyuki Furukawa; Tomio Ueno; Shigefumi Yoshino; Tomonobu Fujita; Yutaka Kawakami; Yusuke Nakamura; Masaaki Oka; Hiroaki Nagano

BackgroundThe purpose of the present study was to explore novel biomarkers that can predict the clinical outcome of patients before treatment or during vaccination. These would be useful for the selection of appropriate patients who would be expected to exhibit better treatment outcomes from vaccination, and for facilitating the development of cancer vaccine treatments.MethodsFrom a single-arm, non-randomized, human leukocyte antigen (HLA)-A-status-blind phase II trial of a vaccine treatment using three HLA-A*2402-restricted peptides for advanced pancreatic cancer (PC), we obtained peripheral blood samples from 36 patients of an HLA-A*2402-matched group and 27 patients of an HLA-A*2402-unmatched group.ResultsMultivariate analysis (HR = 2.546; 95% CI = 1.138 to 5.765; p = 0.0231) and log-rank test (p = 0.0036) showed that a high expression level of programmed death-1 (PD-1) on CD4+ T cells was a negative predictive biomarker of overall survival in the HLA-A*2402-matched group . Moreover, a high expression level of PD-1 on CD4+ T cells was a negative predictor for the induction of cytotoxic T lymphocytes (p = 0.0007). After treatment, we found that the upregulation of PD-1 and T cell immunoglobulin mucin-3 (Tim-3) expression on CD4+ and CD8+ T cells was significantly associated with a poor clinical outcome in the HLA-A*2402-matched group (p = 0.0330, 0.0282, 0.0046, and 0.0068, respectively). In contrast, there was no significant difference for these factors in the HLA-A*2402-unmatched group.ConclusionsOur results indicate that the upregulation of PD-1 and Tim-3 expression on CD4+ and CD8+ T cells may restrict T cell responses in advanced PC patients; therefore, combination immunotherapy with blockade of PD-1 and Tim-3 to restore T cell responses may be a potential therapeutic approach for advanced PC patients.Trial registrationClinical-Trail-Registration: UMIN000008082.

Collaboration


Dive into the Hiroyuki Furukawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fumito Tsuchiya

International University of Health and Welfare

View shared research outputs
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