Network


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

Hotspot


Dive into the research topics where Masaaki Motoyama is active.

Publication


Featured researches published by Masaaki Motoyama.


Annals of Oncology | 2015

Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies

Andrea Li Ann Wong; Ross A. Soo; D. S. Tan; Soo-Chin Lee; J. S. Lim; P. C. Marban; L. R. Kong; Y. J. Lee; Lingzhi Wang; Win-Lwin Thuya; Richie Chuan Teck Soong; M. Q. Yee; T. M. Chin; M. T. Cordero; B. R. Asuncion; Brendan Pang; Shazib Pervaiz; Jayshree L. Hirpara; Arvind Kumar Sinha; W. W. Xu; M. Yuasa; Takeshi Tsunoda; Masaaki Motoyama; T. Yamauchi; Boon Cher Goh

BACKGROUND The aim of this study was to determine the maximum-tolerated dose (MTD), safety, pharmacokinetics, and pharmacodynamics of OPB-51602, an oral, direct signal transduction activator of transcription 3 (STAT3) inhibitor, in patients with refractory solid tumors. PATIENTS AND METHODS Three cohorts were studied: cohort A, a sequential dose escalation of OPB-51602 administered intermittently (days 1-14 every 21 days); cohort B, an expansion cohort evaluating the dose lower than the MTD; cohort C, evaluating continuous daily dosing. RESULTS Fifty-one patients were studied at 2, 4, and 5 mg per day dosing. The MTD was 5 mg; first-cycle dose-limiting toxicities (DLTs) were grade 3 hyponatremia in one patient, and grade 3 dehydration in another. Intermittent dosing of both 2 and 4 mg doses were tolerable, and the recommended phase II dose was 4 mg. Cohort B investigated 4 mg intermittently, whereas cohort C investigated 4 mg continuously. Common toxicities included fatigue, nausea/vomiting, diarrhea, anorexia, and early-onset peripheral neuropathy. Drug-induced pneumonitis occurred in two patients in cohort C. Continuous dosing was associated with a higher incidence of peripheral neuropathy and a lower mean relative dose intensity, compared with intermittent dosing. Steady-state pharmacokinetics was characterized by high oral clearance, mean elimination half-life ranging from 44 to 61 h, and a large terminal-phase volume of distribution. An active metabolite, OPB-51822, accumulated to a greater extent than OPB-51602. Flow cytometry of peripheral blood mononuclear cells demonstrated pSTAT3 (Tyr(705)) inhibition following exposure. Two patients achieved partial responses at 5 mg intermittently and 4 mg continuously; both had epidermal growth factor receptor (EGFR) mutation-positive non-small-cell lung cancer (NSCLC) with prior EGFR tyrosine kinase inhibitor exposure. CONCLUSION OPB-51602 demonstrates promising antitumor activity, particularly in NSCLC. Its long half-life and poorer tolerability of continuous dosing, compared with intermittent dosing, suggest that less frequent dosing should be explored. CLINICALTRIALSGOV IDENTIFIER NCT01184807.


Japanese Journal of Cancer Research | 1989

Effects of the Plasma Concentration of 5‐Fluorouracil and the Duration of Continuous Venous Infusion of 5‐Fluorouracil with an Inhibitor of 5‐Fluorouracil Degradation on Yoshida Sarcomas in Rats

Setsuro Fujii; Yuji Shimamoto; Hideyuki Ohshimo; Takeshi Imaoka; Masaaki Motoyama; Masakazu Fukushima; Tetsuhiko Shirasaka

The correlations of the 5‐fluorouracil (5‐FU) level in the plasma and the duration of continuous 5‐FU infusion with the antitumor activity of 5‐FU on Yoshida sarcomas in rats were examined. The circadian variation in the plasma level of 5‐FU during continuous infusion was prevented by treatment with 3‐cyano‐2,6‐dihydroxypyridine (CNDP), which strongly inhibits 5‐FU degradation. On continuous venous infusion of 2 to 30 mg/kg of 5‐FU over 24 h with CNDP at a molar ratio of 1:10 into normal rats, the 5‐FU level in the blood was linearly proportional to the dose of 5‐FU. The optimum schedule for antitumor activity on Yoshida sarcomas in rats was found to be infusion of 5‐FU at 5 mg/kg over 24 h for 6 consecutive days, which gave a plasma 5‐FU level of 176 ng/ml. Continuous infusion of 5‐FU to give a plasma level of 300 ng/ml for 6 consecutive days from day 5 after implantation of tumor cells, when the tumors weighed about 1.0 g, resulted in complete regression of the tumors in all rats.


Cancer Science | 2015

Phase I study of OPB-51602, an oral inhibitor of signal transducer and activator of transcription 3, in patients with relapsed/refractory hematological malignancies.

Michinori Ogura; Toshiki Uchida; Yasuhito Terui; Fumihiko Hayakawa; Yukio Kobayashi; Masafumi Taniwaki; Yasushi Takamatsu; Tomoki Naoe; Kensei Tobinai; Wataru Munakata; Takeshi Yamauchi; Akiko Kageyama; Miyuki Yuasa; Masaaki Motoyama; Takeshi Tsunoda; Kiyohiko Hatake

We carried out a multicenter dose‐escalation phase I study of oral OPB‐51602, a signal transducer and activator of transcription 3 phosphorylation inhibitor, in patients with relapsed or refractory hematological malignancies to evaluate the safety, maximum tolerated dose (MTD), pharmacokinetics, and preliminary antitumor activity. Twenty patients were treated with OPB‐51602 at doses of 1, 2, 3, 4, and 6 mg in the “3 + 3” dose escalation design. The most common treatment‐related adverse events included nausea (55%), peripheral sensory neuropathy (45%), and diarrhea (40%). The most frequently observed grade 3 or 4 drug‐related adverse events were neutropenia (20%), leukopenia (15%), lymphopenia (10%), and thrombocytopenia (10%). The MTD was 6 mg, with dose‐limiting toxicities of grade 3 lactic acidosis and increased blood lactic acid levels observed in one of three patients and grade 1–2 peripheral neuropathy in three of three patients. The recommended dose was determined to be 4 mg. OPB‐51602 was rapidly absorbed, and exposure tended to increase in a dose‐dependent manner. Accumulation of OPB‐51602 was seen with 4 weeks of multiple treatments. No clear therapeutic response was observed. Durable stable disease was observed in two patients with acute myeloid leukemia and one with myeloma. In conclusion, the MTD of OPB‐51602 was 6 mg. OPB‐51602 was safe and well tolerated in a dose range of 1–4 mg. However, long‐term administration at higher doses was difficult with the daily dosing schedule, and no response was seen. Therefore, further clinical development of OPB‐51602 for hematological malignancies with a daily dosing schedule was terminated.


Japanese Journal of Cancer Research | 1999

Inhibition of Spontaneous Rat Osteosarcoma Lung Metastasis by 3S‐[4‐(Nhydroxyamino)‐2R‐isobutylsuccinyl]amino‐1‐methoxy‐3,4‐dihydrocarbostyril, a Novel Matrix Metalloproteinase Inhibitor

Akira Kido; Masahiro Tsutsumi; Katsumichi Iki; Masaaki Motoyama; Makoto Takahama; Toshifumi Tsujiuchi; Toru Morishita; Kunihiko Tatsumi; Susumu Tamai; Yoichi Konishi

In the present experiment, we examined the effects of OPB‐3206, 3S‐[4‐(N‐hydroxyamino)‐2R isobutylsuccinyl] amino‐1‐methoxy‐3,4‐dihydrocarbostyril, a novel metalloproteinase inhibitor, on the growth and metastasis of transplantable osteosarcomas (spontaneous osteosarcoma, selected lung metastatic lesions; S‐SLM), which were previously established in rats. OPB‐3206 inhibited the activities of interstitial collagenase, gelatinases A and B, and stromelysin in vitro. After oral administration to rats, its serum concentration peaked at 40 min and the drug was no longer detectable at 8 h. When OPB‐3206 was orally administered at 0%, 0.1% and 0.4% in the diet for 4 weeks, starting 7 days after subcutaneous transplantation of osteosarcomas to male Fischer 344 rats, numbers of lung metastatic nodules were significantly reduced by the highest dose, while the growth of subcutaneous tumors was not affected. Zymographic analysis showed the presence of pro matrix metalloproteinase (proMMP)‐2, proMMP‐9 and MMP‐9 activities in S‐SLM. In animals fed 0.4% OPB‐3206, the activity of proMMP‐9 was increased, but that for MMP‐9 had become undetectable. The results thus suggest that OPB‐3206 selectively inhibits lung metastasis of rat transplantable osteosarcomas by inhibiting MMP‐9 activation.


Angiogenesis | 1998

A new synthetic matrix metalloproteinase inhibitor modulates both angiogenesis and urokinase type plasminogen activator activity

Tadahisa Shono; Masaaki Motoyama; Kunihiko Tatsumi; Norbert Ulbrich; Yukihide Iwamoto; Michihiko Kuwano; Mayumi Ono

Proteolytic degradation of the extracellular matrix is essential to angiogenesis. Two families of proteases, the serine proteases of plasminogen activator/plasmin system and the matrix metalloproteinases (MMPs) are closely involved in these processes. The treatment of mice with a diet containing a new synthetic MMP inhibitor, OPB-3206: 3S-[4-(N-hydroxyamino)-2R-isobutylsuccinyl] amino-1methoxy-3, 4-dihydrocarbostyril, abrogated the development of new vessels in a rat corneal assay, and in a mouse Matrigel assay. In an in vitro angiogenesis model, OPB-3206 inhibited the migration and the tube formation of bovine aortic endothelial cells at 10–100 times lower concentrations than those required to inhibit the growth of these cells. OPB-3206 as well as other MMP inhibitory drugs, batimastat/BB-94 and marimastat/BB-2516, also selectively inhibited tubular morphogenesis in vitro. OPB-3206 reduced the activities of interstitial collagenase and type IV collagenase, but the concentrations of 50% inhibition against these MMPs were much higher than those of BB-94 and BB-2516. However, this new compound also inhibited urokinase type plasminogen activator activity on fibrin zymogram, while BB-94 and BB-2516 did not. Furthermore, the addition of urokinase type plasminogen activator reduced the inhibitory effect of the tubular morphogenesis of vascular endothelial cells by OPB-3206. The treatment of mice with a diet containing this new compound also reduced the growth of implanted mammary carcinomas as well as the lung metastasis of colon carcinoma. The anti-angiogenic effect of OPB-3206 appeared to be associated with its inhibition of tumor growth and metastasis.


Molecular Cancer Therapeutics | 2011

Abstract B56: A phase 1, open-label, dose escalation, nonrandomized study to assess the maximum tolerated dose, dose limiting toxicity, and pharmacokinetics of OPB-31121 in subjects with advanced solid tumors.

Razelle Kurzrock; Aung Naing; Gerald S. Falchook; David S. Hong; Agnes Elekes; Takumi Sumida; Patricia Bricmont; Takeshi Kodama; Masaaki Motoyama; Shiva Patil; Jennifer A. Lee; Suzanne F. Jones; Jeffrey R. Infante; Howard A. Burris; Johanna C. Bendell

Background: OPB-31121 is a novel compound exhibiting potent growth inhibition of cancer cell lines in vitro and xenografts in vivo. The exact mechanism of action of OPB-31121 has not been fully characterized, but studies indicate that a major effect is inhibition of STAT3 phosphorylation. STAT3 is frequently activated in a variety of solid and hematologic malignancies and may present an important target for antitumor therapy. Methods: Open-label, non-randomized, multi-center study in subjects with advanced solid tumors, using a 3+3 dose escalation design. OPB-31121 was administered orally for 21 days followed by 7 days rest per cycle (28-day cycle). The starting dose was 50 mg BID with escalations planned until the dose-limiting toxicity (DLT) was reached. The primary endpoint was determination of maximum tolerated dose (MTD). Additional endpoints included safety, pharmacokinetics, and anti-tumor effect of OPB-31121. Results: 30 subjects received treatment with OPB-31121. Most common tumor types were colorectal cancer (15), breast (3) and thyroid (2). Mean age was 55.9 (range 35–80) years and 17 of the patients were female. Most common adverse events (AEs) potentially attributed to treatment were gastrointestinal: nausea (80%), vomiting (73%), diarrhea (63%), anorexia (20%), and constipation (17%). Most AEs were CTCAE grades 1–2 and manageable with supportive treatment. Three DLTs were observed: one at 300 mg BID (grade 3 lactic acidosis), and two at 350 mg BID (a grade 3 diarrhea and a grade 3 vomiting); the MTD was 300 mg BID. All patients recovered from DLTs after discontinuing the drug. Six additional (9 total) subjects discontinued during the first cycle. Eight subjects completed only one cycle and 13 completed two cycles. No objective responses were observed. Disease progression was observed in all evaluable patients at first restaging. Pharmacokinetic measurements showed low and transient plasma levels of OPB-31121. Inter-patient variability was high. Exposure was low - area-under-the-curve (AUC) values were 2–3 orders of magnitude lower than those measured at active doses in mouse models. Analysis of metabolites in plasma samples indicates extensive CYP3A4 metabolism and suggests a large first-pass effect in humans, which had not been observed in rodents. Conclusions: OPB-31121 does not show potential as a therapeutic option for most malignancies. The extensive first-pass metabolism and dose-limiting gastrointestinal AEs limit the level of systemic exposure that can be achieved with this agent. Further studies may be warranted in cancers of organs where local concentrations of the compound may be higher (e.g., liver) and an exploratory study in hepatocellular carcinoma is ongoing in Asia. Because STAT3 remains an attractive antitumor target, chemically related compounds with similar pharmacologic activities are currently being evaluated preclinically. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr B56.


Archive | 1993

Maillard reaction inhibitor

Keisuke Miyajima; Bonpei Yasui; Masaaki Motoyama; Shintaro Ishikawa; Koichi Yasumura


Archive | 1995

Carbostyril derivatives as matrix metalloproteinases inhibitors

Makoto Sakamoto; Takeshi Imaoka; Masaaki Motoyama; Yoshihito Yamamoto; Hideki Takasu


Carcinogenesis | 1999

Expression of matrix metalloproteinase 2 (MMP-2), membrane-type 1 MMP and tissue inhibitor of metalloproteinase 2 and activation of proMMP-2 in pancreatic duct adenocarcinomas in hamsters treated with N-nitrosobis(2-oxopropyl)amine

Katsumichi Iki; Masahiro Tsutsumi; Akira Kido; Hiroyuki Sakitani; Makoto Takahama; Masatoshi Yoshimoto; Masaaki Motoyama; Kunihiko Tatsumi; Tsukasa Tsunoda; Yoichi Konishi


Archive | 2006

DIARYLETHER DERIVATIVES AS ANTITUMOR AGENTS

Hironori Matsuyama; Kenji Ohnishi; Takashi Nakagawa; Hideki Takasu; Makoto Sakamoto; Kumi Higuchi; Keisuke Miyajima; Satoshi Yamada; Masaaki Motoyama; Yutaka Kojima; Koichi Yasumura; Takeshi Kodama; Shun Otsuji; Keizo Kan; Takumi Sumida

Collaboration


Dive into the Masaaki Motoyama'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

Naoto Ohi

Otsuka Pharmaceutical

View shared research outputs
Top Co-Authors

Avatar

Takashi Nakagawa

Tokyo Metropolitan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge