Network


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

Hotspot


Dive into the research topics where Makoto Ogo is active.

Publication


Featured researches published by Makoto Ogo.


PLOS ONE | 2015

PE859, a Novel Tau Aggregation Inhibitor, Reduces Aggregated Tau and Prevents Onset and Progression of Neural Dysfunction In Vivo

Michiaki Okuda; Ichiro Hijikuro; Yuki Fujita; Xiaofeng Wu; Shinichi Nakayama; Yoko Sakata; Yuji Noguchi; Makoto Ogo; Shigeru Akasofu; Yoshimasa Ito; Yoshiyuki Soeda; Nobuhiko Tsuchiya; Naoki Tanaka; Takashi Takahashi; Hachiro Sugimoto

In tauopathies, a neural microtubule-associated protein tau (MAPT) is abnormally aggregated and forms neurofibrillary tangle. Therefore, inhibition of the tau aggregation is one of the key approaches for the treatment of these diseases. Here, we have identified a novel tau aggregation inhibitor, PE859. An oral administration of PE859 resulted in the significant reduction of sarkosyl-insoluble aggregated tau along with the prevention of onset and progression of the motor dysfunction in JNPL3 P301L-mutated human tau transgenic mice. These results suggest that PE859 is useful for the treatment of tauopathies.


Alzheimers & Dementia | 2015

A single dose of the beta-secretase inhibitor, e2609, decreases CSF bace1 enzymatic activity in cynomolgus monkeys

Mark Matijevic; Hideki Watanabe; Yoshiaki Sato; Francois Bernier; Shannon McGrath; Lynne Burns; Noboru Yamamoto; Makoto Ogo; Zoltan Dezso; Jesse Chow; Yoshiya Oda; June Kaplow; Bruce Albala

Figure 2. Correlation Between CSF BACE1 Activity Levels and CSF Ab 1-x levels in All Monkey Samples Mark Matijevic, Hideki Watanabe, Yoshiaki Sato, Francois Bernier, Shannon McGrath, Lynne Burns, Noboru Yamamoto, Makoto Ogo, Zoltan Dezso, Jesse Chow, Yoshiya Oda, June Kaplow, Bruce Albala, Eisai, Andover, MA, USA; Eisai, Tsukuba, Japan; Formerly Eisai (when work was completed), Worcester, MA, USA; Eisai, Woodcliff Lake, NJ, USA; Eisai,Woodcliffe Lake, NJ, USA. Contact e-mail: [email protected]


Cancer Research | 2016

Abstract 3264: Lenvatinib in combination with everolimus demonstrated enhanced antiangiogenesis and antitumor activity in human RCC xenograft models

Yusuke Adachi; Masahiro Matsuki; Atsumi Yamaguchi; Yoichi Ozawa; Kiyochi Okamoto; Kaoru Mitsuhashi; Takayuki Kimura; Taisuke Hoshi; Osamu Tohyama; Kenji Tai; Makoto Ogo; Yasuhiro Funahashi; Junji Matsui

Lenvatinib mesilate (lenvatinib) is an oral multiple receptor tyrosine kinase (RTK) inhibitor that selectively inhibits the kinase activities of vascular endothelial growth factor (VEGF) receptors VEGFR1 (FLT1), VEGFR2 (KDR), and VEGFR3 (FLT4), in addition to other proangiogenic and oncogenic pathway-related RTKs including fibroblast growth factor (FGF) receptors FGFR1, 2, 3, and 4; the platelet-derived growth factor (PDGF) receptor PDGFRα; KIT; and RET. Lenvatinib showed antitumor activity against various tumor types mainly through its potent inhibition of angiogenesis, and is currently marketed for the treatment of patients with radioactive iodine-refractory differentiated thyroid cancer. Recently, lenvatinib in combination with everolimus has shown longer progression free survival compared to lenvatinib or everolimus alone in renal cell carcinoma in Phase 2 study. The aim of this study is to elucidate the activity of the combination of lenvatinib and everolimus in preclinical human RCC xenograft models. We examined antitumor activity in two human RCC (A-498 and Caki-1) xenograft models orally treated with lenvatinib (10 mg/kg), everolimus (30 mg/kg), and the combination of lenvatinib and everolimus for 1 or 2 weeks. The antitumor proliferation and antiangiogenic effects were evaluated by immunohistochemistry (IHC) using anti Ki67 antibody and anti CD31 antibody, respectively. The induction of apoptosis was detected by TUNEL assay. To analyze the gene expression profile of tumor samples, microarray analysis were also conducted. The antitumor activity of the combination of lenvatinib and everolimus was greater than that of either agent administered alone in A-498 and Caki-1 xenograft models. The combination caused tumor regression and had no remarkable body weight loss. IHC analysis revealed decrease of microvessel density in lenvatinib and combination groups, and also decrease in the proportion of proliferative cells in everolimus treated and combination-treated group in A-498 model. In TUNEL assay, significant induction of apoptosis was observed only in the combination-treatment group. The analysis of gene expression profile in A-498 xenograft tumors also supported these results: lenvatinib alone upregulated hypoxia-related genes and everolimus decreased proliferation-related genes. The combination of these 2 drugs induced blends of the gene expression changes caused by each single treatment. Our results indicate that treatment of lenvatinib in combination with everolimus caused significant antitumor effect by combining the potent antiangiogenic activity of lenvatinib as well as direct antitumor activity of everolimus in A-498 model. These preclinical results provide one of the mechanisms to support the significant clinical benefit observed in RCC with the combination of lenvatinib and everolimus. Citation Format: Yusuke Adachi, Masahiro Matsuki, Atsumi Yamaguchi, Yoichi Ozawa, Kiyochi Okamoto, Kaoru Mitsuhashi, Takayuki Kimura, Taisuke Hoshi, Osamu Tohyama, Kenji Tai, Makoto Ogo, Yasuhiro Funahashi, Junji Matsui. Lenvatinib in combination with everolimus demonstrated enhanced antiangiogenesis and antitumor activity in human RCC xenograft models. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3264.


Cancer Research | 2016

Abstract 3262: Effects of lenvatinib mesilate in combination with everolimus on VEGF and FGF-driven angiogenesis and tumor growth

Kaoru Mitsuhashi; Takayuki Kimura; Taisuke Hoshi; Osamu Tohyama; Kenji Tai; Makoto Ogo; Masahiro Matsuki; Atsumi Yamaguchi; Yoichi Ozawa; Yusuke Adachi; Kiyoshi Okamoto; Junji Matsui; Yasuhiro Funahashi

Lenvatinib mesilate (lenvatinib) is an orally available inhibitor for multiple receptor tyrosine kinases (RTKs) that selectively inhibits the kinase activities of VEGFR1, 2, and 3, in addition to other proangiogenic and oncogenic pathway-related RTKs including FGFR1, 2, 3, and 4; PDGFRα; KIT; and RET. Recently, lenvatinib in combination with everolimus has shown longer progression free survival compared to lenvatinib or everolimus alone in renal cell carcinoma in a Phase 2 study. In this study, we evaluated the effect of lenvatinib in combination with everolimus on VEGF and bFGF-driven angiogenesis to elucidate the mechanism of combination action in preclinical models. Preclinical studies provide a plausible biologic rationale for the significant clinical benefit observed in RCC with the combination of lenvatinib and everolimus. Effects of lenvatinib, everolimus, and its combination on VEGF or bFGF activated intracellular signaling were analyzed in HUVEC by western blotting. Combination effects of lenvatinib and everolimus on VEGF and bFGF-induced proliferation or tube formation of HUVEC were examined using combination indexes (CI). Antitumor activities were tested in the KP-1/VEGF or KP-1/FGF models, where VEGF or FGF-induced tumor angiogenesis and tumor growth were enhanced in nude mice due to overexpressed VEGF or FGF in human pancreatic cancer KP-1 cells. Lenvatinib inhibited the VEGF or bFGF-driven phosphorylation of Erk1/2 (Thr202/Tyr204), S6K (Thr389), and S6K (Thr421/Ser424), and S6 (Ser235/Ser236), indicating the inhibition of both the MAPK pathway and the mTOR-S6K-S6 pathway. Everolimus inhibited the phosphorylation of S6K (Thr389), S6K (Thr421/Ser424), and S6 (Ser235/Ser236), but not Erk1/2. The combination showed greater inhibition for the phosphorylation of S6K (Thr421/Ser424) and S6 (Ser235/Ser236) than each single agent. Inhibitory activity of the combination at several molar ratios was mostly additive for VEGF-driven proliferation (CI: 0.799-1.167) and mostly synergistic for bFGF-driven tube formation (CI: 0.469-0.741). In the KP-1/VEGF or KP-1/FGF xenogtaft models, lenvatinib, everolimus, and the combination (p.o., qd x 14) significantly inhibited tumor growth compared to vehicle. In addition the combination of lenvatinib (7.5 mg/kg) and everolimus (15 mg/kg) showed significantly greater antitumor activity than higher dose of either lenvatinib (10 mg/kg) or everolimus (30 mg/kg) monotherapy. These results demonstrated enhancement of the inhibitory activity against VEGF and FGF-induced angiogenesis by the combination of lenvatinib with everolimus, and the synergistic enhancement against bFGF-induced angiogenesis unlike other VEGFR2 TKIs. The vertical inhibition of angiogenic signaling pathways with lenvatinib (RTK) and everolimus (mTOR) may contribute to enhanced antiangiogenic activity by dual targeting of the mTOR-S6K-S6 pathway. Citation Format: Kaoru Mitsuhashi, Takayuki Kimura, Taisuke Hoshi, Osamu Tohyama, Kenji Tai, Makoto Ogo, Masahiro Matsuki, Atsumi Yamaguchi, Yoichi Ozawa, Yusuke Adachi, Kiyoshi Okamoto, Junji Matsui, Yasuhiro Funahashi. Effects of lenvatinib mesilate in combination with everolimus on VEGF and FGF-driven angiogenesis and tumor growth. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3262.


Archive | 2001

1,2-dihydropyridine compounds, process for preparation of the same and use thereof

Satoshi Nagato; Kohshi Ueno; Koki Kawano; Yoshihiko Norimine; Koichi Ito; Takahisa Hanada; Masataka Ueno; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Yoshio Urawa; Hiroyuki Naka; Anthony Groom; Leanne Rivers; Terence Smith


Archive | 2009

1, 2-Dihydropyridine compounds, manufacturing method thereof and use thereof

Satoshi Nagato; Kohshi Ueno; Koki Kawano; Yoshihiko Norimine; Koichi Ito; Takahisa Hanada; Masataka Ueno; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Yoshio Urawa; Hiroyuki Naka; Anthony John Groom; Leanne Rivers; Terence Smith


Archive | 2001

Pyridazinones and triazinones and medicinal use thereof

Satoshi Nagato; Koki Kawano; Koichi Ito; Yoshihiko Norimine; Kohshi Ueno; Takahisa Hanada; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Masataka Ueno; Anthony John Groom; Leanne Rivers; Terence Smith


Archive | 2006

Pyridazinone and triazinone compounds and use thereof as pharmaceutical preparations

Satoshi Nagato; Koki Kawano; Koichi Ito; Yoshihiko Norimine; Kohshi Ueno; Takahisa Hanada; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Masataka Ueno; Anthony John Groom; Leanne Rivers; Terence Smith


Archive | 2009

TRIAZINONES AND MEDICINAL USE THEREOF

Satoshi Nagato; Koki Kawano; Koichi Ito; Yoshihiko Norimine; Kohshi Ueno; Takahisa Hanada; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Masataka Ueno; Anthony Groom; Leanne Rivers; Terence Smith


Archive | 2001

1,3,5-Trisubstituted 2(1H)-pyridones as AMPA receptor inhibitors useful for the treatment of eg Parkinson's disease

Satoshi Nagato; Kohshi Ueno; Koki Kawano; Yoshihiko Norimine; Koichi Ito; Takahisa Hanada; Masataka Ueno; Hiroyuki Amino; Makoto Ogo; Shinji Hatakeyama; Yoshio Urawa; Hiroyuki Naka; Anthony John Groom; Leanne Rivers; Terence Smith

Collaboration


Dive into the Makoto Ogo's collaboration.

Researchain Logo
Decentralizing Knowledge