Network


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

Hotspot


Dive into the research topics where Toshiyuki Tsukaguchi is active.

Publication


Featured researches published by Toshiyuki Tsukaguchi.


Cancer Cell | 2011

CH5424802, a Selective ALK Inhibitor Capable of Blocking the Resistant Gatekeeper Mutant

Hiroshi Sakamoto; Toshiyuki Tsukaguchi; Sayuri Hiroshima; Tatsushi Kodama; Takamitsu Kobayashi; Takaaki A. Fukami; Nobuhiro Oikawa; Takuo Tsukuda; Nobuya Ishii; Yuko Aoki

Anaplastic lymphoma kinase (ALK) is a tyrosine kinase that is constitutively activated in certain cancers, following gene alterations such as chromosomal translocation, amplification, or point mutation. Here, we identified CH5424802, a potent, selective, and orally available ALK inhibitor with a unique chemical scaffold, showing preferential antitumor activity against cancers with gene alterations of ALK, such as nonsmall cell lung cancer (NSCLC) cells expressing EML4-ALK fusion and anaplastic large-cell lymphoma (ALCL) cells expressing NPM-ALK fusion in vitro and in vivo. CH5424802 inhibited ALK L1196M, which corresponds to the gatekeeper mutation conferring common resistance to kinase inhibitors, and blocked EML4-ALK L1196M-driven cell growth. Our results support the potential for clinical evaluation of CH5424802 for the treatment of patients with ALK-driven tumors.


Cancer Letters | 2014

Selective ALK inhibitor alectinib with potent antitumor activity in models of crizotinib resistance

Tatsushi Kodama; Toshiyuki Tsukaguchi; Miyuki Yoshida; Osamu Kondoh; Hiroshi Sakamoto

The clinical efficacy of the ALK inhibitor crizotinib has been demonstrated in ALK fusion-positive NSCLC; however, resistance to crizotinib certainly occurs through ALK secondary mutations in clinical use. Here we examined the efficacy of a selective ALK inhibitor alectinib/CH5424802 in models of crizotinib resistance. Alectinib led to tumor size reduction in EML4-ALK-positive xenograft tumors that failed to regress fully during the treatment with crizotinib. In addition, alectinib inhibited the growth of some EML4-ALK mutant-driven tumors, including the G1269A model. These results demonstrated that alectinib might provide therapeutic opportunities for crizotinib-treated patients with ALK secondary mutations.


Bioorganic & Medicinal Chemistry | 2012

Design and synthesis of a highly selective, orally active and potent anaplastic lymphoma kinase inhibitor (CH5424802).

Kazutomo Kinoshita; Kohsuke Asoh; Noriyuki Furuichi; Toshiya Ito; Hatsuo Kawada; Sousuke Hara; Jun Ohwada; Takuho Miyagi; Takamitsu Kobayashi; Kenji Takanashi; Toshiyuki Tsukaguchi; Hiroshi Sakamoto; Takuo Tsukuda; Nobuhiro Oikawa

Anaplastic lymphoma kinase (ALK) receptor tyrosine kinase is considered an attractive therapeutic target for human cancers, especially non-small cell lung cancer (NSCLC). Our previous study revealed that 8,9-side-chains of 6,6-dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazole scaffold crucially affected kinase selectivity, cellular activity, and metabolic stability. In this work, we optimized the side-chains and identified highly selective, orally active and potent ALK inhibitor CH5424802 (18a) as the clinical candidate.


Molecular Cancer Therapeutics | 2014

Alectinib Shows Potent Antitumor Activity against RET-Rearranged Non–Small Cell Lung Cancer

Tatsushi Kodama; Toshiyuki Tsukaguchi; Yasuko Satoh; Miyuki Yoshida; Yoshiaki Watanabe; Osamu Kondoh; Hiroshi Sakamoto

Alectinib/CH5424802 is a known inhibitor of anaplastic lymphoma kinase (ALK) and is being evaluated in clinical trials for the treatment of ALK fusion–positive non–small cell lung cancer (NSCLC). Recently, some RET and ROS1 fusion genes have been implicated as driver oncogenes in NSCLC and have become molecular targets for antitumor agents. This study aims to explore additional target indications of alectinib by testing its ability to inhibit the activity of kinases other than ALK. We newly verified that alectinib inhibited RET kinase activity and the growth of RET fusion–positive cells by suppressing RET phosphorylation. In contrast, alectinib hardly inhibited ROS1 kinase activity unlike other ALK/ROS1 inhibitors such as crizotinib and LDK378. It also showed antitumor activity in mouse models of tumors driven by the RET fusion. In addition, alectinib showed kinase inhibitory activity against RET gatekeeper mutations (RET V804L and V804M) and blocked cell growth driven by the KIF5B-RET V804L and V804M. Our results suggest that alectinib is effective against RET fusion–positive tumors. Thus, alectinib might be a therapeutic option for patients with RET fusion–positive NSCLC. Mol Cancer Ther; 13(12); 2910–8. ©2014 AACR.


Journal of Medicinal Chemistry | 2011

9-Substituted 6,6-Dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazoles as Highly Selective and Potent Anaplastic Lymphoma Kinase Inhibitors

Kazutomo Kinoshita; Takamitsu Kobayashi; Kohsuke Asoh; Noriyuki Furuichi; Toshiya Ito; Hatsuo Kawada; Sousuke Hara; Jun Ohwada; Kazuo Hattori; Takuho Miyagi; Woo-Sang Hong; Min-Jeong Park; Kenji Takanashi; Toshiyuki Tsukaguchi; Hiroshi Sakamoto; Takuo Tsukuda; Nobuhiro Oikawa

9-Substituted 6,6-dimethyl-11-oxo-6,11-dihydro-5H-benzo[b]carbazoles were discovered as highly selective and potent anaplastic lymphoma kinase (ALK) inhibitors by structure-based drug design. The high target selectivity was achieved by introducing a substituent close to the E(0) region of the ATP binding site, which has a unique amino acid sequence. Among the identified inhibitors, compound 13d showed highly selective and potent inhibitory activity against ALK with an IC(50) value of 2.9 nM and strong antiproliferative activity against KARPAS-299 with an IC(50) value of 12.8 nM. The compound also displayed significant antitumor efficacy in an established ALK fusion gene-positive anaplastic large-cell lymphoma (ALCL) xenograft model in mice without body weight loss.


Molecular Cancer Therapeutics | 2014

The Fibroblast Growth Factor Receptor Genetic Status as a Potential Predictor of the Sensitivity to CH5183284/Debio 1347, a Novel Selective FGFR Inhibitor

Yoshito Nakanishi; Nukinori Akiyama; Toshiyuki Tsukaguchi; Toshihiko Fujii; Kiyoaki Sakata; Hitoshi Sase; Takehito Isobe; Kenji Morikami; Hidetoshi Shindoh; Toshiyuki Mio; Hirosato Ebiike; Naoki Taka; Yuko Aoki; Nobuya Ishii

The FGF receptors (FGFR) are tyrosine kinases that are constitutively activated in a subset of tumors by genetic alterations such as gene amplifications, point mutations, or chromosomal translocations/rearrangements. Recently, small-molecule inhibitors that can inhibit the FGFR family as well as the VEGF receptor (VEGFR) or platelet-derived growth factor receptor (PDGFR) family displayed clinical benefits in cohorts of patients with FGFR genetic alterations. However, to achieve more potent and prolonged activity in such populations, a selective FGFR inhibitor is still needed. Here, we report the identification of CH5183284/Debio 1347, a selective and orally available FGFR1, FGFR2, and FGFR3 inhibitor that has a unique chemical scaffold. By interacting with unique residues in the ATP-binding site of FGFR1, FGFR2, or FGFR3, CH5183284/Debio 1347 selectively inhibits FGFR1, FGFR2, and FGFR3 but does not inhibit kinase insert domain receptor (KDR) or other kinases. Consistent with its high selectivity for FGFR enzymes, CH5183284/Debio 1347 displayed preferential antitumor activity against cancer cells with various FGFR genetic alterations in a panel of 327 cancer cell lines and in xenograft models. Because of its unique binding mode, CH5183284/Debio 1347 can inhibit FGFR2 harboring one type of the gatekeeper mutation that causes resistance to other FGFR inhibitors and block FGFR2 V564F–driven tumor growth. CH5183284/Debio 1347 is under clinical investigation for the treatment of patients harboring FGFR genetic alterations. Mol Cancer Ther; 13(11); 2547–58. ©2014 AACR.


Bioorganic & Medicinal Chemistry Letters | 2009

Synthesis and structure-activity relationships of novel benzofuran farnesyltransferase inhibitors.

Kohsuke Asoh; Masami Kohchi; Ikumi Hyoudoh; Tatsuo Ohtsuka; Miyako Masubuchi; Kenichi Kawasaki; Hirosato Ebiike; Yasuhiko Shiratori; Takaaki A. Fukami; Osamu Kondoh; Toshiyuki Tsukaguchi; Nobuya Ishii; Yuko Aoki; Nobuo Shimma; Masahiro Sakaitani

A series of benzofuran-based farnesyltransferase inhibitors have been designed and synthesized as antitumor agents. Among them, 11f showed the most potent enzyme inhibitory activity (IC(50)=1.1nM) and antitumor activity in human cancer xenografts in mice.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of novel tetracyclic compounds as anaplastic lymphoma kinase inhibitors.

Kazutomo Kinoshita; Yoshiyuki Ono; Takashi Emura; Kohsuke Asoh; Noriyuki Furuichi; Toshiya Ito; Hatsuo Kawada; Shota Tanaka; Kenji Morikami; Toshiyuki Tsukaguchi; Hiroshi Sakamoto; Takuo Tsukuda; Nobuhiro Oikawa

Anaplastic lymphoma kinase (ALK) receptor tyrosine kinase is considered a promising therapeutic target for human cancers. We identified novel tetracyclic derivatives as potent ALK inhibitors. Among them, compound 27 showed strong cytotoxicity against KARPAS-299 with an IC(50) value of 21 nM and significant antitumor efficacy in ALK fusion-positive blood and solid cancer xenograft models in mice without body weight loss.


Molecular Cancer Therapeutics | 2015

Mechanism of Oncogenic Signal Activation by the Novel Fusion Kinase FGFR3-BAIAP2L1

Yoshito Nakanishi; Nukinori Akiyama; Toshiyuki Tsukaguchi; Toshihiko Fujii; Yasuko Satoh; Nobuya Ishii; Masahiro Aoki

Recent cancer genome profiling studies have identified many novel genetic alterations, including rearrangements of genes encoding FGFR family members. However, most fusion genes are not functionally characterized, and their potentials in targeted therapy are unclear. We investigated a recently discovered gene fusion between FGFR3 and BAI1-associated protein 2-like 1 (BAIAP2L1). We identified 4 patients with bladder cancer and 2 patients with lung cancer harboring the FGFR3–BAIAP2L1 fusion through PCR and FISH assay screens. To investigate the oncogenic potential of the fusion gene, we established an FGFR3–BAIAP2L1 transfectant with Rat-2 fibroblast cells (Rat-2_F3-B). The FGFR3–BAIAP2L1 fusion had transforming activity in Rat2 cells, and Rat-2_F3-B cells were highly tumorigenic in mice. Rat-2_F3-B cells showed in vitro and in vivo sensitivity in the selective FGFR inhibitor CH5183284/Debio 1347, indicating that FGFR3 kinase activity is critical for tumorigenesis. Gene signature analysis revealed that FGFR3–BAIAP2L1 activates growth signals, such as the MAPK pathway, and inhibits tumor-suppressive signals, such as the p53, RB1, and CDKN2A pathways. We also established Rat-2_F3-B-ΔBAR cells expressing an FGFR3–BAIAP2L1 variant lacking the Bin–Amphiphysin–Rvs (BAR) dimerization domain of BAIAP2L1, which exhibited decreased tumorigenic activity, FGFR3 phosphorylation, and F3-B-ΔBAR dimerization, compared with Rat-2_F3-B cells. Collectively, these data suggest that constitutive dimerization through the BAR domain promotes constitutive FGFR3 kinase activation and is essential for its potent oncogenic activity. Mol Cancer Ther; 14(3); 704–12. ©2015 AACR.


Cancer Science | 2013

Enhanced antitumor activity of erlotinib in combination with the Hsp90 inhibitor CH5164840 against non-small-cell lung cancer

Naomi Ono; Toshikazu Yamazaki; Toshiyuki Tsukaguchi; Toshihiko Fujii; Kiyoaki Sakata; Atsushi Suda; Takuo Tsukuda; Toshiyuki Mio; Nobuya Ishii; Osamu Kondoh; Yuko Aoki

Inhibition of heat shock protein 90 (Hsp90) can lead to degradation of multiple client proteins, which are involved in tumor progression. Epidermal growth factor receptor (EGFR) is one of the most potent oncogenic client proteins of Hsp90. Targeted inhibition of EGFR has shown clinical efficacy in the treatment of patients with non‐small‐cell lung cancer (NSCLC). However, primary and acquired resistance to the existing EGFR inhibitors is a major clinical problem. In the present study, we investigated the effect of the novel Hsp90 inhibitor CH5164840 on the antitumor activity of erlotinib. The NSCLC cell lines and xenograft models were treated with CH5164840 and erlotinib to examine their mechanisms of action and cell growth inhibition. We found that CH5164840 showed remarkable antitumor activity against NSCLC cell lines and xenograft models. The addition of CH5164840 enhanced the antitumor activity of erlotinib against NCI‐H292 EGFR‐overexpressing xenograft models. Phosphorylation of Stat3 increased with erlotinib treatment in NCI‐H292 cells, which was abrogated by Hsp90 inhibition. Furthermore, in a NCI‐H1975 T790M mutation erlotinib‐resistant model, CH5164840 enhanced the antitumor activity of erlotinib despite the low efficacy of erlotinib treatment alone. In addition, ERK signaling was effectively suppressed by combination treatment with erlotinib and CH5164840 in a NCI‐H1975 erlotinib‐resistant model. Taken together, these data indicate that CH5164840 has potent antitumor activity and is highly effective in combination with erlotinib against NSCLC tumors with EGFR overexpression and mutations. Our results support the therapeutic potential of CH5164840 as a Hsp90 inhibitor for combination therapy with EGFR‐targeting agents against EGFR‐addicted NSCLC.

Collaboration


Dive into the Toshiyuki Tsukaguchi's collaboration.

Top Co-Authors

Avatar

Nobuya Ishii

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tatsushi Kodama

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Yuko Aoki

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Osamu Kondoh

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Takuo Tsukuda

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Yasuko Satoh

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Toshihiko Fujii

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nobuhiro Oikawa

Chugai Pharmaceutical Co.

View shared research outputs
Researchain Logo
Decentralizing Knowledge