Network


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

Hotspot


Dive into the research topics where Hiroko Yamakawa is active.

Publication


Featured researches published by Hiroko Yamakawa.


Bioorganic & Medicinal Chemistry | 2012

Discovery of the investigational drug TAK-441, a pyrrolo[3,2-c]pyridine derivative, as a highly potent and orally active hedgehog signaling inhibitor: modification of the core skeleton for improved solubility.

Tomohiro Ohashi; Yuya Oguro; Toshio Tanaka; Zenyu Shiokawa; Yuta Tanaka; Sachio Shibata; Yoshihiko Sato; Hiroko Yamakawa; Harumi Hattori; Yukiko Yamamoto; Shigeru Kondo; Maki Miyamoto; Mitsuhiro Nishihara; Yoshimasa Ishimura; Hideaki Tojo; Atsuo Baba; Satoshi Sasaki

We recently reported the discovery of the novel pyrrolo[3,2-c]quinoline-4-one derivative 1 as a potent inhibitor of Hedgehog (Hh) pathway signaling. However, the PK evaluation of 1 at high dosage (100 mg/kg) revealed the C(max) value 3.63 μg/mL, likely due to poor solubility of this compound. Efforts to improve solubility by reducing the aromatic ring count of the core system led to N-methylpyrrolo[3,2-c]pyridine derivative 11. Further optimization of the 3-alkoxy group led to compound 11d with acceptable solubility and potent Hh inhibitory activity. Compound 11d suppressed transcription factor Gli1 mRNA expression in tumor-associated stromal tissue and inhibited tumor growth (treatment/control ratio, 3%) in a mouse medulloblastoma allograft model owing to the improved PK profile based on increased solubility. Compound 11d (TAK-441) is currently in clinical trials for the treatment of advanced solid tumors.


Bioorganic & Medicinal Chemistry | 2012

Discovery of pyrrolo[3,2-c]quinoline-4-one derivatives as novel hedgehog signaling inhibitors.

Tomohiro Ohashi; Yuya Oguro; Toshio Tanaka; Zenyu Shiokawa; Sachio Shibata; Yoshihiko Sato; Hiroko Yamakawa; Harumi Hattori; Yukiko Yamamoto; Shigeru Kondo; Maki Miyamoto; Hideaki Tojo; Atsuo Baba; Satoshi Sasaki

The Hedgehog (Hh) signaling pathway plays a significant role in the regulation of cell growth and differentiation during embryonic development. Since activation of the Hh signaling pathway is implicated in several types of human cancers, inhibitors of this pathway could be promising anticancer agents. Using high throughput screening, thieno[3,2-c]quinoline-4-one derivative 9a was identified as a compound of interest with potent in vitro activity but poor metabolic stability. Our efforts focused on enhancement of in vitro inhibitory activity and metabolic stability, including core ring conversion and side chain optimization. This led to the discovery of pyrrolo[3,2-c]quinoline-4-one derivative 12b, which has a structure distinct from previously reported Hh signaling inhibitors. Compound 12b suppressed stromal Gli1 mRNA expression in a murine model and demonstrated antitumor activity in a murine medulloblastoma allograft model.


Cancer Research | 2011

Abstract 2823: TAK-441, a novel investigational small molecule hedgehog pathway inhibitor for use in cancer therapy

Hideaki Tojo; Sachio Shibata; Yoshihiko Satoh; Mihoko Kawamura; Masakazu Inazuka; Hiroko Yamakawa; Masahide Kashiwagi; Maki Miyamoto; Shigeru Kondo; Tomohiro Oohashi; Yuya Oguro; Satoshi Sasaki

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Dysregulation of hedgehog (Hh) signal, which is caused by Hh-ligand overexpression or patched1 (Ptch1) mutation, has been found to play an important role in tumorigenesis. TAK-441 is a potent and selective smoothened (Smo) antagonist that blocks Hh signaling. In the Gli-responsive promoter-luciferase (Gli-luc) reporter assay in NIH3T3/Gli-luc cells, TAK-441 inhibited Gli transcriptional activity with an IC50 of 4.4 nM. TAK-441 also inhibited expression of human Gli1 messenger mRNA with an IC50 of 1.9 nM in MRC5 human embryonic fibroblasts. In addition, TAK-441 inhibited binding of cyclopamine, a plant-derived well-known Smo inhibitor, to human Smo on 293T cells overexpressing human Smo with an IC50 of 8.6 nM. These data suggest that TAK-441 inhibits the Hh signaling pathway through its binding to Smo. Oral administration of TAK-441 (0.5, 1, 5, 25 mg/kg QD) in mice bearing a Ptch1 (+/-), p53(−/-) medulloblastoma allograft resulted in dose-dependent antitumor activity with concomitant reduction of Gli1 mRNA expression in the tumors. Complete remissions were observed in the 25 mg/kg QD dose cohort after two consecutive weeks of treatment. Significant (p≤0.025 by one-tailed Williams test) antitumor activity was also detected in sonic hedgehog expressing human primary pancreatic and ovarian cancer xenografts when dosed at 10 and 25 mg/kg QD for two weeks, and in a colon cancer xenograft when dosed at 6.25 and 25 mg/kg BID for three weeks. In these models, the expression of tumor-associated mouse stromal Gli1 mRNA was markedly decreased following TAK-441 treatment, suggesting that Hh signaling in these tumors was driven in a paracrine mode of action. The combination of TAK-441 and rapamycin, an inhibitor of mammalian target of rapamycin (mTOR), demonstrated significantly better antitumor activity than either agent alone in human primary pancreatic cancer xenograft models (p≤0.05 by Students’ t-test). In addition, delayed tumor re-growth was observed in the TAK-441/rapamycin combination treatment group compared with the rapamycin single-agent treatment group after terminating treatment. As a result of these preclinical studies, TAK-441 has recently entered Phase I clinical studies in cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2823. doi:10.1158/1538-7445.AM2011-2823


Bioorganic & Medicinal Chemistry | 2015

Synthesis and evaluation of hedgehog signaling inhibitor with novel core system.

Tomohiro Ohashi; Yuta Tanaka; Zenyu Shiokawa; Hiroshi Banno; Toshio Tanaka; Sachio Shibata; Yoshihiko Satoh; Hiroko Yamakawa; Yukiko Yamamoto; Harumi Hattori; Shigeru Kondo; Maki Miyamoto; Hideaki Tojo; Atsuo Baba; Satoshi Sasaki

As we previously reported, N-methylpyrrolo[3,2-c]pyridine derivatives 1 (TAK-441) was discovered as a clinical candidate of hedgehog (Hh) signaling inhibitor by modification of the upper part. We next focused on modification of the lower part including core skeletons to discover new Hh signaling inhibitors with novel core rings. Efforts to find novel chemotypes by using X-ray single crystal structure analysis led to some potent Hh signaling inhibitors (2c, 2d, 2e, 2f) with novel core ring systems, which had benzamide moiety at the 5-position as a key component for potent activity. The suppression of Gli1 expression with these new Hh signaling inhibitors were weaker than that of compound 1 (TAK-441) because of low pharmacokinetic property. We recognized again TAK-441 is a good compound as clinical candidate with good structural and pharmacokinetic advantages.


Pharmacology Research & Perspectives | 2018

Antidyslipidemic potential of a novel farnesoid X receptor antagonist in a hamster model of dyslipidemia: Comparative studies of other nonstatin agents

Emiko Shinozawa; Yuichiro Amano; Hiroko Yamakawa; Megumi Haba; Mitsuyuki Shimada; Ryuichi Tozawa

We attempted to clarify the therapeutic capability of antagonists of the farnesoid X receptor (FXR), a nuclear receptor that regulates lipid and bile acid metabolism. Herein, we report the antidyslipidemic effects of a novel synthesized FXR antagonist, compound‐T1, utilizing a dyslipidemic hamster model. Compound‐T1 selectively inhibited chenodeoxycholic acid‐induced FXR activation (IC50, 2.1 nmol·L−1). A hamster model of diet‐induced hyperlipidemia was prepared to investigate the antidyslipidemic effects of compound‐T1 through comparative studies of the nonstatin lipid‐modulating agents ezetimibe, cholestyramine, and torcetrapib. In the hamster model, compound‐T1 (6 mg·kg−1·day−1, p.o.) increased the level of plasma high‐density lipoprotein (HDL)‐cholesterol (+22.2%) and decreased the levels of plasma non‐HDL‐cholesterol (−43.6%) and triglycerides (−31.1%). Compound‐T1 also increased hepatic cholesterol 7α‐hydroxylase expression and fecal bile acid excretion, and decreased hepatic cholesterol content. Moreover, the hamster model could reflect clinical results of other nonstatin agents. Torcetrapib especially increased large HDL particles compared with compound‐T1. Additionally, in the human hepatoma Huh‐7 cells, compound‐T1 enhanced apolipoprotein A‐I secretion at a concentration close to its IC50 value for FXR. Our results indicated the usefulness of the hamster model in evaluating FXR antagonists and nonstatin agents. Notably, compound‐T1 exhibited beneficial effects on both blood non‐HDL‐cholesterol and HDL‐cholesterol, which are thought to involve enhancement of cholesterol catabolism and apolipoprotein A‐I production. These findings aid the understanding of the antidyslipidemic potential of FXR antagonists with a unique lipid and bile acid modulation.


Archive | 2006

Use of tak-475 together with ezetimibe for treating hyperlipidemia

Tomoyuki Nishimoto; Hiroko Yamakawa; Takeo Wada


Pharmacological Reports | 2017

Farnesoid X receptor antagonist exacerbates dyslipidemia in mice

Yuichiro Amano; Hiroko Yamakawa; Kazuko Yonemori; Mitsuyuki Shimada; Ryuichi Tozawa


Archive | 2006

Novel method of treating hyperlipidemia

Tomoyuki Nishimoto; Ryuichi Tozawa; Takeo Wada; Eiichiro Ishikawa; Toshiya Nishi; Hiroko Yamakawa


Archive | 2006

Combinations of a squalene synthase inhibitor and a hmg-coa reductase inhibitor for treating hyperlipidemia

Tomoyuki Nishimoto; Ryuichi Tozawa; Takeo Wada; Eiichiro Ishikawa; Toshiya Nishi; Hiroko Yamakawa


Archive | 2017

método para prevenir e/ou tratar hiperlipidemia e a toxidade hepatica, composição farmaceutica, método para realçar um efeito na prevenção e/ou no tratamento da hiperlipidemia, e, uso de inibidor da esqualeno sintase

Eiichiro Ishikawa; Hiroko Yamakawa; Ryuichi Tozawa; Takeo Wada; Tomoyuki Nishimoto; Toshiya Nishi

Collaboration


Dive into the Hiroko Yamakawa's collaboration.

Top Co-Authors

Avatar

Takeo Wada

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tomoyuki Nishimoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Ryuichi Tozawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hideaki Tojo

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Maki Miyamoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Sachio Shibata

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Satoshi Sasaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shigeru Kondo

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Toshiya Nishi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsuo Baba

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge