Network


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

Hotspot


Dive into the research topics where Tatsuki Koike is active.

Publication


Featured researches published by Tatsuki Koike.


Journal of Medicinal Chemistry | 2011

1,6-Dihydro-2H-indeno[5,4-b]furan Derivatives: Design, Synthesis, and Pharmacological Characterization of a Novel Class of Highly Potent MT2-Selective Agonists

Tatsuki Koike; Yasutaka Hoashi; Takafumi Takai; Masaharu Nakayama; Nobuhito Yukuhiro; Takashi Ishikawa; Keisuke Hirai; Osamu Uchikawa

A novel series of 1,6-dihydro-2H-indeno[5,4-b]furan derivatives were designed and synthesized as MT(2)-selective ligands. This scaffold was identified as a potent mimic of the 5-methoxy indole core of melatonin, and introduction of a cyclohexylmethyl group at the 7-position of this scaffold afforded an MT(2)-selective ligand 15 (K(i) = 0.012 nM) with high MT(1)/MT(2) selectivity (799). Compound 15 was identified as a potent full agonist for the MT(2) subtype and exhibited reentrainment effects to a new light/dark cycle in ICR mice at 3-30 mg/kg. This result demonstrated the involvement of the MT(2) receptors in chronobiotic activity.


Journal of Medicinal Chemistry | 2011

Synthesis of a Novel Series of Tricyclic Dihydrofuran Derivatives: Discovery of 8,9-Dihydrofuro[3,2-c]pyrazolo[1,5-a]pyridines as Melatonin Receptor (MT1/MT2) Ligands

Tatsuki Koike; Takafumi Takai; Yasutaka Hoashi; Masaharu Nakayama; Yohei Kosugi; Masato Nakashima; Shin-ichi Yoshikubo; Keisuke Hirai; Osamu Uchikawa

Novel tricyclic dihydrofuran derivatives were designed, synthesized, and evaluated as melatonin receptor (MT(1)/MT(2)) ligands based on the previously reported 1,6-dihydro-2H-indeno[5,4-b]furan 1a. By screening the central tricyclic cores, we identified 8,9-dihydrofuro[3,2-c]pyrazolo[1,5-a]pyridine as a potent scaffold with a high ligand-lipophilicity efficiency (LLE) value. Subsequent optimization of the side chains led to identification of the potent MT(1)/MT(2) agonist 4d (MT(1), K(i) = 0.062 nM; MT(2), K(i) = 0.420 nM) with good oral absorption and blood-brain barrier (BBB) penetration in rats. The oral administration of compound 4d exhibited a sleep-promoting action in freely moving cats at 0.1 mg/kg.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of novel 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives as γ-secretase modulators.

Takafumi Takai; Yasutaka Hoashi; Yoshihide Tomata; Sachie Morimoto; Minoru Nakamura; Tomomichi Watanabe; Tomoko Igari; Tatsuki Koike

Novel 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives were designed, synthesized, and evaluated as γ-secretase modulators (GSMs). An optimization study of this series resulted in the identification of (R)-11j, which showed a potent Aβ42-lowering effect, high bioavailability and good blood-brain barrier permeability in mice. Oral administration of (R)-11j significantly reduced brain Aβ42 in mice at a dose of 10 mg/kg.


Brain Research | 2016

Pharmacological properties of a novel and potent γ-secretase modulator as a therapeutic option for the treatment of Alzheimer’s disease

Koji Murakami; Tomomichi Watanabe; Tatsuki Koike; Makoto Kamata; Tomoko Igari; Shinichi Kondo

Previous studies of γ-secretase inhibitors (GSIs) and Notch-sparing GSIs have shown reduced amyloid-β (Aβ) peptide levels but increased Notch-related and -unrelated adverse effects. In this study, we examined the effects of compound-1 on Aβ processing and cognitive function and assessed Notch-related and -unrelated adverse effects. Compound-1 reduced Aβ40 and Aβ42 levels but inversely increased Aβ37 in Neuro2a cells, leading to no net changes in total Aβ levels, indicating that compound-1 is a γ-secretase modulator (GSM). In time-course experiments, compound-1 reduced Aβ40 and Aβ42 levels in tris-soluble fractions, with peak reduction at approximately 3h after oral administration in C57BL mice. Moreover, at >1mg/kg, compound-1 dose dependently reduced Aβ40 and Aβ42 levels in Tg2576 mice. Chronic treatment with compound-1 in Tg2576 mice for 4 months significantly reduced both soluble and insoluble Aβ42 levels and ameliorated cognitive impairments, even after drug withdrawal for 10 days following oral administration for 2 months. In contrast with compound-1, at 100-fold higher doses (100mg/kg), the GSI LY450139 decreased HES1 mRNA expression in thymus tissues and increased the intensity of periodic acid-Schiff (PAS)-positive areas in the intestine. Moreover, the Notch-sparing GSI BMS708163 led to amyloid precursor protein (APP)-β-C-terminal fragment accumulation in mouse primary neurons. BMS708163 significantly hampered cognitive function in normal mice 1 month after administration, whereas compound-1 significantly improved cognitive function. Taken together, the present novel and orally active GSM is a promising molecule for the treatment of pathologies associated with Aβ42 and Aβ40.


Bioorganic & Medicinal Chemistry | 2016

Discovery of novel 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives as γ-secretase modulators (Part 2).

Takafumi Takai; Tatsuki Koike; Minoru Nakamura; Yuichi Kajita; Toshiro Yamashita; Naohiro Taya; Tetsuya Tsukamoto; Tomomichi Watanabe; Koji Murakami; Tomoko Igari; Makoto Kamata

γ-Secretase modulators (GSMs), which lower pathogenic amyloid beta (Aβ) without affecting the production of total Aβ or Notch signal, have emerged as a potential therapeutic agent for Alzheimers disease (AD). A novel series of 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives was discovered and characterized as GSMs. Optimization of substituents at the 8-position of the core scaffold using ligand-lipophilicity efficiency (LLE) as a drug-likeness guideline led to identification of various types of high-LLE GSMs. Phenoxy compound (R)-17 exhibited especially high LLE as well as potent in vivo Aβ42-lowering effect by single administration. Furthermore, multiple oral administration of (R)-17 significantly reduced soluble and insoluble brain Aβ42, and ameliorated cognitive deficit in novel object recognition test (NORT) using Tg2576 mice as an AD model.


Bioorganic & Medicinal Chemistry | 2015

Design and synthesis of piperazine derivatives as a novel class of γ-secretase modulators that selectively lower Aβ42 production

Takafumi Takai; Tatsuki Koike; Eiji Honda; Yuichi Kajita; Minoru Nakamura; Sachie Morimoto; Yasutaka Hoashi; Makoto Kamata; Tomomichi Watanabe; Tomoko Igari; Jun Terauchi

Novel piperazine derivatives as γ-secretase modulators (GSMs) were prepared and tested for their ability to selectively lower Aβ₄₂ production. Lead compound 3, with selective Aβ₄₂-lowering activity, was modified by replacing its imidazolylphenyl moiety with an oxazolylphenyl moiety. Optimization of the urea group significantly improved mouse microsomal stability, while retaining both activity and selectivity. These efforts led to the successful identification of an orally available and brain-penetrant GSM, 6j, which selectively reduced brain Aβ₄₂ in mice.


Journal of Medicinal Chemistry | 2018

Design, Synthesis, and Evaluation of Piperazinyl Pyrrolidin-2-ones as a Novel Series of Reversible Monoacylglycerol Lipase Inhibitors

Jumpei Aida; Makoto Fushimi; Tomokazu Kusumoto; Hideyuki Sugiyama; Naoto Arimura; Shuhei Ikeda; Masako Sasaki; Satoshi Sogabe; Kazunobu Aoyama; Tatsuki Koike

Monoacylglycerol lipase (MAGL) is a major serine hydrolase that hydrolyzes 2-arachidonoylglycerol (2-AG) to arachidonic acid (AA) and glycerol in the brain. Because 2-AG and AA are endogenous biologically active ligands in the brain, inhibition of MAGL is an attractive therapeutic target for CNS disorders, particularly neurodegenerative diseases. In this study, we report the structure-based drug design of novel piperazinyl pyrrolidin-2-ones starting from our hit compounds 2a and 2b. By enhancing the interaction of the piperazinyl pyrrolidin-2-one core and its substituents with the MAGL enzyme via design modifications, we identified a potent and reversible MAGL inhibitor, compound ( R)-3t. Oral administration of compound ( R)-3t to mice decreased AA levels and elevated 2-AG levels in the brain.


Alzheimers & Dementia | 2015

Pharmacologic properties of compound-1, a novel and orally active γ-secretase modulator, as a therapeutic option for the treatment of Alzheimer's disease

Koji Murakami; Tomomichi Watanabe; Tatsuki Koike; Makoto Kamata; Tomoko Igari; Shinichi Kondou

compounds in which the anionic terminus interacted with the cationic “H” residues of Ab and the cationic terminus interacted with the anionic “E” residue of Ab were identified. Although the standard alpha amino acids showed no capacity to inhibit Ab aggregation, a number of atypical amino acids including b-alanine, 2aminoethanesulfonic acid, and L-phosphoserine demonstrated varying capacities to inhibit the oligomerization and aggregation of Ab. These predictions were verified using in vitro assays, including the kinetic Thioflavin T [ThT] aggregation assay, to demonstrate the capacity of these compounds to inhibit misfolding. Conclusions: Searching for an “endogenous anti-AD compound” represents an unexplored concept. Our in silico and in vitro studies suggest that compounds endogenous to the human brain, including several zwitterionic small molecules, can inhibit pathological protein misfolding of Ab. The value of a novel in silico screening assay to identify bioactive endogenous agents within brain has also been demonstrated.


Archive | 2003

Substituted amino compounds and use thereof

Osamu Uchikawa; Kazuyoshi Aso; Tatsuki Koike; Naoki Tarui; Keisuke Hirai


Tetrahedron Letters | 2011

Synthesis of 4-aza analog of ramelteon: a novel tricyclic 1,6,7,8-tetrahydro-2H-cyclopenta[d]furo[2,3-b]pyridine derivative as melatonin receptor ligand

Tatsuki Koike; Yasutaka Hoashi; Takafumi Takai; Osamu Uchikawa

Collaboration


Dive into the Tatsuki Koike's collaboration.

Top Co-Authors

Avatar

Takafumi Takai

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yasutaka Hoashi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Osamu Uchikawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Makoto Kamata

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masato Yoshikawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Minoru Nakamura

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tomoko Igari

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tomomichi Watanabe

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

William Farnaby

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yuichi Kajita

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge