13 results on '"Okaniwa M"'
Search Results
2. The DSL domain in mutant JAG1 ligand is essential for the severity of the liver defect in Alagille syndrome
- Author
-
Yuan, Z R, Okaniwa, M, Nagata, I, Tazawa, Y, Ito, M, Kawarazaki, H, Inomata, Y, Okano, S, Yoshida, T, Kobayashi, N, and Kohsaka, T
- Published
- 2001
3. TAK-676: A Novel Stimulator of Interferon Genes (STING) Agonist Promoting Durable IFN-dependent Antitumor Immunity in Preclinical Studies.
- Author
-
Carideo Cunniff E, Sato Y, Mai D, Appleman VA, Iwasaki S, Kolev V, Matsuda A, Shi J, Mochizuki M, Yoshikawa M, Huang J, Shen L, Haridas S, Shinde V, Gemski C, Roberts ER, Ghasemi O, Bazzazi H, Menon S, Traore T, Shi P, Thelen TD, Conlon J, Abu-Yousif AO, Arendt C, Shaw MH, and Okaniwa M
- Subjects
- Animals, Humans, Mice, Cytokines, Interferons, Signal Transduction, Tumor Microenvironment, Clinical Trials, Phase I as Topic, Immunity, Innate, Neoplasms drug therapy
- Abstract
Oncology therapies targeting the immune system have improved patient outcomes across a wide range of tumor types, but resistance due to an inadequate T-cell response in a suppressive tumor microenvironment (TME) remains a significant problem. New therapies that activate an innate immune response and relieve this suppression may be beneficial to overcome this hurdle. TAK-676 is a synthetic novel stimulator of interferon genes (STING) agonist designed for intravenous administration. Here we demonstrate that TAK-676 dose-dependently triggers activation of the STING signaling pathway and activation of type I interferons. Furthermore, we show that TAK-676 is a highly potent modulator of both the innate and adaptive immune system and that it promotes the activation of dendritic cells, natural killer cells, and T cells in preclinical models. In syngeneic murine tumor models in vivo, TAK-676 induces dose-dependent cytokine responses and increases the activation and proliferation of immune cells within the TME and tumor-associated lymphoid tissue. We also demonstrate that TAK-676 dosing results in significant STING-dependent antitumor activity, including complete regressions and durable memory T-cell immunity. We show that TAK-676 is well tolerated, exhibits dose-proportional pharmacokinetics in plasma, and exhibits higher exposure in tumor. The intravenous administration of TAK-676 provides potential treatment benefit in a broad range of tumor types. Further study of TAK-676 in first-in-human phase I trials is ongoing., Significance: TAK-676 is a novel systemic STING agonist demonstrating robust activation of innate and adaptive immune activity resulting in durable antitumor responses within multiple syngeneic tumor models. Clinical investigation of TAK-676 is ongoing., Competing Interests: V.A. Appleman reports personal fees from Takeda Development Centers of America, Inc during the conduct of the study; personal fees from Takeda and other from Takeda outside the submitted work. V. Kolev reports other from Takeda during the conduct of the study. M. Mochizuki reports patent application WO2018/100558. S. Haridas reports personal fees from Takeda during the conduct of the study; personal fees from Takeda outside the submitted work. T.D. Thelen reports other from Takeda outside the submitted work. A.O. Abu-Yousif reports other from Takeda during the conduct of the study. M. Okaniwa reports patent application WO2018/100558. No other disclosures were reported., (© 2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
4. New Series of Potent Allosteric Inhibitors of Deoxyhypusine Synthase.
- Author
-
Tanaka Y, Kurasawa O, Yokota A, Klein MG, Saito B, Matsumoto S, Okaniwa M, Ambrus-Aikelin G, Uchiyama N, Morishita D, Kimura H, and Imamura S
- Abstract
Deoxyhypusine synthase (DHPS) is the primary enzyme responsible for the hypusine modification and, thereby, activation of the eukaryotic translation initiation factor 5A (eIF5A), which is key in regulating the protein translation processes associated with tumor proliferation. Although DHPS inhibitors could be a promising therapeutic option for treating cancer, only a few studies reported druglike compounds with this inhibition property. Thus, in this work, we designed and synthesized a new chemical series possessing fused ring scaffolds designed from high-throughput screening hit compounds, discovering a 5,6-dihydrothieno[2,3- c ]pyridine derivative ( 26d ) with potent inhibitory activity; furthermore, the X-ray crystallographic analysis of the DHPS complex with 26d demonstrated a distinct allosteric binding mode compared to a previously reported inhibitor. These findings could be significantly useful in the functional analysis of conformational changes in DHPS as well as the structure-based design of allosteric inhibitors., Competing Interests: The authors declare no competing financial interest.
- Published
- 2020
- Full Text
- View/download PDF
5. Discovery and pharmacological characterization of a new class of prolyl-tRNA synthetase inhibitor for anti-fibrosis therapy.
- Author
-
Shibata A, Kuno M, Adachi R, Sato Y, Hattori H, Matsuda A, Okuzono Y, Igaki K, Tominari Y, Takagi T, Yabuki M, and Okaniwa M
- Subjects
- Animals, Cells, Cultured, Drug Discovery, Enzyme Inhibitors therapeutic use, Humans, Male, Mice, Mice, Inbred C3H, Amino Acyl-tRNA Synthetases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Scleroderma, Systemic drug therapy
- Abstract
Scleroderma has clinical characteristics including skin and other tissue fibrosis, but there is an unmet need for anti-fibrotic therapy. Halofuginone (HF) is a well-known anti-fibrosis agent in preclinical and clinical studies which exerts its effect via inhibition of TGF-β/Smad3 signaling pathway. Recently, prolyl-tRNA synthetase (PRS) was elucidated as a target protein for HF that binds to the proline binding site of the catalytic domain of PRS. Here, we characterized a new class of PRS inhibitor (T-3833261) that is carefully designed in a way that binds to the ATP site of the catalytic domain and does not disrupt binding of proline. The anti-fibrotic activity and the mechanism of action for T-3833261 on TGF-β-induced fibrotic assay were compared with those of HF in primary human skin fibroblast. We evaluated in vivo effect of topical application of T-3833261 and HF on TGF-β-induced fibrotic genes expression in mice. We found that T-3833261 suppressed TGF-β-induced α-smooth muscle actin (α-SMA) and type I collagen α1 (COL1A1) expression through the Smad3 axis in a similar fashion to HF. In vivo topical application of T-3833261 reduced the increase of fibrotic genes expression such as α-Sma, Col1a1 and Col1a2 by TGF-β intradermal injection to the ear of a mouse. We revealed that T-3833261 is more effective than HF under the conditions of high proline concentration, as reported in fibrotic tissues. These results suggest the potential of ATP competitive PRS inhibitors for the treatment of fibrotic diseases such as scleroderma.
- Published
- 2017
- Full Text
- View/download PDF
6. A Kinase Inhibitor Targeted to mTORC1 Drives Regression in Glioblastoma.
- Author
-
Fan Q, Aksoy O, Wong RA, Ilkhanizadeh S, Novotny CJ, Gustafson WC, Truong AY, Cayanan G, Simonds EF, Haas-Kogan D, Phillips JJ, Nicolaides T, Okaniwa M, Shokat KM, and Weiss WA
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Inbred BALB C, Sirolimus therapeutic use, Tacrolimus Binding Protein 1A physiology, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Multiprotein Complexes antagonists & inhibitors, Protein Kinase Inhibitors therapeutic use, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Although signaling from phosphatidylinositol 3-kinase (PI3K) and AKT to mechanistic target of rapamycin (mTOR) is prominently dysregulated in high-grade glial brain tumors, blockade of PI3K or AKT minimally affects downstream mTOR activity in glioma. Allosteric mTOR inhibitors, such as rapamycin, incompletely block mTORC1 compared with mTOR kinase inhibitors (TORKi). Here, we compared RapaLink-1, a TORKi linked to rapamycin, with earlier-generation mTOR inhibitors. Compared with rapamycin and Rapalink-1, TORKi showed poor durability. RapaLink-1 associated with FKBP12, an abundant mTOR-interacting protein, enabling accumulation of RapaLink-1. RapaLink-1 showed better efficacy than rapamycin or TORKi, potently blocking cancer-derived, activating mutants of mTOR. Our study re-establishes mTOR as a central target in glioma and traces the failure of existing drugs to incomplete/nondurable inhibition of mTORC1., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
7. Two exfoliation approaches for organic layered compounds: hydrophilic and hydrophobic polydiacetylene nanosheets.
- Author
-
Ishijima Y, Okaniwa M, Oaki Y, and Imai H
- Abstract
Nanosheets have attracted much interest because of their characteristic properties originating from anisotropic and flexible structures. Inorganic nanosheets are synthesized from precursor layered compounds through exfoliation in a liquid phase. In contrast, a versatile exfoliation approach has not been fully studied for organic layered compounds. Here we report two exfoliation approaches for organic layered compounds. Hydrophilic and hydrophobic polydiacetylene (PDA) nanosheets, around 5 nm in thickness, are obtained through exfoliation of the layered precursor in aqueous and nonpolar organic media, respectively. The intercalation of ions and molecules in the interlayer space facilitates swelling and exfoliation. The resultant PDA nanosheets showed characteristic photochemical properties originating from the flexible structure. The exfoliation approach can be applied to a variety of organic layered compounds for the generation of designed nanosheets.
- Published
- 2017
- Full Text
- View/download PDF
8. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor.
- Author
-
Rodrik-Outmezguine VS, Okaniwa M, Yao Z, Novotny CJ, McWhirter C, Banaji A, Won H, Wong W, Berger M, de Stanchina E, Barratt DG, Cosulich S, Klinowska T, Rosen N, and Shokat KM
- Subjects
- Animals, Binding Sites drug effects, Cell Line, Tumor, Female, Humans, Mice, Mutation drug effects, Neoplasms drug therapy, Neoplasms enzymology, Neoplasms genetics, Neoplasms pathology, Protein Kinase Inhibitors classification, Protein Structure, Tertiary genetics, Signal Transduction drug effects, TOR Serine-Threonine Kinases chemistry, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, Drug Resistance drug effects, Drug Resistance genetics, Mutation genetics, Protein Kinase Inhibitors pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics
- Abstract
Precision medicines exert selective pressure on tumour cells that leads to the preferential growth of resistant subpopulations, necessitating the development of next-generation therapies to treat the evolving cancer. The PIK3CA-AKT-mTOR pathway is one of the most commonly activated pathways in human cancers, which has led to the development of small-molecule inhibitors that target various nodes in the pathway. Among these agents, first-generation mTOR inhibitors (rapalogs) have caused responses in 'N-of-1' cases, and second-generation mTOR kinase inhibitors (TORKi) are currently in clinical trials. Here we sought to delineate the likely resistance mechanisms to existing mTOR inhibitors in human cell lines, as a guide for next-generation therapies. The mechanism of resistance to the TORKi was unusual in that intrinsic kinase activity of mTOR was increased, rather than a direct active-site mutation interfering with drug binding. Indeed, identical drug-resistant mutations have been also identified in drug-naive patients, suggesting that tumours with activating MTOR mutations will be intrinsically resistant to second-generation mTOR inhibitors. We report the development of a new class of mTOR inhibitors that overcomes resistance to existing first- and second-generation inhibitors. The third-generation mTOR inhibitor exploits the unique juxtaposition of two drug-binding pockets to create a bivalent interaction that allows inhibition of these resistant mutants.
- Published
- 2016
- Full Text
- View/download PDF
9. A Novel Time-Dependent CENP-E Inhibitor with Potent Antitumor Activity.
- Author
-
Ohashi A, Ohori M, Iwai K, Nambu T, Miyamoto M, Kawamoto T, and Okaniwa M
- Subjects
- Animals, Cell Line, Tumor, Chromosome Segregation, Heterografts, Metaphase, Mice, Mice, Nude, Microtubules metabolism, Mitosis, Antineoplastic Agents pharmacology, Chromosomal Proteins, Non-Histone antagonists & inhibitors, Spindle Apparatus drug effects
- Abstract
Centromere-associated protein E (CENP-E) regulates both chromosome congression and the spindle assembly checkpoint (SAC) during mitosis. The loss of CENP-E function causes chromosome misalignment, leading to SAC activation and apoptosis during prolonged mitotic arrest. Here, we describe the biological and antiproliferative activities of a novel small-molecule inhibitor of CENP-E, Compound-A (Cmpd-A). Cmpd-A inhibits the ATPase activity of the CENP-E motor domain, acting as a time-dependent inhibitor with an ATP-competitive-like behavior. Cmpd-A causes chromosome misalignment on the metaphase plate, leading to prolonged mitotic arrest. Treatment with Cmpd-A induces antiproliferation in multiple cancer cell lines. Furthermore, Cmpd-A exhibits antitumor activity in a nude mouse xenograft model, and this antitumor activity is accompanied by the elevation of phosphohistone H3 levels in tumors. These findings demonstrate the potency of the CENP-E inhibitor Cmpd-A and its potential as an anticancer therapeutic agent.
- Published
- 2015
- Full Text
- View/download PDF
10. Aneuploidy generates proteotoxic stress and DNA damage concurrently with p53-mediated post-mitotic apoptosis in SAC-impaired cells.
- Author
-
Ohashi A, Ohori M, Iwai K, Nakayama Y, Nambu T, Morishita D, Kawamoto T, Miyamoto M, Hirayama T, Okaniwa M, Banno H, Ishikawa T, Kandori H, and Iwata K
- Subjects
- Animals, Caspase 3 genetics, Caspase 3 metabolism, Caspase 7, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Gene Expression Regulation, HeLa Cells, Heterografts, Humans, Kinesins antagonists & inhibitors, Mice, Mice, Nude, Mitosis, Neoplasms, Experimental, Stress, Physiological, Aneuploidy, Apoptosis, DNA Damage, M Phase Cell Cycle Checkpoints physiology, Tumor Suppressor Protein p53 physiology
- Abstract
The molecular mechanism responsible that determines cell fate after mitotic slippage is unclear. Here we investigate the post-mitotic effects of different mitotic aberrations--misaligned chromosomes produced by CENP-E inhibition and monopolar spindles resulting from Eg5 inhibition. Eg5 inhibition in cells with an impaired spindle assembly checkpoint (SAC) induces polyploidy through cytokinesis failure without a strong anti-proliferative effect. In contrast, CENP-E inhibition causes p53-mediated post-mitotic apoptosis triggered by chromosome missegregation. Pharmacological studies reveal that aneuploidy caused by the CENP-E inhibitor, Compound-A, in SAC-attenuated cells causes substantial proteotoxic stress and DNA damage. Polyploidy caused by the Eg5 inhibitor does not produce this effect. Furthermore, p53-mediated post-mitotic apoptosis is accompanied by aneuploidy-associated DNA damage response and unfolded protein response activation. Because Compound-A causes p53 accumulation and antitumour activity in an SAC-impaired xenograft model, CENP-E inhibitors could be potential anticancer drugs effective against SAC-impaired tumours.
- Published
- 2015
- Full Text
- View/download PDF
11. Antitumor activity of the selective pan-RAF inhibitor TAK-632 in BRAF inhibitor-resistant melanoma.
- Author
-
Nakamura A, Arita T, Tsuchiya S, Donelan J, Chouitar J, Carideo E, Galvin K, Okaniwa M, Ishikawa T, and Yoshida S
- Subjects
- Animals, Antineoplastic Agents pharmacology, Benzothiazoles pharmacology, Cells, Cultured, Humans, MAP Kinase Signaling System drug effects, Melanoma pathology, Mice, Mice, Nude, Nitriles pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Skin Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Benzothiazoles therapeutic use, Drug Resistance, Neoplasm drug effects, Melanoma drug therapy, Nitriles therapeutic use, Protein Kinase Inhibitors therapeutic use, Skin Neoplasms drug therapy, raf Kinases antagonists & inhibitors
- Abstract
The mitogen-activated protein kinase (MAPK) pathway is particularly important for the survival and proliferation of melanoma cells. Somatic mutations in BRAF and NRAS are frequently observed in melanoma. Recently, the BRAF inhibitors vemurafenib and dabrafenib have emerged as promising agents for the treatment of melanoma patients with BRAF-activating mutations. However, as BRAF inhibitors induce RAF paradoxical activation via RAF dimerization in BRAF wild-type cells, rapid emergence of acquired resistance and secondary skin tumors as well as presence of few effective treatment options for melanoma bearing wild-type BRAF (including NRAS-mutant melanoma) are clinical concerns. Here, we demonstrate that the selective pan-RAF inhibitor TAK-632 suppresses RAF activity in BRAF wild-type cells with minimal RAF paradoxical activation. Our analysis using RNAi and TAK-632 in preclinical models reveals that the MAPK pathway of NRAS-mutated melanoma cells is highly dependent on RAF. We also show that TAK-632 induces RAF dimerization but inhibits the kinase activity of the RAF dimer, probably because of its slow dissociation from RAF. As a result, TAK-632 demonstrates potent antiproliferative effects both on NRAS-mutated melanoma cells and BRAF-mutated melanoma cells with acquired resistance to BRAF inhibitors through NRAS mutation or BRAF truncation. Furthermore, we demonstrate that the combination of TAK-632 and the MAPK kinase (MEK) inhibitor TAK-733 exhibits synergistic antiproliferative effects on these cells. Our findings characterize the unique features of TAK-632 as a pan-RAF inhibitor and provide rationale for its further investigation in NRAS-mutated melanoma and a subset of BRAF-mutated melanomas refractory to BRAF inhibitors.
- Published
- 2013
- Full Text
- View/download PDF
12. Aluminum-controlled reactivity and diastereoselectivity toward radical reactions of optically active aldimines with metallic samarium.
- Author
-
Yanada R, Okaniwa M, Kaieda A, Ibuka T, and Takemoto Y
- Abstract
The intermolecular pinacol-type coupling reaction and allylation reaction of optically active imines bearing a beta-hydroxy group were performed stereoselectively with metallic samarium after treatment of the imines with trimethylaluminum.
- Published
- 2001
- Full Text
- View/download PDF
13. Neonatal hepatitis and extrahepatic biliary atresia in the same sibship.
- Author
-
Suda J, Nakajima S, Okaniwa M, and Kamoshita S
- Subjects
- Hepatitis etiology, Hepatitis pathology, Humans, Infant, Infant, Newborn, Infant, Newborn, Diseases pathology, Male, Bile Ducts abnormalities, Hepatitis genetics, Infant, Newborn, Diseases genetics
- Abstract
An instance of the rare occurrence of neonatal hepatitis and extrahepatic biliary atresia in the same sibship is reported. The older brother with neonatal hepatitis developed jaundice at the age of 4 days and had clay-colored stools from early infancy. Cholangiography by exploratory laparotomy at the age of 3 months showed a normal bile duct pattern. After laparotomy, jaundice rapidly disappeared, and stools became yellow. His liver function has been normal since age 6 months to the present (6 years old). The younger brother developed jaundice and clay-colored stools at the age of 1 month. The diagnosis of extrahepatic biliary atresia was made at laparotomy at the age of 4 1/2 months. Hepatojejunostomy was performed with successful bile drainage, although he had frequent attacks of ascending cholangitis since operation. These cases support a recent hypothesis that neonatal hepatitis and extrahepatic biliary atresia may be produced by the same disease process.
- Published
- 1981
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.