Network


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

Hotspot


Dive into the research topics where Toshihiko Ohtomo is active.

Publication


Featured researches published by Toshihiko Ohtomo.


Journal of Biological Chemistry | 2003

A resorcylic acid lactone, 5Z-7-oxozeaenol, prevents inflammation by inhibiting the catalytic activity of TAK1 MAPK kinase kinase.

Jun Ninomiya-Tsuji; Taisuke Kajino; Koichiro Ono; Toshihiko Ohtomo; Masahiko Matsumoto; Masashi Shiina; Masahiko Mihara; Masayuki Tsuchiya; Kunihiro Matsumoto

TAK1, a member of the mitogen-activated kinase kinase kinase (MAPKKK) family, participates in proinflammatory cellular signaling pathways by activating JNK/p38 MAPKs and NF-κB. To identify drugs that prevent inflammation, we screened inhibitors of TAK1 catalytic activity. We identified a natural resorcylic lactone of fungal origin, 5Z-7-oxozeaenol, as a highly potent inhibitor of TAK1. This compound did not effectively inhibit the catalytic activities of the MEKK1 or ASK1 MAPKKKs, suggesting that 5Z-7-oxozeaenol is a selective inhibitor of TAK1. In cell culture, 5Z-7-oxozeaenol blocked interleukin-1-induced activation of TAK1, JNK/p38 MAPK, IκB kinases, and NF-κB, resulting in inhibition of cyclooxgenase-2 production. Furthermore, in vivo 5Z-7-oxozeaenol was able to inhibit picryl chloride-induced ear swelling. Thus, 5Z-7-oxozeaenol blocks proinflammatory signaling by selectively inhibiting TAK1 MAPKKK.


Clinical Cancer Research | 2013

First-in-Man Phase I Study of GC33, A Novel Recombinant Humanized Antibody against Glypican-3, in Patients with Advanced Hepatocellular Carcinoma

Andrew X. Zhu; Philip J. Gold; Anthony B. El-Khoueiry; Thomas Adam Abrams; Hideo Morikawa; Norihisa Ohishi; Toshihiko Ohtomo; Philip A. Philip

Purpose: GC33 is a novel recombinant fully humanized monoclonal antibody that binds to human glypican-3 (GPC3). The antitumor activity of GC33 was shown in preclinical models of hepatocellular carcinoma (HCC). This first-in-man clinical trial was conducted to evaluate the safety, pharmacokinetic characteristics, and preliminary efficacy of GC33 in patients with advanced HCC. Experimental Design: Patients with measurable, histologically proven, advanced HCC were enrolled to a dose-escalation study of GC33 (2.5–20 mg/kg) given intravenously weekly. The primary endpoint was to determine the maximum tolerated dose of GC33 for further development. Pharmacokinetic characteristics were measured in serum samples. Immunohistochemistry was conducted on tumor biopsies to evaluate GPC3 expression. Tumor response was assessed every 8 weeks using Response Evaluation Criteria in Solid Tumors criteria. Results: Twenty patients were enrolled and treated with GC33. A maximum tolerated dose was not reached as there were no dose-limiting toxicities (DLT) up to the highest planned dose level. Common adverse events with all grades included fatigue (50%), constipation (35%), headache (35%), and hyponatremia (35%). The incidence of adverse events seemed not to be dose dependent. Trough serum concentrations at steady state were in excess of target concentration at doses of 5 mg/kg or greater. Median time to progression (TTP) was 26.0 weeks in the GPC3 high expression group and 7.1 weeks in the low expression group (P = 0.033). Conclusion: This study shows that GC33 was well tolerated in advanced HCC and provides preliminary evidence that GPC3 expression in HCC may be associated with the clinical benefit to GC33 that warrants prospective evaluation. Clin Cancer Res; 19(4); 920–8. ©2012 AACR.


Molecular Immunology | 1999

The humanized anti-HM1.24 antibody effectively kills multiple myeloma cells by human effector cell-mediated cytotoxicity

Koichiro Ono; Toshihiko Ohtomo; Yasushi Yoshimura; Shigeto Kawai; Yasuo Koishihara; Shuji Ozaki; Masaaki Kosaka; Masayuki Tsuchiya

A mouse monoclonal antibody, anti-HM1.24 (IgG2a/kappa), binds to a surface antigen preferentially overexpressed on multiple myeloma (MM) cells, and exhibits potent antitumor cell activity against MM cells by antibody-dependent cell-mediated cytotoxicity (ADCC). To develop an antibody-based immunotherapy against MM, a humanized anti-HM1.24 antibody, in which all FRs correspond to naturally processed human FRs, has been successfully constructed with the aid of both the hybrid variable region and two-step design methods. This humanized anti-HM1.24 antibody (IgG1/kappa) is able to effectively induce ADCC against human myeloma KPMM2 and ARH77 cells in the presence of human PBMCs as effectively as a chimeric anti-HM1.24 antibody. The humanized anti-HM1.24 antibody, therefore, could be expected as a potent immunotherapeutic agent for MM patients.


Clinical Cancer Research | 2006

Identification of ROBO1 as a Novel Hepatocellular Carcinoma Antigen and a Potential Therapeutic and Diagnostic Target

Hirotaka Ito; Shin-Ichi Funahashi; Naoko Yamauchi; Junji Shibahara; Yutaka Midorikawa; Shigeto Kawai; Yasuko Kinoshita; Akira Watanabe; Yoshitaka Hippo; Toshihiko Ohtomo; Hiroko Iwanari; Atsushi Nakajima; Masatoshi Makuuchi; Masashi Fukayama; Yuichi Hirata; Takao Hamakubo; Tatsuhiko Kodama; Masayuki Tsuchiya; Hiroyuki Aburatani

Purpose: Hepatocellular carcinoma is the most common primary malignancy of the liver and accounts for as many as one million deaths annually worldwide. The present study was done to identify new transmembrane molecules for antibody therapy in hepatocellular carcinoma. Experimental Design: Gene expression profiles of pooled total RNA from three tissues each of moderately differentiated and poorly differentiated hepatocellular carcinoma were compared with those of normal liver, noncancerous liver tissue in hepatocellular carcinoma patients, 30 normal tissue samples, and five fetal tissue samples. Target genes up-regulated specifically in hepatocellular carcinoma were validated by immunohistochemical analysis and complement-dependent cytotoxicity assay using monoclonal antibodies generated against target molecules. Results: The human homologue of the Drosophila Roundabout gene, axon guidance receptor homologue 1, ROBO1/DUTT1, a member of the immunoglobulin superfamily, was highly expressed in hepatocellular carcinoma, whereas it showed only a limited distribution in normal tissues. On immunohistochemical analysis using a newly generated anti-ROBO1 monoclonal antibody, positive signals were observed in 83 of 98 cases of hepatocellular carcinoma (84.7%). The mAb B2318C induced complement-dependent cytotoxicity in ROBO1-expressing cell lines and in the liver cancer cell line PLC/PRF/5. Strikingly, the ectodomain of ROBO1 was detected not only in the culture medium of liver cancer cell lines (PLC/PRF/5, HepG2, etc.) but also in sera from hepatocellular carcinoma patients (6 of 11). Conclusions: This is the first report that ROBO1 is overexpressed in hepatocellular carcinoma and shed into serum in humans. These observations suggest that ROBO1 is a potential new serologic marker for hepatocellular carcinoma and may represent a new therapeutic target.


Journal of Biological Chemistry | 2001

An Evolutionarily Conserved Motif in the TAB1 C-terminal Region Is Necessary for Interaction with and Activation of TAK1 MAPKKK

Koichiro Ono; Toshihiko Ohtomo; Shigeo Sato; Yasuhiro Sugamata; Maho Suzuki; Naoki Hisamoto; Jun Ninomiya-Tsuji; Masayuki Tsuchiya; Kunihiro Matsumoto

TAK1, a member of the MAPKKK family, is involved in the intracellular signaling pathways mediated by transforming growth factor β, interleukin 1, and Wnt. TAK1 kinase activity is specifically activated by the TAK1-binding protein TAB1. The C-terminal 68-amino acid sequence of TAB1 (TAB1-C68) is sufficient for TAK1 interaction and activation. Analysis of various truncated versions of TAB1-C68 defined a C-terminal 30-amino acid sequence (TAB1-C30) necessary for TAK1 binding and activation. NMR studies revealed that the TAB1-C30 region has a unique α-helical structure. We identified a conserved sequence motif, PYVDXA/TXF, in the C-terminal domain of mammalian TAB1, Xenopus TAB1, and itsCaenorhabditis elegans homolog TAP-1, suggesting that this motif constitutes a specific TAK1 docking site. Alanine substitution mutagenesis showed that TAB1 Phe-484, located in the conserved motif, is crucial for TAK1 binding and activation. The C. eleganshomolog of TAB1, TAP-1, was able to interact with and activate theC. elegans homolog of TAK1, MOM-4. However, the site in TAP-1 corresponding to Phe-484 of TAB1 is an alanine residue (Ala-364), and changing this residue to Phe abrogates the ability of TAP-1 to interact with and activate MOM-4. These results suggest that the Phe or Ala residue within the conserved motif of the TAB1-related proteins is important for interaction with and activation of specific TAK1 MAPKKK family members in vivo.


Cancer Science | 2005

Overexpression of MUC13 is associated with intestinal-type gastric cancer

Takahiro Shimamura; Hirotaka Ito; Junji Shibahara; Akira Watanabe; Yoshitaka Hippo; Hirokazu Taniguchi; Yongxin Chen; Takeshi Kashima; Toshihiko Ohtomo; Fumihiko Tanioka; Hiroko Iwanari; Tatsuhiko Kodama; Teruhisa Kazui; Haruhiko Sugimura; Masashi Fukayama; Hiroyuki Aburatani

Mucins are secreted or transmembrane glycoproteins that are expressed mainly in the digestive tract. This family of proteins has been the focus of much gastric cancer research as some transmembrane mucins are implicated in tumorigenesis and make attractive targets for cancer diagnosis and therapeutics. Mucins have also been utilized to classify gastric cancer by differentiating between gastric and intestinal phenotypes. Here we show that transmembrane mucin MUC13 is upregulated in gastric cancer. By quantitative real‐time reverse transcription‐polymerase chain reaction and immunoblot analysis, overexpression of MUC13 was verified in more than half of the samples examined. In immunohistochemical analysis, MUC13 staining was observed in 74 of 114 cases of gastric cancer (64.9%), predominantly in intestinal type (P < 0.001), and in 9 of 10 cases of intestinal metaplasia, precancerous lesions of intestinal‐type gastric cancer, but not observed in normal gastric mucosa. Moreover, MUC13 staining patterns characteristic of histological type were identified: staining was on the apical side of tubular glands in intestinal type and on the cytoplasm in diffuse type. These results suggest that MUC13 is a good differentiation marker for gastrointestinal mucosa and that it may have a causal role that correlates with two distinct gastric tumorigenesis pathways. (Cancer Sci 2005; 96: 265 –274)


Biochemical and Biophysical Research Communications | 2003

A dominant negative TAK1 inhibits cellular fibrotic responses induced by TGF-β

Koichiro Ono; Toshihiko Ohtomo; Jun Ninomiya-Tsuji; Masayuki Tsuchiya

Transforming growth factor-beta (TGF-beta) is crucially virulent in the progression of fibrotic disorders. TAK1 (TGF-beta activated kinase 1) is one of the mitogen-activated kinase kinase kinase (MAPKKK) that is involved in TGF-beta signal transduction. To elucidate the importance of TAK1 in TGF-beta-induced fibrotic marker expression, we investigated whether dominant negative TAK1 could suppress TGF-beta signaling. Based on the finding that TAB1 (TAK1 binding protein 1) binding to TAK1 is required for TAK1 activation, a minimal portion of TAK1 lacking kinase activity that binds to TAB1 was designed as a TAK1 dominant negative inhibitor (TAK1-DN). The effect of TAK1-DN was assessed in the cells that respond to TGF-beta stimulation and that lead to the increase in production of extracellular matrix (ECM) proteins. TAK1-DN, indeed, decreased the ECM protein production, indicating that TAK1-DN retains the ability to intercept the TGF-beta signaling effectively.


Liver International | 2011

Prognostic significance of circumferential cell surface immunoreactivity of glypican‐3 in hepatocellular carcinoma

Kenji Yorita; Nobuyasu Takahashi; Hirotake Takai; Atsuhiko Kato; Masami Suzuki; Takahiro Ishiguro; Toshihiko Ohtomo; Koki Nagaike; Kazuhiro Kondo; Kazuo Chijiiwa; Hiroaki Kataoka

Background: GC33 is a recently developed monoclonal antibody against human glypican‐3 (GPC3), which is significantly upregulated in hepatocellular carcinoma (HCC). GC33 recognizes a GPC3 ectodomain and shows significant antitumour activity in vivo. Thus, humanized GC33 antibody may be a promising tool for treating HCC having cell surface GPC3 expression.


Molecular Immunology | 1995

Humanization of mouse ONS-M21 antibody with the aid of hybrid variable regions.

Toshihiko Ohtomo; Masayuki Tsuchiya; Koh Sato; Keiji Shimizu; Syusuke Moriuchi; Yasuyoshi Miyao; Toshio Akimoto; Kenichi Akamatsu; Toru Hayakawa; Yoshiyuki Ohsugi

Mouse monoclonal antibody, ONS-M21, directed against human medulloblastoma cells, has been humanized by complementarity determining region (CDR) grafting. A humanized ONS-M21 VH region, comparable to the original mouse ONS-M21 VH region, was easily constructed based on framework regions (FRs) 1, 2 and 3 from human EU antibody and on FR4 from human ND antibody. Five alterations in the FRs were made at amino acids 27, 28, 29, 30 and 94 which are all part of the canonical structure for CDR1 (H1). The humanized ONS-M21 VL regions were constructed based on the FRs from human REI antibody. We first identified five amino acid residues in the FRs at positions 20, 21, 71, 73 and 87 as having a possible adverse influences on antigen binding. None of the versions with a variety of combinations at these five positions showed any bindings to antigen. In order to identify the mouse residues that must be retained in the human FRs, hybrid VL regions were constructed by joining the mouse ONS-M21 VL region and the first humanized version within CDR2. The hybrid VL regions revealed that residues in FR1 and/or FR2 were critical in creating a functional antigen binding site. Redesigning several versions with alterations in FR1 and FR2 revealed that the Pro-46 residue was the only critical residue for creating an antigen binding site. This approach should be helpful in identifying key residues in difficult cases of antibody humanization.


Human antibodies | 1996

Humanization of an anti-human IL-6 mouse monoclonal antibody glycosylated in its heavy chain variable region

Koh Sato; Toshihiko Ohtomo; Yuichi Hirata; Hiroyuki Saito; Tetsu Matsuura; Toshio Akimoto; Kenichi Akamatsu; Yasuo Koishihara; Yoshiyuki Ohsugi; Masayuki Tsuchiya

Interleukin-6 (IL-6) inhibitors are good potential therapeutic agents in human patients, and anti-IL-6 antibodies are among the best candidates. Here, we have successfully humanized mouse monoclonal antibody SK2, which specifically binds to IL-6 and strongly inhibits IL-6 functions. Since this antibody possesses N-linked carbohydrates on Asn-30 of VH region, which seems to be very close to an antigen-binding site, influence of these carbohydrates on antigen-binding was investigated. A biosensor study showed that the mouse SK2 Fab and its deglycosylated fragments had almost equal Kd (Kon/Koff), 26.8 nM (1.05 x 10(6)/2.81 x 10(-2)) and 24.7 nM (1.28 x 10(6)/3.15 x 10(-2)), respectively. Furthermore, a mutant chimeric SK2 antibody, in which the N-glycosylation site was removed from the VH region, showed a Kd of 11 nM, almost similar to that of the original chimeric SK2 antibody, determined by Scatchard analysis with 125I-IL-6. These data indicate the carbohydrates of mouse SK2 VH region do not significantly influence antigen-binding activity. In the next step, two versions of each humanized SK2 VL and VH regions were carefully designed based on the amino acid sequences of human REI and DAW, respectively. Only one alteration, Tyr to Phe, was made at position 71 in the two light chains, according to the canonical residue for LI. A N-glycosylation site was introduced on the two heavy chains, by changing Ser to Asn at position 30. All four combinations of humanized light and heavy chains could bind to IL-6 as well as the chimeric SK2 antibody. The light chain first version, however, could not efficiently inhibit IL-6 binding to its receptor, indicating the importance of the LI loop conformation for the inhibitory activity of SK2 antibody. In contrast, both versions of the heavy chains were comparable, in yielding good humanized SK2 antibodies, suggesting that the glycosylation of the SK2 VH region has no influence in recreating a functional antigen-binding site in this humanization.

Collaboration


Dive into the Toshihiko Ohtomo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Koichiro Ono

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Naohiro Yabuta

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroyuki Aburatani

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge