Network


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

Hotspot


Dive into the research topics where Shojiro Kadono is active.

Publication


Featured researches published by Shojiro Kadono.


Protein Engineering Design & Selection | 2013

Engineered antibody Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIaR131 and FcγRIIaH131

Futa Mimoto; Hitoshi Katada; Shojiro Kadono; Tomoyuki Igawa; Taichi Kuramochi; M. Muraoka; Y. Wada; Kenta Haraya; T. Miyazaki; Kunihiro Hattori

Engaging inhibitory FcγRIIb by Fc region has been recently reported to be an attractive approach for improving the efficacy of antibody therapeutics. However, the previously reported S267E/L328F variant with enhanced binding affinity to FcγRIIb, also enhances binding affinity to FcγRIIaR131 allotype to a similar degree because FcγRIIb and FcγRIIaR131 are structurally similar. In this study, we applied comprehensive mutagenesis and structure-guided design based on the crystal structure of the Fc/FcγRIIb complex to identify a novel Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIaR131 and FcγRIIaH131. This novel variant has more than 200-fold stronger binding affinity to FcγRIIb than wild-type IgG1, while binding affinity to FcγRIIaR131 and FcγRIIaH131 is comparable with or lower than wild-type IgG1. This selectivity was achieved by conformational change of the CH2 domain by mutating Pro to Asp at position 238. Fc variant with increased binding to both FcγRIIb and FcγRIIa induced platelet aggregation and activation in an immune complex form in vitro while our novel variant did not. When applied to agonistic anti-CD137 IgG1 antibody, our variant greatly enhanced the agonistic activity. Thus, the selective enhancement of FcγRIIb binding achieved by our Fc variant provides a novel tool for improving the efficacy of antibody therapeutics.


mAbs | 2013

Novel asymmetrically engineered antibody Fc variant with superior FcγR binding affinity and specificity compared with afucosylated Fc variant.

Futa Mimoto; Tomoyuki Igawa; Taichi Kuramochi; Hitoshi Katada; Shojiro Kadono; Takayuki Kamikawa; Meiri Shida-Kawazoe; Kunihiro Hattori

Fc engineering is a promising approach to enhance the antitumor efficacy of monoclonal antibodies (mAbs) through antibody-dependent cell-mediated cytotoxicity (ADCC). Glyco- and protein-Fc engineering have been employed to enhance FcγR binding and ADCC activity of mAbs; the drawbacks of previous approaches lie in their binding affinity to both FcγRIIIa allotypes, the ratio of activating FcγR binding to inhibitory FcγR binding (A/I ratio) or the melting temperature (TM) of the CH2 domain. To date, no engineered Fc variant has been reported that satisfies all these points. Herein, we present a novel Fc engineering approach that introduces different substitutions in each Fc domain asymmetrically, conferring optimal binding affinity to FcγR and specificity to the activating FcγR without impairing the stability. We successfully designed an asymmetric Fc variant with the highest binding affinity for both FcγRIIIa allotypes and the highest A/I ratio compared with previously reported symmetrically engineered Fc variants, and superior or at least comparable in vitro ADCC activity compared with afucosylated Fc variants. In addition, the asymmetric Fc engineering approach offered higher stability by minimizing the use of substitutions that reduce the TM of the CH2 domain compared with the symmetric approach. These results demonstrate that the asymmetric Fc engineering platform provides best-in-class effector function for therapeutic antibodies against tumor antigens.


Molecular Immunology | 2014

Crystal structure of a novel asymmetrically engineered Fc variant with improved affinity for FcγRs

Futa Mimoto; Shojiro Kadono; Hitoshi Katada; Tomoyuki Igawa; T. Kamikawa; Kunihiro Hattori

Enhancing the effector function by optimizing the interaction between Fc and Fcγ receptor (FcγR) is a promising approach to enhance the potency of anticancer monoclonal antibodies (mAbs). To date, a variety of Fc engineering approaches to modulate the interaction have been reported, such as afucosylation in the heavy chain Fc region or symmetrically introducing amino acid substitutions into the region, and there is still room to improve FcγR binding and thermal stability of the CH2 domain with these approaches. Recently, we have reported that asymmetric Fc engineering, which introduces different substitutions into each Fc region of heavy chain, can further improve the FcγR binding while maintaining the thermal stability of the CH2 domain by fine-tuning the asymmetric interface between the Fc domain and FcγR. However, the structural mechanism by which the asymmetrically engineered Fc improved FcγR binding remained unclear. In order to elucidate the mechanism, we solved the crystal structure of a novel asymmetrically engineered Fc, asym-mAb23, in complex with FcγRIIIa. Asym-mAb23 has enhanced binding affinity for both FcγRIIIa and FcγRIIa at the highest level of previously reported Fc variants. The structural analysis reveals the features of the asymmetrically engineered Fc in comparison with symmetric Fc and how each asymmetrically introduced substitution contributes to the improved interaction between asym-mAb23 and FcγRIIIa. This crystal structure could be utilized to enable us to design a more potent asymmetric Fc.


Bioorganic & Medicinal Chemistry Letters | 2008

Factor VIIa inhibitors: target hopping in the serine protease family using X-ray structure determination.

Takuya Shiraishi; Shojiro Kadono; Masayuki Haramura; Hirofumi Kodama; Yoshiyuki Ono; Hitoshi Iikura; Tohru Esaki; Takaki Koga; Kunihiro Hattori; Yoshiaki Watanabe; Akihisa Sakamoto; Kazutaka Yoshihashi; Takehisa Kitazawa; Keiko Esaki; Masateru Ohta; Haruhiko Sato; Toshiro Kozono

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is regarded as a promising target for developing new anticoagulant drugs. Compound 1 was discovered from focused screening of serine protease-directed compounds from our internal collection. Using parallel synthesis supported by structure-based drug design, we identified peptidemimetic FVIIa/TF inhibitors (compounds 4-11) containing L-Gln or L-Met as the P2 moiety. However, these compounds lacked the selectivity of other serine proteases in the coagulation cascade, especially thrombin. Further optimization of these compounds was carried out with a focus on the P4 moiety. Among the optimized compounds, 12b-f showed improved selectivity.


mAbs | 2016

Calcium-dependent antigen binding as a novel modality for antibody recycling by endosomal antigen dissociation

N Hironiwa; Shinya Ishii; Shojiro Kadono; Yuki Iwayanagi; Futa Mimoto; K Habu; Tomoyuki Igawa; Kunihiro Hattori

The pH-dependent antigen binding antibody, termed a recycling antibody, has recently been reported as an attractive type of second-generation engineered therapeutic antibody. A recycling antibody can dissociate antigen in the acidic endosome, and thus bind to its antigen multiple times. As a consequence, a recycling antibody can neutralize large amounts of antigen in plasma. Because this approach relies on histidine residues to achieve pH-dependent antigen binding, which could limit the epitopes that can be targeted and affect the rate of antigen dissociation in the endosome, we explored an alternative approach for generating recycling antibodies. Since calcium ion concentration is known to be lower in endosome than in plasma, we hypothesized that an antibody with antigen-binding properties that are calcium-dependent could be used as recycling antibody. Here, we report a novel anti-interleukin-6 receptor (IL-6R) antibody, identified from a phage library that binds to IL-6R only in the presence of a calcium ion. Thermal dynamics and a crystal structure study revealed that the calcium ion binds to the heavy chain CDR3 region (HCDR3), which changes and possibly stabilizes the structure of HCDR3 to make it bind to antigen calcium dependently (PDB 5AZE). In vitro and in vivo studies confirmed that this calcium-dependent antigen-binding antibody can dissociate its antigen in the endosome and accelerate antigen clearance from plasma, making it a novel approach for generating recycling antibody.


Acta Crystallographica Section F-structural Biology and Crystallization Communications | 2005

Structure of human factor VIIa/tissue factor in complex with a peptide-mimetic inhibitor: high selectivity against thrombin by introducing two charged groups in P2 and P4.

Shojiro Kadono; Akihisa Sakamoto; Yasufumi Kikuchi; Masayoshi Oh-eda; Naohiro Yabuta; Takaki Koga; Kunihiro Hattori; Takuya Shiraishi; Masayuki Haramura; Hirofumi Kodama; Yoshiyuki Ono; Toru Esaki; Haruhiko Sato; Yoshiaki Watanabe; Susumu Itoh; Masateru Ohta; Toshiro Kozono

The crystal structure of human factor VIIa/soluble tissue factor (FVIIa/sTF) in complex with a highly selective peptide-mimetic FVIIa inhibitor which shows 1670-fold selectivity against thrombin inhibition has been solved at 2.6 A resolution. The inhibitor is bound to FVIIa/sTF at the S1, S2 and S3 sites and at the additional S1 subsite. Two charged groups, the amidino group in P2 and the carboxylate group in P4, form ionic interactions with Asp60 and Lys192 of FVIIa, respectively. Structural comparisons between factor VIIa and thrombin show that thrombin has oppositely charged residues, Lys60F and Glu192, in the S2 site and the S1 subsites, respectively. These data suggest that the utilization of the differences of charge distribution in the S2 site and the S1 subsites between FVIIa and thrombin is critical for achieving high selectivity against thrombin inhibition. These results will provide valuable information for the structure-based drug design of specific inhibitors for FVIIa/TF.


Archive | 2012

Fcγriib-specific fc antibody

Futa Mimoto; 風太 味元; Taichi Kuramochi; 太一 倉持; Tomoyuki Igawa; 智之 井川; Hitoshi Katada; 仁 堅田; Shojiro Kadono; 正次郎 門野


Archive | 2013

ANTIGEN-BINDING MOLECULE FOR PROMOTING DISAPPEARANCE OF ANTIGEN VIA FcγRIIB

Tomoyuki Igawa; 智之 井川; Atsuhiko Maeda; 敦彦 前田; Yuki Iwayanagi; 有起 岩柳; Kenta Haraya; 健太 原谷; Hitoshi Katada; 仁 堅田; Shojiro Kadono; 正次郎 門野; Futa Mimoto; 風太 味元


Biochemical and Biophysical Research Communications | 2004

Crystal structure of human factor VIIa/tissue factor in complex with peptide mimetic inhibitor

Shojiro Kadono; Akihisa Sakamoto; Yasufumi Kikuchi; Masayoshi Oh-eda; Naohiro Yabuta; Takaki Koga; Kunihiro Hattori; Takuya Shiraishi; Masayuki Haramura; Hirofumi Kodama; Toru Esaki; Haruhiko Sato; Yoshiaki Watanabe; Susumu Itoh; Masateru Ohta; Toshiro Kozono


Archive | 2012

Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity

Tomoyuki Igawa; 智之 井川; Atsuhiko Maeda; 敦彦 前田; Kenta Haraya; 健太 原谷; Yuki Iwayanagi; 有起 岩柳; Tatsuhiko Tachibana; 橘 達彦; Futa Mimoto; 風太 味元; Taichi Kuramochi; 太一 倉持; Hitoshi Katada; 仁 堅田; Shojiro Kadono; 正次郎 門野

Collaboration


Dive into the Shojiro Kadono's collaboration.

Top Co-Authors

Avatar

Tomoyuki Igawa

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Futa Mimoto

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Hitoshi Katada

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masateru Ohta

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hirofumi Kodama

Chugai Pharmaceutical Co.

View shared research outputs
Researchain Logo
Decentralizing Knowledge