Network


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

Hotspot


Dive into the research topics where Taichi Kuramochi is active.

Publication


Featured researches published by Taichi Kuramochi.


mAbs | 2011

Engineering the variable region of therapeutic IgG antibodies

Tomoyuki Igawa; Hiroyuki Tsunoda; Taichi Kuramochi; Zenjiro Sampei; Shinya Ishii; Kunihiro Hattori

Since the first generation of humanized IgG1 antibodies reached the market in the late 1990s, IgG antibody molecules have been extensively engineered. The success of antibody therapeutics has introduced severe competition in developing novel therapeutic monoclonal antibodies, especially for promising or clinically validated targets. Such competition has led researchers to generate so-called second or third generation antibodies with clinical differentiation utilizing various engineering and optimization technologies. Parent IgG antibodies can be engineered to have improved antigen binding properties, effector functions, pharmacokinetics, pharmaceutical properties and safety issues. Although the primary role of the antibody variable region is to bind to the antigen, it is also the main source of antibody diversity and its sequence affects various properties important for developing antibody therapeutics. Here we review recent research activity in variable region engineering to generate superior antibody therapeutics.


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.


Blood | 2014

Anti-factor IXa/X bispecific antibody ACE910 prevents joint bleeds in a long-term primate model of acquired hemophilia A

Atsushi Muto; Kazutaka Yoshihashi; Minako Takeda; Takehisa Kitazawa; Tetsuhiro Soeda; Tomoyuki Igawa; Zenjiro Sampei; Taichi Kuramochi; Akihisa Sakamoto; Kenta Haraya; Kenji Adachi; Yoshiki Kawabe; Keiji Nogami; Midori Shima; Kunihiro Hattori

ACE910 is a humanized anti-factor IXa/X bispecific antibody mimicking the function of factor VIII (FVIII). We previously demonstrated in nonhuman primates that a single IV dose of ACE910 exerted hemostatic activity against hemophilic bleeds artificially induced in muscles and subcutis, and that a subcutaneous (SC) dose of ACE910 showed a 3-week half-life and nearly 100% bioavailability, offering support for effective prophylaxis for hemophilia A by user-friendly SC dosing. However, there was no direct evidence that such SC dosing of ACE910 would prevent spontaneous bleeds occurring in daily life. In this study, we newly established a long-term primate model of acquired hemophilia A by multiple IV injections of an anti-primate FVIII neutralizing antibody engineered in mouse-monkey chimeric form to reduce its antigenicity. The monkeys in the control group exhibited various spontaneous bleeding symptoms as well as continuous prolongation of activated partial thromboplastin time; notably, all exhibited joint bleeds, which are a hallmark of hemophilia. Weekly SC doses of ACE910 (initial 3.97 mg/kg followed by 1 mg/kg) significantly prevented these bleeding symptoms; notably, no joint bleeding symptoms were observed. ACE910 is expected to prevent spontaneous bleeds and joint damage in hemophilia A patients even with weekly SC dosing, although appropriate clinical investigation is required.


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.


Methods of Molecular Biology | 2014

Humanization and Simultaneous Optimization of Monoclonal Antibody

Taichi Kuramochi; Tomoyuki Igawa; Hiroyuki Tsunoda; Kunihiro Hattori

Antibody humanization is an essential technology for reducing the potential risk of immunogenicity associated with animal-derived antibodies and has been applied to a majority of the therapeutic antibodies on the market. For developing an antibody molecule as a pharmaceutical at the current biotechnology level, however, other properties also have to be considered in parallel with humanization in antibody generation and optimization. This section describes the critical properties of therapeutic antibodies that should be sufficiently qualified, including immunogenicity, binding affinity, physiochemical stability, expression in host cells and pharmacokinetics, and the basic methodologies of antibody engineering involved. By simultaneously optimizing the antibody molecule in the light of these properties, it should prove possible to shorten the research and development period necessary to identify a highly qualified clinical candidate and consequently accelerate the start of the clinical trial.


Experimental Dermatology | 2018

Cynomolgus monkey model of interleukin-31-induced scratching depicts blockade of human interleukin-31 receptor A by a humanized monoclonal antibody

Sohei Oyama; Hidetomo Kitamura; Taichi Kuramochi; Yoshinobu Higuchi; Hiroaki Matsushita; Tsukasa Suzuki; Masaaki Goto; Hideki Adachi; Keiko Kasutani; Akihisa Sakamoto; Yuki Iwayanagi; Akihisa Kaneko; Masahiko Nanami; Etsuko Fujii; Keiko Esaki; Yoshiaki Takashima; Shin Shimaoka; Kunihiro Hattori; Yoshiki Kawabe

Scratching is an important factor exacerbating skin lesions through the so‐called itch‐scratch cycle in atopic dermatitis (AD). In mice, interleukin (IL)‐31 and its receptor IL‐31 receptor A (IL‐31RA) are known to play a critical role in pruritus and the pathogenesis of AD; however, study of their precise roles in primates is hindered by the low sequence homologies between primates and mice and the lack of direct evidence of itch sensation by IL‐31 in primates. We showed that administration of cynomolgus IL‐31 induces transient scratching behaviour in cynomolgus monkeys and by that were able to establish a monkey model of scratching. We then showed that a single subcutaneous injection of 1 mg/kg nemolizumab, a humanized anti‐human IL‐31RA monoclonal antibody that also neutralizes cynomolgus IL‐31 signalling and shows a good pharmacokinetic profile in cynomolgus monkeys, suppressed the IL‐31‐induced scratching for about 2 months. These results suggest that the IL‐31 axis and IL‐31RA axis play as critical a role in the induction of scratching in primates as in mice and that the blockade of IL‐31 signalling by an anti‐human IL‐31RA antibody is a promising therapeutic approach for treatment of AD. Nemolizumab is currently under investigation in clinical trials.


Current Pharmaceutical Biotechnology | 2016

Fc Engineering to Improve the Function of Therapeutic Antibodies

Futa Mimoto; Taichi Kuramochi; Hitoshi Katada; Tomoyuki Igawa; Kunihiro Hattori

Monoclonal antibodies are currently the most attractive therapeutic modality in a broad range of disease areas, including infectious diseases, autoimmune diseases, and oncology. Fc engineering is one attractive application to maximize the value or overcome the drawbacks of monoclonal antibodies for therapeutic use. With the Fc region, antibodies bind to several types of receptors, such as Fc gamma receptors, a complement receptor, and a neonatal Fc receptor. Through this interaction with the receptors, antibodies demonstrate unique functions, such as antibody-dependent cellular cytotoxicity, antibody- dependent cellular phagocytosis, complement dependent cytotoxicity, agonistic activity, and endosomal recycling. Fc engineering technology is conducted mainly to maximize the receptor-mediated functions of antibodies. Moreover, Fc engineering of the two heavy chains to facilitate heterodimerization is indispensable for generating IgG-like bispecific antibodies that are asymmetric. Fc engineering is also conducted to avoid the undesired properties, such as cytokine release and protease-mediated cleavage of the hinge region, of wild-type antibodies, as well as providing additional functions. Thus, Fc engineering technology is an attractive approach for maximizing the potency and convenience of therapeutic antibodies. This review will cover a variety of Fc engineering technologies that improve the functions of therapeutic antibodies.


Archive | 2008

Method of modifying isoelectric point of antibody via amino acid substitution in cdr

Tomoyuki Igawa; Hiroyuki Tsunoda; Tatsuhiko Tachibana; Taichi Kuramochi


Archive | 2008

Anti-glypican-3 antibody having improved kinetics in plasma

Tomoyuki Igawa; Taichi Kuramochi; Hirotake Shiraiwa; Hiroyuki Tsunoda; Tatsuhiko Tachibana; Takahiro Ishiguro


Archive | 2009

Anti-nr10 antibody and use thereof

Taichi Kuramochi; Keiko Kasutani; Souhei Ohyama; Hiroyuki Tsunoda; Tomoyuki Igawa; Tatsuhiko Tachibana; Hirotake Shiraiwa; Keiko Esaki

Collaboration


Dive into the Taichi Kuramochi's collaboration.

Top Co-Authors

Avatar

Tomoyuki Igawa

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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

Atsuhiko Maeda

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Keiko Esaki

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Shojiro Kadono

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Keiko Kasutani

Chugai Pharmaceutical Co.

View shared research outputs
Researchain Logo
Decentralizing Knowledge