Network


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

Hotspot


Dive into the research topics where Kenta Haraya is active.

Publication


Featured researches published by Kenta Haraya.


Nature Biotechnology | 2010

Antibody recycling by engineered pH-dependent antigen binding improves the duration of antigen neutralization

Tomoyuki Igawa; Shinya Ishii; Tatsuhiko Tachibana; Atsuhiko Maeda; Yoshinobu Higuchi; Shin Shimaoka; Chifumi Moriyama; Tomoyuki Watanabe; Ryoko Takubo; Yoshiaki Doi; Tetsuya Wakabayashi; Akira Hayasaka; Shoujiro Chugai Seiyaku Kabushiki Kaisha Kadono; Takuya Miyazaki; Kenta Haraya; Yasuo Sekimori; Tetsuo Kojima; Yoshiaki Nabuchi; Yoshinori Aso; Yoshiki Kawabe; Kunihiro Hattori

For many antibodies, each antigen-binding site binds to only one antigen molecule during the antibodys lifetime in plasma. To increase the number of cycles of antigen binding and lysosomal degradation, we engineered tocilizumab (Actemra), an antibody against the IL-6 receptor (IL-6R), to rapidly dissociate from IL-6R within the acidic environment of the endosome (pH 6.0) while maintaining its binding affinity to IL-6R in plasma (pH 7.4). Studies using normal mice and mice expressing human IL-6R suggested that this pH-dependent IL-6R dissociation within the acidic environment of the endosome resulted in lysosomal degradation of the previously bound IL-6R while releasing the free antibody back to the plasma to bind another IL-6R molecule. In cynomolgus monkeys, an antibody with pH-dependent antigen binding, but not an affinity-matured variant, significantly improved the pharmacokinetics and duration of C-reactive protein inhibition. Engineering pH dependency into the interactions of therapeutic antibodies with their targets may enable them to be delivered less frequently or at lower doses.


PLOS ONE | 2013

Identification and Multidimensional Optimization of an Asymmetric Bispecific IgG Antibody Mimicking the Function of Factor VIII Cofactor Activity

Zenjiro Sampei; Tomoyuki Igawa; Tetsuhiro Soeda; Yukiko Okuyama-Nishida; Chifumi Moriyama; Tetsuya Wakabayashi; Eriko Tanaka; Atsushi Muto; Tetsuo Kojima; Takehisa Kitazawa; Kazutaka Yoshihashi; Aya Harada; Miho Funaki; Kenta Haraya; Tatsuhiko Tachibana; Sachiyo Suzuki; Keiko Esaki; Yoshiaki Nabuchi; Kunihiro Hattori

In hemophilia A, routine prophylaxis with exogenous factor VIII (FVIII) requires frequent intravenous injections and can lead to the development of anti-FVIII alloantibodies (FVIII inhibitors). To overcome these drawbacks, we screened asymmetric bispecific IgG antibodies to factor IXa (FIXa) and factor X (FX), mimicking the FVIII cofactor function. Since the therapeutic potential of the lead bispecific antibody was marginal, FVIII-mimetic activity was improved by modifying its binding properties to FIXa and FX, and the pharmacokinetics was improved by engineering the charge properties of the variable region. Difficulties in manufacturing the bispecific antibody were overcome by identifying a common light chain for the anti-FIXa and anti-FX heavy chains through framework/complementarity determining region shuffling, and by pI engineering of the two heavy chains to facilitate ion exchange chromatographic purification of the bispecific antibody from the mixture of byproducts. Engineering to overcome low solubility and deamidation was also performed. The multidimensionally optimized bispecific antibody hBS910 exhibited potent FVIII-mimetic activity in human FVIII-deficient plasma, and had a half-life of 3 weeks and high subcutaneous bioavailability in cynomolgus monkeys. Importantly, the activity of hBS910 was not affected by FVIII inhibitors, while anti-hBS910 antibodies did not inhibit FVIII activity, allowing the use of hBS910 without considering the development or presence of FVIII inhibitors. Furthermore, hBS910 could be purified on a large manufacturing scale and formulated into a subcutaneously injectable liquid formulation for clinical use. These features of hBS910 enable routine prophylaxis by subcutaneous delivery at a long dosing interval without considering the development or presence of FVIII inhibitors. We expect that hBS910 (investigational drug name: ACE910) will provide significant benefit for severe hemophilia A patients.


Journal of Thrombosis and Haemostasis | 2014

Anti-factor IXa/X bispecific antibody (ACE910): hemostatic potency against ongoing bleeds in a hemophilia A model and the possibility of routine supplementation.

Atsushi Muto; Kazutaka Yoshihashi; Minako Takeda; Takehisa Kitazawa; Tetsuhiro Soeda; Tomoyuki Igawa; Yuichiro Sakamoto; Kenta Haraya; Yoshiki Kawabe; Midori Shima; Akira Yoshioka; Kunihiro Hattori

We previously reported that a humanized anti‐factor IXa/X bispecific antibody, hBS23, mimics the function of FVIII even in the presence of FVIII inhibitors, and has preventive hemostatic activity against bleeding in an animal model of acquired hemophilia A. After further molecular engineering of hBS23, we recently identified an improved humanized bispecific antibody, ACE910, for clinical investigation.


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.


PLOS ONE | 2013

Engineered monoclonal antibody with novel antigen-sweeping activity in vivo.

Tomoyuki Igawa; Atsuhiko Maeda; Kenta Haraya; Tatsuhiko Tachibana; Yuki Iwayanagi; Futa Mimoto; Yoshinobu Higuchi; Shinya Ishii; Shigero Tamba; Naoka Hironiwa; Kozue Nagano; Tetsuya Wakabayashi; Hiroyuki Tsunoda; Kunihiro Hattori

Monoclonal antibodies are widely used to target disease-related antigens. However, because conventional antibody binds to the antigen but cannot eliminate the antigen from plasma, and rather increases the plasma antigen concentration by reducing the clearance of the antigen, some clinically important antigens are still difficult to target with monoclonal antibodies because of the huge dosages required. While conventional antibody can only bind to the antigen, some natural endocytic receptors not only bind to the ligands but also continuously eliminate them from plasma by pH-dependent dissociation of the ligands within the acidic endosome and subsequent receptor recycling to the cell surface. Here, we demonstrate that an engineered antibody, named sweeping antibody, having both pH-dependent antigen binding (to mimic the receptor-ligand interaction) and increased binding to cell surface neonatal Fc receptor (FcRn) at neutral pH (to mimic the cell-bound form of the receptor), selectively eliminated the antigen from plasma. With this novel antigen-sweeping activity, antibody without in vitro neutralizing activity exerted in vivo efficacy by directly eliminating the antigen from plasma. Moreover, conversion of conventional antibody with in vitro neutralizing activity into sweeping antibody further potentiated the in vivo efficacy. Depending on the binding affinity to FcRn at neutral pH, sweeping antibody reduced antigen concentration 50- to 1000-fold compared to conventional antibody. Thereby, sweeping antibody antagonized excess amounts of antigen in plasma against which conventional antibody was completely ineffective, and could afford marked reduction of dosage to a level that conventional antibody can never achieve. Thus, the novel mode of action of sweeping antibody provides potential advantages over conventional antibody and may allow access to the target antigens which were previously undruggable by conventional antibody.


Immunological Reviews | 2016

Sweeping antibody as a novel therapeutic antibody modality capable of eliminating soluble antigens from circulation

Tomoyuki Igawa; Kenta Haraya; Kunihiro Hattori

Monoclonal antibodies have become a general modality in therapeutic development, and a variety of monoclonal antibodies targeting soluble antigens have been developed. However, even with infinite binding affinity to an antigen, a conventional antibody can bind to the antigen only once and results in an increase in total plasma antigen concentration in vivo. This antibody‐mediated antigen accumulation generally occurs because the clearance from circulation of an antibody–antigen complex is much slower than that of a free antigen. This limitation has recently been overcome by sweeping antibodies, which are capable of actively eliminating soluble antigens from circulation. A sweeping antibody incorporates two antibody engineering technologies: one is variable region engineering to enable the antibody to bind to an antigen in plasma and dissociate from the antigen in endosome (after which the antigen undergoes lysosomal degradation), and the other is constant region engineering to increase the cellular uptake of the antibody–antigen complex into endosome. By enhancing the elimination of soluble antigens from circulation, sweeping antibodies can therapeutically target soluble antigens that conventional antibodies cannot. This review discusses the features, engineering technologies, advantages, and applications of sweeping antibodies that target soluble antigens.


Scientific Reports | 2017

Long lasting neutralization of C5 by SKY59, a novel recycling antibody, is a potential therapy for complement-mediated diseases.

Taku Fukuzawa; Zenjiro Sampei; Kenta Haraya; Yoshinao Ruike; Meiri Shida-Kawazoe; Yuichiro Shimizu; Siok Wan Gan; Machiko Irie; Yoshinori Tsuboi; Hitoshi Tai; Tetsushi Sakiyama; Akihisa Sakamoto; Shinya Ishii; Atsuhiko Maeda; Yuki Iwayanagi; Norihito Shibahara; Mitsuko Shibuya; Genki Nakamura; Takeru Nambu; Akira Hayasaka; Futa Mimoto; Yuu Okura; Yuji Hori; Kiyoshi Habu; Manabu Wada; Takaaki Miura; Tatsuhiko Tachibana; Kiyofumi Honda; Hiroyuki Tsunoda; Takehisa Kitazawa

Dysregulation of the complement system is linked to the pathogenesis of a variety of hematological disorders. Eculizumab, an anti-complement C5 monoclonal antibody, is the current standard of care for paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). However, because of high levels of C5 in plasma, eculizumab has to be administered biweekly by intravenous infusion. By applying recycling technology through pH-dependent binding to C5, we generated a novel humanized antibody against C5, SKY59, which has long-lasting neutralization of C5. In cynomolgus monkeys, SKY59 suppressed C5 function and complement activity for a significantly longer duration compared to a conventional antibody. Furthermore, epitope mapping by X-ray crystal structure analysis showed that a histidine cluster located on C5 is crucial for the pH-dependent interaction with SKY59. This indicates that the recycling effect of SKY59 is driven by a novel mechanism of interaction with its antigen and is distinct from other known pH-dependent antibodies. Finally, SKY59 showed neutralizing effect on C5 variant p.Arg885His, while eculizumab does not inhibit complement activity in patients carrying this mutation. Collectively, these results suggest that SKY59 is a promising new anti-C5 agent for patients with PNH and other complement-mediated disorders.


Xenobiotica | 2014

Application of human FcRn transgenic mice as a pharmacokinetic screening tool of monoclonal antibody

Kenta Haraya; Tatsuhiko Tachibana; Masahiko Nanami; Masaki Ishigai

Abstract 1. For drug discovery, useful screening tools are essential to select superior candidates. Here, we evaluated the applicability of transgenic mice expressing human neonatal Fc receptor (FcRn) (hFcRn Tgm) as a pharmacokinetic screening tool of therapeutic monoclonal antibodies (mAbs) and Fc-fusion proteins that overcomes the species difference in FcRn binding. 2. Marketed 11 mAbs and 2 Fc-fusion proteins were intravenously administered to hFcRn Tgm and WT mice. The half-lives in hFcRn Tgm and WT mice were compared with those in human obtained from literature. The linear half-lives in human and monkey were also calculated by nonlinear pharmacokinetic analysis. For comparison, correlations of half-lives between monkey and human were also evaluated. 3. The half-lives of mAbs and Fc-fusion proteins after intravenous administration ranged from 1.1 to 13.2 days in hFcRn Tgm and from 1.2 to 30.3 days in WT mice. The half-lives in human correlated more closely with those in hFcRn Tgm than in WT mice and monkey. 4. Our results suggest that hFcRn Tgm are a valuable and useful tool for pharmacokinetic screening of mAbs and Fc-fusion proteins in the preclinical stage. Furthermore, we believe that hFcRn Tgm are broadly applicable to preclinical pharmacokinetic screening of mAbs-based therapeutics.


Journal of Immunology | 2015

Inhibitory FcγRIIb-Mediated Soluble Antigen Clearance from Plasma by a pH-Dependent Antigen-Binding Antibody and Its Enhancement by Fc Engineering

Yuki Iwayanagi; Tomoyuki Igawa; Atsuhiko Maeda; Kenta Haraya; Naoko A. Wada; Norihito Shibahara; Ken Ohmine; Takeru Nambu; Genki Nakamura; Futa Mimoto; Hitoshi Katada; Shunsuke Ito; Tatsuhiko Tachibana; Kou-ichi Jishage; Kunihiro Hattori

Fc engineering can modulate the Fc–FcγR interaction and thus enhance the potency of Abs that target membrane-bound Ags, but it has not been applied to Abs that target soluble Ags. In this study, we revealed a previously unknown function of inhibitory FcγRII in vivo and, using an Ab that binds to Ag pH dependently, demonstrated that the function can be exploited to target soluble Ag. Because pH-dependent Ab dissociates Ag in acidic endosome, its Ag clearance from circulation reflects the cellular uptake rate of Ag/Ab complexes. In vivo studies showed that FcγR but not neonatal FcR contributes to Ag clearance by the pH-dependent Ab, and when Fc binding to mouse FcγRII and III was increased, Ag clearance was markedly accelerated in wild-type mice and FcR γ-chain knockout mice, but the effect was diminished in FcγRII knockout mice. This demonstrates that mouse FcγRII efficiently promotes Ab uptake into the cell and its subsequent recycling back to the cell surface. Furthermore, when a human IgG1 Fc variant with selectively increased binding to human FcγRIIb was tested in human FcγRIIb transgenic mice, Ag clearance was accelerated without compromising the Ab half-life. Taken together, inhibitory FcγRIIb was found to play a prominent role in the cellular uptake of monomeric Ag/Ab immune complexes in vivo, and when the Fc of a pH-dependent Ab was engineered to selectively enhance human FcγRIIb binding, the Ab could accelerate soluble Ag clearance from circulation. We assume such a function would enhance the therapeutic potency of Abs that target soluble Ags.

Collaboration


Dive into the Kenta Haraya's collaboration.

Top Co-Authors

Avatar

Tomoyuki Igawa

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Atsuhiko Maeda

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Yuki Iwayanagi

Chugai Pharmaceutical Co.

View shared research outputs
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

Shojiro Kadono

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Atsushi Muto

Chugai Pharmaceutical Co.

View shared research outputs
Researchain Logo
Decentralizing Knowledge