Network


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

Hotspot


Dive into the research topics where Akihisa Sakamoto is active.

Publication


Featured researches published by Akihisa Sakamoto.


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.


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.


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.


Science Translational Medicine | 2017

An anti–glypican 3/CD3 bispecific T cell–redirecting antibody for treatment of solid tumors

Takahiro Ishiguro; Yuji Sano; Shun-ichiro Komatsu; Mika Kamata-Sakurai; Akihisa Kaneko; Yasuko Kinoshita; Hirotake Shiraiwa; Yumiko Azuma; Toshiaki Tsunenari; Yoko Kayukawa; Yukiko Sonobe; Natsuki Ono; Kiyoaki Sakata; Toshihiko Fujii; Yoko Miyazaki; Mizuho Noguchi; Mika Endo; Asako Harada; Werner Frings; Etsuko Fujii; Eitaro Nanba; Atsushi Narita; Akihisa Sakamoto; Tetsuya Wakabayashi; Hiroko Konishi; Hiroaki Segawa; Tomoyuki Igawa; Takashi Tsushima; Hironori Mutoh; Yukari Nishito

An anti–glypican 3/CD3 bispecific T cell–redirecting antibody (ERY974) is a promising therapeutic agent for solid tumors. Double trouble for solid tumors Because the endogenous immune response is not enough to clear a patient’s cancer, therapies are being designed to redirect T cells to tumor cells. This can be done by engineering the cells ex vivo, such as in CAR T cell therapy, or in vivo, such as with bispecific antibodies. Ishiguro et al. describe the development and preclinical testing of a bispecific antibody recognizing CD3 and glypican 3, a common antigen on solid tumors. This bispecific antibody was effective in a variety of mouse cancer models, even when treatment was initiated after the tumor was quite large. Treatment also appeared to be safe when administered to monkeys. These results suggest further development of this antibody for therapeutic use in multiple cancer types. Cancer care is being revolutionized by immunotherapies such as immune checkpoint inhibitors, engineered T cell transfer, and cell vaccines. The bispecific T cell–redirecting antibody (TRAB) is one such promising immunotherapy, which can redirect T cells to tumor cells by engaging CD3 on a T cell and an antigen on a tumor cell. Because T cells can be redirected to tumor cells regardless of the specificity of T cell receptors, TRAB is considered efficacious for less immunogenic tumors lacking enough neoantigens. Its clinical efficacy has been exemplified by blinatumomab, a bispecific T cell engager targeting CD19 and CD3, which has shown marked clinical responses against hematological malignancies. However, the success of TRAB in solid tumors has been hampered by the lack of a target molecule with sufficient tumor selectivity to avoid “on-target off-tumor” toxicity. Glypican 3 (GPC3) is a highly tumor-specific antigen that is expressed during fetal development but is strictly suppressed in normal adult tissues. We developed ERY974, a whole humanized immunoglobulin G–structured TRAB harboring a common light chain, which bispecifically binds to GPC3 and CD3. Using a mouse model with reconstituted human immune cells, we revealed that ERY974 is highly effective in killing various types of tumors that have GPC3 expression comparable to that in clinical tumors. ERY974 also induced a robust antitumor efficacy even against tumors with nonimmunogenic features, which are difficult to treat by inhibiting immune checkpoints such as PD-1 (programmed cell death protein–1) and CTLA-4 (cytotoxic T lymphocyte–associated protein–4). Immune monitoring revealed that ERY974 converted the poorly inflamed tumor microenvironment to a highly inflamed microenvironment. Toxicology studies in cynomolgus monkeys showed transient cytokine elevation, but this was manageable and reversible. No organ toxicity was evident. These data provide a rationale for clinical testing of ERY974 for the treatment of patients with GPC3-positive solid tumors.


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.


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 | 2011

Cytotoxicity-inducing therapeutic agent

Jun-ichi Nezu; Takahiro Ishiguro; Atsushi Narita; Akihisa Sakamoto; Yumiko Kawai; Tomoyuki Igawa; Taichi Kuramochi


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


Biochemical and Biophysical Research Communications | 2005

Structure-based design of P3 moieties in the peptide mimetic factor VIIa inhibitor

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


Biochemical and Biophysical Research Communications | 2005

Novel interactions of large P3 moiety and small P4 moiety in the binding of the peptide mimetic factor VIIa inhibitor

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

Collaboration


Dive into the Akihisa Sakamoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hirofumi Kodama

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Masateru Ohta

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shojiro Kadono

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Takaki Koga

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshiro Kozono

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge