Network


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

Hotspot


Dive into the research topics where Shun-ichiro Komatsu is active.

Publication


Featured researches published by Shun-ichiro Komatsu.


Toxicologic Pathology | 2013

Blinded by the Light Retinal Phototoxicity in the Context of Safety Studies

Maria Cristina de Vera Mudry; Sven Kronenberg; Shun-ichiro Komatsu; Gustavo D. Aguirre

In this review, we discuss concepts and mechanisms of light damage and indicate how these might relate to preclinical safety studies. As well, we provide recommendations for evaluating the retina and developing protocols for evaluating a potential light damage mechanism in studies where retinal degeneration is observed. Language: en


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.


Cancer Research | 2016

Abstract DDT01-05: First-in-class T cell-redirecting bispecific antibody targeting glypican-3: a highly tumor-selective antigen

Takahiro Ishiguro; Yasuko Kinoshita; Yuji Sano; Yumiko Azuma; Toshiaki Tsunenari; Natsuki Ono; Yoko Kayukawa; Mika Kamata-Sakurai; Hirotake Shiraiwa; Akihisa Kaneko; Werner Frings; Shun-ichiro Komatsu; Jun-ichi Nezu; Mika Endo

Immune checkpoint inhibitors such as anti-PD1 antibodies have shown promising clinical responses in several solid tumors, however there remain patients who do not show an adequate response. Recent biomarker studies have revealed that the presence of neoantigens in the tumor can determine the level of response, and thus the next challenge will be how to target tumors with a neoantigen level that is too low to be recognized by endogenous cytotoxic T cells. Hope in this area is offered by a T cell-redirecting antibody (TRAB), which bispecifically engages CD3 and a tumor antigen, even at very low expression levels, to activate the inherent cytolytic potential of T cells against target tumor cells. A TRAB is highly potent because T cells are activated only in the presence of the targeted antigens and are not restricted by the specificity of the T cell receptor. Given this very potent cytotoxicity, the key to successfully achieving strong antitumor efficacy while avoiding on-target off-tumor toxicity is to select a highly tumor-selective antigen. Our fully humanized IgG TRAB recognizes CD3 and a highly tumor-selective antigen, glypican-3 (GPC3), which is a fetal protein expressed in a wide variety of tissues during development but suppressed in most adult tissues. On the other hand, an inct101e in GPC3 expression has been reported in hepatocellular carcinoma, gastric cancer, lung squamous cell carcinoma, and other cancers. In nonclinical in vitro pharmacology studies, the anti-GPC3 TRAB elicited activation and proliferation of T cells and T cell-dependent cellular cytotoxicity against a wide variety of GPC3-expressing tumor cells, and showed long-lasting in vivo efficacy against tumor expressing very low levels of GPC3 at a few thousand molecules per cell. Furthermore, in an immunocompetent mouse model using human CD3 transgenic mice, anti-GPC3 TRAB showed strong antitumor efficacy against poorly immunogenic tumors, whereas both the immune checkpoint inhibitors and a conventional ADCC-inducing antibody recognizing GPC3 did not show significant efficacy. Pharmacokinetics and toxicology studies in nonhuman primates showed a plasma half-life comparable to a standard IgG drug, allowing a QW or Q2W regimen in humans, with toxicity which was manageable and reversible; the main observations of transient cytokine elevation and associated clinical symptoms were markedly reduced by steroid premedication. Our anti-GPC3 TRAB, which is supported by proprietary antibody engineering technology (ART-Ig) that enables large-scale GMP manufacturing, has promise as a new approach in cancer immunotherapy. Citation Format: Takahiro Ishiguro, Yasuko Kinoshita, Yuji Sano, Yumiko Azuma, Toshiaki Tsunenari, Natsuki Ono, Yoko Kayukawa, Mika Kamata-Sakurai, Hirotake Shiraiwa, Akihisa Kaneko, Werner Frings, Shunichiro Komatsu, Junichi Nezu, Mika Endo. First-in-class T cell-redirecting bispecific antibody targeting glypican-3: a highly tumor-selective antigen. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr DDT01-05.


Journal of Toxicologic Pathology | 2012

Stimulation of adrenal chromaffin cell proliferation by hypercalcemia induced by intravenous infusion of calcium gluconate in rats.

Kaori Isobe; Tsuneo Ito; Shun-ichiro Komatsu; Kentaro Asanuma; Etsuko Fujii; Chie Kato; Kenji Adachi; Atsuhiko Kato; Tetsuro Sugimoto; Masami Suzuki

Increased incidence of adrenal pheochromocytoma is frequently encountered in rat carcinogenicity studies. In some of the studies, the finding is judged to be due to a rat-specific mechanism of carcinogenesis caused by a disturbance of calcium homeostasis. However, direct evidence that the proliferation of chromaffin cells in the adrenal medulla is induced solely by hypercalcemia is not available. In this study, calcium gluconate was intravenously infused for 7 days to rat chromaffin cells by a tail cuff method, and cumulative labeling with bromodeoxyuridine (BrdU) was carried out to evaluate the proliferative activity. The serum calcium concentration was dose-dependently increased, and a high calcium concentration was stably sustained from day 2 to 7. In the adrenal medulla, BrdU-positive chromaffin cells increased in the calcium gluconate-treated animals, and the BrdU-labeling index increased in a dose-dependent manner. In addition, an increased BrdU-labeling index of chromaffin cells was shown to correlate with the serum calcium concentration. Our results demonstrate that hypercalcemia directly enhances the proliferative activity of chromaffin cells and that the proliferative activity is correlated with the serum calcium concentration.


Journal of Toxicological Sciences | 2006

TOTAL PARENTERAL NUTRITION USING CONTINUOUS INTRAVENOUS INFUSION VIA THE POSTERIOR VENA CAVA IN RATS

Kentaro Asanuma; Shun-ichiro Komatsu; Takayuki Sakurai; Ryo Takai; Shuichi Chiba


Journal of Toxicological Sciences | 2009

Application of an indwelling vascular access port for intravenous administration in a repeated and intermittent dose toxicity study in rats.

Junko Taketoh; Shun-ichiro Komatsu; Kenji Adachi; Ryo Takai; Kentaro Asanuma


Journal of Clinical Oncology | 2016

A phase I dose escalation and cohort expansion study of T-cell redirecting bispecific antibody against Glypican 3 in patients with advanced solid tumors.

Kenji Hashimoto; Ayesh Perera; Yoshitaka Ogita; Mikiko Nakamura; Takahiro Ishiguro; Yuji Sano; Yasuko Kinoshita; Mika Kamata Sakurai; Werner Frings; Shun-ichiro Komatsu; Akihisa Kaneko; Masamichi Ueda; Shohei Kishishita; Athos Gianella-Borradori


Journal of Toxicological Sciences | 2006

CONTRIBUTION OF INTESTINAL CALCIUM ABSORPTION TO 1,25-DIHYDROXYVITAMIN D3-INDUCED CALCIUM ACTION IN THE TOTAL PARENTERAL NUTRITION RAT

Kentaro Asanuma; Shun-ichiro Komatsu; Takayuki Sakurai; Ryo Takai; Shuichi Chiba


Diabetes | 2018

OWL833, an Orally Active Nonpeptide GLP-1 Receptor Agonist, Improves Glucose Tolerance by Increasing Insulin Secretion and Reduces Food Intake of Cynomolgus Monkeys

Takahiro Kawai; Fumihito Tanino; Masanori Fukazawa; Kotaro Ogawa; Shunsuke Nagao; Hitoshi Yoshino; Shun-ichiro Komatsu; Yoshiyuki Suzuki; Yoshiki Kawabe


Journal of Clinical Oncology | 2017

A phase 1 dose escalation (DE) and cohort expansion (CE) study of ERY974, an anti-Glypican 3 (GPC3)/CD3 bispecific antibody, in patients with advanced solid tumors.

Kenji Hashimoto; Ayesh Perera; Naofumi Sugaya; Yoshitaka Ogita; Mikiko Nakamura; Sheila Rossi; Takahiro Ishiguro; Yuji Sano; Sumire Shimada; Werner Frings; Shun-ichiro Komatsu; Akihisa Kaneko; Masamichi Ueda; Junnosuke Matsushima; Shohei Kishishita; Athos Gianella-Borradori; Asco Ery

Collaboration


Dive into the Shun-ichiro Komatsu's collaboration.

Top Co-Authors

Avatar

Kentaro Asanuma

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Shuichi Chiba

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Akihisa Kaneko

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Ryo Takai

Rakuno Gakuen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Werner Frings

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Yuji Sano

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge