Network


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

Hotspot


Dive into the research topics where Kiyotaka Nakano is active.

Publication


Featured researches published by Kiyotaka Nakano.


Cancer Research | 2004

Identification of Soluble NH2-Terminal Fragment of Glypican-3 as a Serological Marker for Early-Stage Hepatocellular Carcinoma

Yoshitaka Hippo; Kiyotaka Watanabe; Akira Watanabe; Yutaka Midorikawa; Shogo Yamamoto; Sigeo Ihara; Susumu Tokita; Hiroko Iwanari; Yukio Ito; Kiyotaka Nakano; Jun-ichi Nezu; Hiroyuki Tsunoda; Takeshi Yoshino; Iwao Ohizumi; Masayuki Tsuchiya; Shin Ohnishi; Masatoshi Makuuchi; Takao Hamakubo; Tatsuhiko Kodama; Hiroyuki Aburatani

For detection of hepatocellular carcinoma (HCC) in patients with liver cirrhosis, serum α-fetoprotein has been widely used, but its sensitivity has not been satisfactory, especially in small, well-differentiated HCC, and complementary serum marker has been clinically required. Glypican-3 (GPC3), a heparan sulfate proteoglycan anchored to the plasma membrane, is a good candidate marker of HCC because it is an oncofetal protein overexpressed in HCC at both the mRNA and protein levels. In this study, we demonstrated that its NH2-terminal portion [soluble GPC3 (sGPC3)] is cleaved between Arg358 and Ser359 of GPC3 and that sGPC3 can be specifically detected in the sera of patients with HCC. Serum levels of sGPC3 were 4.84 ± 8.91 ng/ml in HCC, significantly higher than the levels seen in liver cirrhosis (1.09 ± 0.74 ng/ml; P < 0.01) and healthy controls (0.65 ± 0.32 ng/ml; P < 0.001). In well- or moderately-differentiated HCC, sGPC3 was superior to α-fetoprotein in sensitivity, and a combination measurement of both markers improved overall sensitivity from 50% to 72%. These results indicate that sGPC3 is a novel serological marker essential for the early detection of HCC.


Cancer Research | 2008

Anti–Glypican 3 Antibody as a Potential Antitumor Agent for Human Liver Cancer

Takahiro Ishiguro; Masamichi Sugimoto; Yasuko Kinoshita; Yoko Miyazaki; Kiyotaka Nakano; Hiroyuki Tsunoda; Izumi Sugo; Iwao Ohizumi; Hiroyuki Aburatani; Takao Hamakubo; Tatsuhiko Kodama; Masayuki Tsuchiya; Hisafumi Yamada-Okabe

Human glypican 3 (GPC3) is preferentially expressed in the tumor tissues of liver cancer patients. In this study, we obtained a monoclonal antibody (mAb) against the COOH-terminal part of GPC3, which induced antibody-dependent cellular cytotoxicity (ADCC). The mAb, designated GC33, exhibited marked tumor growth inhibition of s.c. transplanted Hep G2 and HuH-7 xenografts that expressed GPC3 but did not inhibit growth of the SK-HEP-1 that was negative for GPC3. GC33 was efficacious even in an orthotopic model; it markedly reduced the blood alpha-fetoprotein levels of mice intrahepatically transplanted with Hep G2 cells. Humanized GC33 (hGC33) was as efficacious as GC33 against the Hep G2 xenograft, but hGC33 lacking carbohydrate moieties caused neither ADCC nor tumor growth inhibition. Depletion of CD56+ cells from human peripheral blood mononuclear cells markedly abrogated the ADCC caused by hGC33. The results show that the antitumor activity of hGC33 is mainly attributable to ADCC, and in human, natural killer cell-mediated ADCC is one possible mechanism of the antitumor effects by GC33. hGC33 will provide a novel treatment option for liver cancer patients with GPC3-positive tumors.


Biochemical and Biophysical Research Communications | 2009

Anti-glypican 3 antibodies cause ADCC against human hepatocellular carcinoma cells

Kiyotaka Nakano; Tetsuro Orita; Jun-ichi Nezu; Takeshi Yoshino; Iwao Ohizumi; Masamichi Sugimoto; Koh Furugaki; Yasuko Kinoshita; Takahiro Ishiguro; Takao Hamakubo; Tatsuhiko Kodama; Hiroyuki Aburatani; Hisafumi Yamada-Okabe; Masayuki Tsuchiya

Glypican 3 (GPC3), a GPI-anchored heparan sulfate proteoglycan, is expressed in the majority of hepatocellular carcinoma (HCC) tissues. Using MRL/lpr mice, we successfully generated a series of anti-GPC3 monoclonal antibodies (mAbs). GPC3 was partially cleaved between Arg358 and Ser359, generating a C-terminal 30-kDa fragment and an N-terminal 40-kDa fragment. All mAbs that induced antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC) against cells expressing GPC3 recognized the 30-kDa fragment, indicating that the C-terminal region of GPC3 serves as an epitope for mAb with ADCC and/or CDC inducing activities. Chimeric mAbs with Fc replaced by human IgG1 were created from GC33, one of the mAbs that reacted with the C-terminal 30-kDa fragment. Chimeric GC33 induced not only ADCC against GPC3-positive human HCC cells but also was efficacious against the Huh-7 human HCC xenograft. Thus, mAbs against the C-terminal 30-kDa fragment such as GC33 are useful in therapy targeting HCC.


Anti-Cancer Drugs | 2010

Generation of a humanized anti-glypican 3 antibody by CDR grafting and stability optimization.

Kiyotaka Nakano; Takahiro Ishiguro; Hiroko Konishi; Megumi Tanaka; Masamichi Sugimoto; Izumi Sugo; Tomoyuki Igawa; Hiroyuki Tsunoda; Yasuko Kinoshita; Kiyoshi Habu; Tetsuro Orita; Masayuki Tsuchiya; Kunihiro Hattori; Hisafumi Yamada-Okabe

Glypican 3 (GPC3), a glycosylphosphatidylinositol-anchored heparan sulfate proteoglycan, is expressed in a majority of hepatocellular carcinoma tissues. The murine monoclonal antibody GC33 that specifically binds to the COOH-terminal part of GPC3 causes strong antibody-dependent cellular cytotoxicity against hepatocellular carcinoma cells and exhibits strong antitumor activity in the xenograft models. To apply GC33 for clinical use, we generated a humanized GC33 from complementarity-determining region grafting with the aid of both the hybrid variable region and two-step design methods. The humanized antibody bound to GPC3 specifically and induced antibody-dependent cellular cytotoxicity as effectively as a chimeric GC33 antibody. To improve stability of the humanized GC33, we further optimized humanized GC33 by replacing the amino acid residues that may affect the structure of the variable region of a heavy chain. Substitution of Glu6 with Gln in the heavy chain significantly improved the stability under high temperatures. GC33 also has the risk of deamidation of the -Asn–Gly- sequence in the complementarity-determining region 1 of the light chain. As substitution of Asn diminished the antigen binding, we changed the neighboring Gly to Arg to avoid deamidation. The resulting humanized anti-GPC3 antibody was as efficacious as chimeric GC33 against the HepG2 xenograft and is now being evaluated in clinical trials.


Journal of Immunological Methods | 2009

Effective screening method of agonistic diabodies based on autocrine growth

Kiyotaka Nakano; Tetsuo Kojima; Keiko Kasutani; Chiaki Senoh; Osamu Natori; Shinya Ishii; Hiroyuki Tsunoda; Kunihiro Hattori

Agonistic diabodies that mimic the function of natural ligands are expected to increase the value of therapeutic antibodies. We have developed a method that detects agonistic diabodies based on their ability to transduce growth signals through receptors, thereby permitting cytokine-independent growth of BaF/3-derived cytokine-dependent cells. Retrovirus-mediated expression of the diabody in cytokine-dependent cells was followed by selection of clones for growth in the absence of cytokine. A diabody library derived from splenocytes of human Mpl immunized mice was constructed. Infection of cells with viral particles led to the isolation of over 500 autonomously growing clones whose cultured supernatants showed agonistic activities against Mpl. Genome-integrated diabodies were cloned; representative clone AB317 showed agonistic activities as potent as a natural ligand and cross-reactive against mouse Mpl.


Oncotarget | 2018

DGC-specific RHOA mutations maintained cancer cell survival and promoted cell migration via ROCK inactivation

Takashi Nishizawa; Kiyotaka Nakano; Aya Harada; Miwako Kakiuchi; Shin-Ichi Funahashi; Masami Suzuki; Shumpei Ishikawa; Hiroyuki Aburatani

RHOA missense mutations exist specifically in diffuse type gastric cancers (DGC) and are considered one of the DGC driver genes, but it is not fully understood how RHOA mutations contribute to DGC development. Here we examined how RHOA mutations affect cancer cell survival and cell motility. We revealed that cell survival was maintained by specific mutation sites, namely G17, Y42, and L57. Because these functional mutations suppressed MLC2 phosphorylation and actin stress fiber formation, we realized they act in a dominant-negative fashion against the ROCK pathway. Through the same inactivating mechanism that maintained cell survival, RHOA mutations also increased cell migration activity. Cell survival and migration studies on CLDN18-ARHGAP (CLG) fusions, which are known to be mutually exclusive to RHOA mutations, showed that CLG fusions complemented cell survival under RHOA knockdown condition and also induced cell migration. Site-directed mutagenesis analysis revealed the importance of the GAP domain and indicated that CLG fusions maintained RHOA in the inactive form. Taken together, these findings show that the inactivation of ROCK would be a key step in DGC development, so ROCK activation might provide novel therapeutic opportunities.


Cancer Research | 2010

Abstract 2426: Anti-Glypican3 antibody for treatment of human liver cancer

Takahiro Ishiguro; Yasuko Kinoshita; Masamichi Sugimoto; Yoko Miyazaki; Atsuhiko Kato; Kiyotaka Nakano; Hirotake Takai; Hiroyuki Tsunoda; Izumi Sugo; Iwao Ohizumi; Hiroyuki Aburatani; Takao Hamakubo; Tatsuhiko Kodama; Masayuki Tsuchiya; Hisafumi Okabe

Glypican-3 (GPC3) is a member of the glypican family of heparan sulfate proteoglycans, which are linked to the cell surface through a glycosyl phosphatidyl inositol anchor. GPC3 has been reported to be highly expressed in the majority (70-100%) of HCC, and considered to play a role in the tumorigenesis of HCC. Although the molecular mechanism by which GPC3 functions in tumorigenesis has not been fully elucidated, the high prevalence in HCC has led to considerable interest in GPC3 as a diagnostic marker and therapeutic target. In this study, we obtained a monoclonal antibody (mAb) against the COOH-terminal part of GPC3, which induced antibody-dependent cellular cytotoxicity (ADCC). The mAb, designated mGC33, exhibited marked tumor growth inhibition of s.c. transplanted Hep G2 and HuH-7 xenografts that expressed GPC3 but did not inhibit growth of the SK-HEP-1 that was negative for GPC3. mGC33 was efficacious even in an orthotopic model; it markedly reduced the blood alpha-fetoprotein levels of mice intrahepatically transplanted with Hep G2 cells. To develop an antibody-based immunotherapy, we generated humanized GC33 (hGC33) by complementarily determining region (CDR) grafting. hGC33 was as efficacious as mGC33 against the Hep G2 xenograft, but hGC33 lacking carbohydrate moieties caused neither ADCC nor tumor growth inhibition. Depletion of CD56+ cells from human peripheral blood mononuclear cells markedly abrogated the ADCC caused by hGC33. The results show that the antitumor activity of hGC33 is mainly attributable to ADCC, and in human, natural killer cell-mediated ADCC is one possible mechanism of the antitumor effects by GC33. We also evaluated the antitumor activity of hGC33 combined with standard chemotherapy agent sorafenib. hGC33 and sorafenib combination was more potent in inhibiting tumor growth than sorafenib alone in the s.c. transplanted Hep G2 xenograft model. Administration of sorafenib alone did not change the GPC3 expression level in xenograft tumor. These suggest that this combination regimen may be clinically useful as an anti-liver cancer therapy. In careful examination of the safety of hGC33 in nonclinical studies, specific adverse findings on GPC3 expressed tissue or organs were not observed after repeated administration. Therefore hGC33 will provide a novel treatment option for liver cancer patients with GPC3-positive tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2426.


Archive | 2005

Anti-glypican-3 antibody

Kiyotaka Nakano; Takeshi Yoshino; Jun-ichi Nezu; Hiroyuki Tsunoda; Tomoyuki Igawa; Hiroko Konishi; Megumi Tanaka; Izumi Sugo; Shigeto Kawai; Takahiro Ishiguro; Yasuko Kinoshita


Blood | 2005

A novel therapeutic approach for thrombocytopenia by minibody agonist of the thrombopoietin receptor

Tetsuro Orita; Hiroyuki Tsunoda; Naohiro Yabuta; Kiyotaka Nakano; Takeshi Yoshino; Yuichi Hirata; Toshihiko Ohtomo; Jun-ichi Nezu; Hirofumi Sakumoto; Kouichiro Ono; Mikiyoshi Saito; Eiji Kumagai; Masahiko Nanami; Akihisa Kaneko; Takashi Yoshikubo; Masayuki Tsuchiya


Archive | 2004

Anti-mpl antibody

Hiroyuki Tsunoda; Kiyotaka Nakano; Tetsuro Orita; Masayuki Tsuchiya; Yuichi Hirata

Collaboration


Dive into the Kiyotaka Nakano's collaboration.

Top Co-Authors

Avatar

Iwao Ohizumi

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Hiroyuki Aburatani

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Izumi Sugo

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Jun-ichi Nezu

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Shigeto Kawai

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar

Takeshi Yoshino

Kyoto Prefectural University of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge