Network


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

Hotspot


Dive into the research topics where Katsuhiro Mori is active.

Publication


Featured researches published by Katsuhiro Mori.


BMC Biotechnology | 2007

Double knockdown of α1,6-fucosyltransferase (FUT8) and GDP-mannose 4,6-dehydratase (GMD) in antibody-producing cells: a new strategy for generating fully non-fucosylated therapeutic antibodies with enhanced ADCC

Harue Imai-Nishiya; Katsuhiro Mori; Miho Inoue; Masako Wakitani; Shigeru Iida; Kenya Shitara; Mitsuo Satoh

BackgroundAntibody-dependent cellular cytotoxicity (ADCC) is greatly enhanced by the absence of the core fucose of oligosaccharides attached to the Fc, and is closely related to the clinical efficacy of anticancer activity in humans in vivo. Unfortunately, all licensed therapeutic antibodies and almost all currently-developed therapeutic antibodies are heavily fucosylated and fail to optimize ADCC, which leads to a large dose requirement at a very high cost for the administration of antibody therapy to cancer patients. In this study, we explored the possibility of converting already-established antibody-producing cells to cells that produce antibodies fully lacking core fucosylation in order to facilitate the rapid development of next-generation therapeutic antibodies.ResultsFirstly, loss-of-function analyses using small interfering RNAs (siRNAs) against the three key genes involved in oligosaccharide fucose modification, i.e. α1,6-fucosyltransferase (FUT8), GDP-mannose 4,6-dehydratase (GMD), and GDP-fucose transporter (GFT), revealed that single-gene knockdown of each target was insufficient to completely defucosylate the products in antibody-producing cells, even though the most effective siRNA (>90% depression of the target mRNA) was employed. Interestingly, beyond our expectations, synergistic effects of FUT8 and GMD siRNAs on the reduction in fucosylation were observed, but not when these were used in combination with GFT siRNA. Secondly, we successfully developed an effective short hairpin siRNA tandem expression vector that facilitated the double knockdown of FUT8 and GMD, and we converted antibody-producing Chinese hamster ovary (CHO) cells to fully non-fucosylated antibody producers within two months, and with high converting frequency. Finally, the stable manufacture of fully non-fucosylated antibodies with enhanced ADCC was confirmed using the converted cells in serum-free fed-batch culture.ConclusionOur results suggest that FUT8 and GMD collaborate synergistically in the process of intracellular oligosaccharide fucosylation. We also demonstrated that double knockdown of FUT8 and GMD in antibody-producing cells could serve as a new strategy for producing next-generation therapeutic antibodies fully lacking core fucosylation and with enhanced ADCC. This approach offers tremendous cost- and time-sparing advantages for the development of next-generation therapeutic antibodies.


Cytotechnology | 2007

Non-fucosylated therapeutic antibodies: the next generation of therapeutic antibodies

Katsuhiro Mori; Shigeru Iida; Naoko Yamane-Ohnuki; Yutaka Kanda; Reiko Kuni-Kamochi; Ryosuke Nakano; Harue Imai-Nishiya; Akira Okazaki; Toyohide Shinkawa; Akihito Natsume; Rinpei Niwa; Kenya Shitara; Mitsuo Satoh

Therapeutic antibody IgG1 has two N-linked oligosaccharide chains bound to the Fc region. The oligosaccharides are of the complex biantennary type, composed of a trimannosyl core structure with the presence or absence of core fucose, bisecting N-acetylglucosamine (GlcNAc), galactose, and terminal sialic acid, which gives rise to structural heterogeneity. Both human serum IgG and therapeutic antibodies are well known to be heavily fucosylated. Recently, antibody-dependent cellular cytotoxicity (ADCC), a lytic attack on antibody-targeted cells, has been found to be one of the critical effector functions responsible for the clinical efficacy of therapeutic antibodies such as anti-CD20 IgG1 rituximab (Rituxan®) and anti-Her2/neu IgG1 trastuzumab (Herceptin®). ADCC is triggered upon the binding of lymphocyte receptors (FcγRs) to the antibody Fc region. The activity is dependent on the amount of fucose attached to the innermost GlcNAc of N-linked Fc oligosaccharide via an α-1,6-linkage, and is dramatically enhanced by a reduction in fucose. Non-fucosylated therapeutic antibodies show more potent efficacy than their fucosylated counterparts both in vitro and in vivo, and are not likely to be immunogenic because their carbohydrate structures are a normal component of natural human serum IgG. Thus, the application of non-fucosylated antibodies is expected to be a powerful and elegant approach to the design of the next generation therapeutic antibodies with improved efficacy. In this review, we discuss the importance of the oligosaccharides attached to the Fc region of therapeutic antibodies, especially regarding the inhibitory effect of fucosylated therapeutic antibodies on the efficacy of non-fucosylated counterparts in one medical agent. The impact of completely non-fucosylated therapeutic antibodies on therapeutic fields will be also discussed.


BMC Cancer | 2009

Two mechanisms of the enhanced antibody-dependent cellular cytotoxicity (ADCC) efficacy of non-fucosylated therapeutic antibodies in human blood

Shigeru Iida; Reiko Kuni-Kamochi; Katsuhiro Mori; Hirofumi Misaka; Miho Inoue; Akira Okazaki; Kenya Shitara; Mitsuo Satoh

BackgroundAntibody-dependent cellular cytotoxicity (ADCC) has recently been identified as one of the critical mechanisms underlying the clinical efficacy of therapeutic antibodies, especially anticancer antibodies. Therapeutic antibodies fully lacking the core fucose of the Fc oligosaccharides have been found to exhibit much higher ADCC in humans than their fucosylated counterparts. However, data which show how fully non-fucosylated antibodies achieve such a high ADCC in human whole blood have not yet been disclosed. The precise mechanisms responsible for the high ADCC mediated by fully non-fucosylated therapeutic antibodies, even in the presence of human plasma, should be explained based on direct evidence of non-fucosylated antibody action in human blood.MethodsUsing a human ex vivo B-cell depletion assay with non-fucosylated and fucosylated anti-CD20 IgG1s rituximab, we monitored the binding of the therapeutic agents both to antigens on target cells (target side interaction) and to leukocyte receptors (FcγR) on effector cells (effector side interaction), comparing the intensities of ADCC in human blood.ResultsIn the target side interaction, down-modulation of CD20 on B cells mediated by anti-CD20 was not observed. Simple competition for binding to the antigens on target B cells between fucosylated and non-fucosylated anti-CD20s was detected in human blood to cause inhibition of the enhanced ADCC of non-fucosylated anti-CD20 by fucosylated anti-CD20. In the effector side interaction, non-fucosylated anti-CD20 showed sufficiently high FcγRIIIa binding activity to overcome competition from plasma IgG for binding to FcγRIIIa on natural killer (NK) cells, whereas the binding of fucosylated anti-CD20 to FcγRIIIa was almost abolished in the presence of human plasma and failed to recruit NK cells effectively. The core fucosylation levels of individual serum IgG1 from healthy donors was found to be so slightly different that it did not affect the inhibitory effect on the ADCC of fucosylated anti-CD20.ConclusionOur results demonstrate that removal of fucosylated antibody ingredients from antibody therapeutics elicits high ADCC in human blood by two mechanisms: namely, by evading the inhibitory effects both of plasma IgG on FcγRIIIa binding (effector side interaction) and of fucosylated antibodies on antigen binding (target side interaction).


Experimental Hematology | 2009

Nonfucosylated rituximab potentiates human neutrophil phagocytosis through its high binding for FcγRIIIb and MHC class II expression on the phagocytotic neutrophils

Mami Shibata-Koyama; Shigeru Iida; Hirofumi Misaka; Katsuhiro Mori; Keiichi Yano; Kenya Shitara; Mitsuo Satoh

OBJECTIVE Antibody-dependent cellular cytotoxicity mediated by natural killer cells via leukocyte receptor IIIa (FcgammaRIIIa) is greatly enhanced by the absence of the core fucose of Fc oligosaccharides, and is closely related to the clinical efficacy of anticancer processes in humans in vivo. Here, we focused on the physiological functions of nonfucosylated anti-CD20 IgG1 rituximab, in particular those functions mediated by human neutrophils, which highly express FcgammaRIIIb, a highly homologous FcgammaR to FcgammaRIIIa. MATERIALS AND METHODS After treatment with anti-CD20, the response of neutrophils to fluorescently labeled CD20(+) B-cell lymphoma in human whole blood was quantitatively analyzed by measuring their activities of antibody-dependent phagocytosis and major histocompatibility complex (MHC) class II expression on the phagocytotic neutrophils using flow cytometry. RESULTS In human whole blood, most of the added CD20(+) B-cell lymphoma died shortly, within 4 hours, irrespective of the presence or absence of anti-CD20. Neutrophils were not directly concerned in the death because depletion of neutrophils from human whole blood did not affect the phenomenon. However, neutrophils aggressively phagocytosed newly dead lymphoma cells, and the nonfucosylated anti-CD20 effectively enhanced neutrophil phagocytosis solely by enhancing binding for the phagocytosis coreceptor FcgammaRIIIb. Noteworthy, more increased expression of MHC class II was also observed on the phagocytotic neutrophils than those observed on spontaneous and fucosylated anti-CD20 stimulated phagocytotic neutrophils. CONCLUSIONS Our data showed that antibody therapy composed of nonfucosylated rituximab can activate human neutrophil functions involving phagocytosis and MHC class II expression, which may favorably potentiate the adaptive immune response in cancer patients.


Glycobiology | 2008

The N-linked oligosaccharide at FcγRIIIa Asn-45: an inhibitory element for high FcγRIIIa binding affinity to IgG glycoforms lacking core fucosylation

Mami Shibata-Koyama; Shigeru Iida; Akira Okazaki; Katsuhiro Mori; Kazuko Kitajima-Miyama; Seiji Saitou; Shingo Kakita; Yutaka Kanda; Kenya Shitara; Koichi Kato; Mitsuo Satoh

Human leukocyte receptor IIIa (FcγRIIIa) plays an important role in mediating therapeutic antibodies’ antibody-dependent cellular cytotoxicity (ADCC), which is closely related to the clinical efficacy of anticancer processes in humans in vivo. The removal of the core fucose from oligosaccharides attached to the Fc region of antibodies improves FcγRIIIa binding, allowing the antibodies to enhance dramatically the antibody effector functions of ADCC. In this study, the contribution of FcγRIIIa oligosaccharides to the strength of the FcγRIIIa/antibody complex was analyzed using a serial set of soluble human recombinant FcγRIIIa lacking the oligosaccharides. A nonfucosylated antibody IgG1 appeared to have a significantly higher affinity to the wild-type FcγRIIIa fully glycosylated at its five N-linked oligosaccharide sites than did the fucosylated IgG1, and this increased binding was almost abolished once all of the FcγRIIIa glycosylation was removed. Our gain-of-function analysis in the FcγRIIIa oligosaccharide at Asn-162 (N-162) confirmed that N-162 is the element required for the high binding affinity to nonfucosylated antibodies, as previously revealed by loss-of-function analyses. Interestingly, beyond our expectation, the FcγRIIIa modified by N-162 alone showed a significantly higher binding affinity to nonfucosylated IgG1 than did the wild-type FcγRIIIa. Attachment of the other four oligosaccharides, especially the FcγRIIIa oligosaccharide at Asn-45 (N-45), hindered the high binding affinity of FcγRIIIa to nonfucosylated IgG1. Our data clearly demonstrated that N-45 is an inhibitory element for the high FcγRIIIa binding affinity mediated by N-162 to nonfucosylated antibodies. This information can be exploited for the structural-based functional study of FcγRIIIa.


Archive | 2006

Generation of an industrially ideal host cell line for producing completely-defucosylated antibody with enhanced antibody-dependent cellular cytotoxicity (ADCC)

Mitsuo Satoh; Naoko Yamane-Ohnuki; Katsuhiro Mori; Ripei Niwa; Toyohide Shinkawa; Harue Imai; Reiko Kuni-Kamochi; Ryosuke Nakano; Kazuya Yamano; Yutaka Kanda; Shigeru Iida; Kazuhisa Uchida; Kenya Shitara

To generate industrially applicable new host cell lines for antibody production with optimizing antibody-dependent cellular cytotoxicity (ADCC) we focused on the most important carbohydrate structure “fucose residues attached to the innermost GlcNAc residue of N-linked oligosaccharides via -1,6 linkage” (Shields, 2002; Shinkawa, 2003), and succeeded in disrupting both FUT8 ( -1,6-fucosyltransferase gene) alleles in Chinese hamster ovary (CHO) cell line by sequential homologous recombination. FUT8-/cell lines have morphology and growth kinetics similar to those of the parent. Antibodies produced by the engineered CHO cell lines strongly bound to human Fc receptor IIIa (Fc RIIIa) and showed approximately two orders of magnitude higher ADCC than antiCD20 antibodies (Rituxan) produced by parental cell lines without changing antigen-binding and complement-dependent cytotoxicity (CDC). Moreover, the engineered cell line remains stable, producing completelydefucosylated antibody with fixed quality and efficacy even in serum-free fed-batch culture. Thus, our approaches provide a new strategy for controlling the glycosylation profile of therapeutic recombinant proteins and could be a considerable advantage for the manufacture of glycoprotein therapeutics, especially antibodies.


Archive | 2003

Cells of which genome is modified

Naoko Yamane; Mitsuo Satoh; Katsuhiro Mori; Kazuya Yamano


Archive | 2003

Production process for antibody composition

Mitsuo Satoh; Reiko Kamachi; Yutaka Kanda; Katsuhiro Mori; Kazuya Yamano; Satoko Kinoshita; Shigeru Iida


Archive | 2003

Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost

Yutaka Kanda; Mitsuo Satoh; Katsuhiro Mori


Archive | 2004

Process for producing antibody composition by using rna inhibiting the function of alpha1,6-fucosyltransferase

Harue Nishiya; Mitsuo Satoh; Katsuhiro Mori

Collaboration


Dive into the Katsuhiro Mori's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yutaka Kanda

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yutaka Kanda

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge