Network


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

Hotspot


Dive into the research topics where Tomofumi Kurokawa is active.

Publication


Featured researches published by Tomofumi Kurokawa.


Journal of Controlled Release | 2003

Sustained release of human growth hormone from microcapsules prepared by a solvent evaporation technique.

Shigeyuki Takada; Yutaka Yamagata; Masafumi Misaki; Keiko Taira; Tomofumi Kurokawa

Biodegradable microcapsules for sustained release of recombinant human growth hormone (rhGH) were prepared by a solid-in-oil-in-water (S/O/W) emulsion solvent evaporation technique using lyophilized protein microparticles. The minimum mean particle size of rhGH in S/O dispersions was 2.8-3.0 microm when ammonium acetate was added at molar ratios of 10-20 times against rhGH. High entrapment of rhGH in microcapsules was achieved by incorporating rhGH powder with a smaller particle size obtained by lyophilizing with ammonium acetate. As the particle size of rhGH decreased, the in vivo initial release decreased, while subsequent serum levels of rhGH in sustained release phase were higher. Addition of zinc oxide to microcapsules resulted in higher serum levels than those prepared without zinc oxide, suggesting a stabilizing effect of zinc oxide after subcutaneous injection into rats. The release profile of rhGH from microcapsules was controllable by selecting the proper copoly(DL-lactic/glycolic)acid (PLGA) with L/G ratio and molecular weight. Utilization of rhGH powder with a smaller particle size obtained by lyophilizing with ammonium acetate is essential for preparation of microcapsules with high entrapment and well-controlled sustained release profile with small initial release.


Molecular Cancer | 2013

Nectin-2 is a potential target for antibody therapy of breast and ovarian cancers

Tsutomu Oshima; Shuji Sato; Junichi Kato; Yuki Ito; Takahiro Watanabe; Isamu Tsuji; Akira Hori; Tomofumi Kurokawa; Toshio Kokubo

BackgroundNectin-2 is a Ca2+-independent cell-cell adhesion molecule that is one of the plasma membrane components of adherens junctions. However, little has been reported about the involvement of Nectin-2 in cancer.MethodsTo determine the expression of Nectin-2 in cancer tissues and cancer cell lines, we performed gene expression profile analysis, immunohistochemistry studies, and flow cytometry analysis. We also investigated the potential of this molecule as a target for antibody therapeutics to treat cancers by generating and characterizing an anti-Nectin-2 rabbit polyclonal antibody (poAb) and 256 fully human anti-Nectin-2 monoclonal antibodies (mAbs). In addition, we tested anti-Nectin-2 mAbs in several in vivo tumor growth inhibition models to investigate the primary mechanisms of action of the mAbs.ResultsIn the present study, we found that Nectin-2 was over-expressed in clinical breast and ovarian cancer tissues by using gene expression profile analysis and immunohistochemistry studies. Nectin-2 was over-expressed in various cancer cell lines as well. Furthermore, the polyclonal antibody specific to Nectin-2 suppressed the in vitro proliferation of OV-90 ovarian cancer cells, which express endogenous Nectin-2 on the cell surface. The anti-Nectin-2 mAbs we generated were classified into 7 epitope bins. The anti-Nectin-2 mAbs demonstrated antibody-dependent cellular cytotoxicity (ADCC) and epitope bin-dependent features such as the inhibition of Nectin-2-Nectin-2 interaction, Nectin-2-Nectin-3 interaction, and in vitro cancer cell proliferation. A representative anti-Nectin-2 mAb in epitope bin VII, Y-443, showed anti-tumor effects against OV-90 cells and MDA-MB-231 breast cancer cells in mouse therapeutic models, and its main mechanism of action appeared to be ADCC.ConclusionsWe observed the over-expression of Nectin-2 in breast and ovarian cancers and anti-tumor activity of anti-Nectin-2 mAbs via strong ADCC. These findings suggest that Nectin-2 is a potential target for antibody therapy against breast and ovarian cancers.


International Journal of Pharmaceutics | 2003

Preparation of a copoly (dl-lactic/glycolic acid)-zinc oxide complex and its utilization to microcapsules containing recombinant human growth hormone

Yutaka Yamagata; Masafumi Misaki; Tomofumi Kurokawa; Keiko Taira; Shigeyuki Takada

A procedure to prepare a complex of copoly (dl-lactic/glycolic acid) and zinc oxide (PLGA-zinc oxide complex) was developed. Out of sparingly water-soluble zinc compounds, zinc oxide was most remarkably soluble in a PLGA/dichloromethane solution and the dissolution rates became faster as the water contents in the PLGA/dichloromethane solutions increased. Since the solubility of zinc oxide was saturated at approximately 0.5-fold molar ratio to PLGA and water was generated with dissolution of zinc oxide in the PLGA/dichloromethane solutions, it is suggested that zinc oxide interacts with the terminal carboxyl group of PLGA. In addition, the glass-transition temperature of a solid material obtained by vacuum-drying the PLGA/dichloromethane solution dissolving zinc oxide became higher as the zinc content increased, suggesting that the formation of a PLGA-zinc oxide complex. Microcapsules were prepared with the PLGA-zinc oxide complex using recombinant human growth hormone (rhGH) in order to evaluate an effect of the complex on protein release and stability of protein in the microcapsules. Released rhGH amount from the microcapsules prepared with the PLGA-zinc oxide complex after subcutaneous administration in rats was significantly larger than that from microcapsules prepared with PLGA alone, indicating that rhGH molecules in the microcapsules was stabilized by the PLGA-zinc oxide complex.


Journal of Biotechnology | 1994

Expression and characterization of a chimeric bispecific antibody against fibrin and against urokinase-type plasminogen activator

Hiroko Tada; Tomofumi Kurokawa; Takeshi Seita; Takeshi Watanabe; Susumu Iwasa

We have produced a chimeric bispecific antibody that has dual specificity of human fibrin and urokinase-type plasminogen activator (u-PA). Complementary DNAs for variable regions of both anti-fibrin and anti-u-PA antibodies were cloned from two murine hybridomas secreting respective antibodies using polymerase chain reaction (PCR) techniques, and joined to cDNAs for human constant regions to form chimeric antibody genes. Both of two expression vectors for chimeric anti-fibrin and chimeric anti-u-PA antibodies were sequentially introduced into Chinese hamster ovary cells, and stable transfectants secreting the chimeric bispecific antibody were obtained. The highest producer transfectant (SULF/C2-30) secreted high level (about 40 micrograms ml-1) of total chimeric IgG and about 2% of the IgG had the bispecific activity of binding with both antigens. The chimeric bispecific antibody was purified by a combination of affinity chromatographies employing antigen-coupled columns and hydroxyapatite high-performance liquid chromatography. The purified chimeric bispecific antibody significantly enhanced the thrombolytic potency of single chain u-PA in an in vitro clot lysis assay as well as the original murine bispecific antibody.


International Journal of Pharmaceutics | 1997

Sustained release of a water-soluble GP IIb/IIIa antagonist from copoly(dl-lactic/glycolic)acid microspheres

Shigeyuki Takada; Tomofumi Kurokawa; Keiko Miyazaki; Susumu Iwasa; Yasuaki Ogawa

Sustained release of TAK-029 [4-(4-amidinobenzoylglycyl)-3-methoxycarbonyl-2-oxopiperazine-l-acetic acid], a glycoprotein (GP) IIb/IIIa antagonist, from injectable microspheres was achieved by a W/O/W emulsion solvent evaporation technique using copoly(d-lactic/glycolic)acid (PLGA). Entrapment of the drug into microspheres increased with addition of sodium chloride into an external aqueous polyvinyl alcohol solution. Addition of l-arginine to an internal water phase dose-dependently reduced initial burst of the drug from the microspheres in vitro and in vivo, probably by forming hydrophobic diffusion barriers with rigid inner structure and increased glass transition temperature (Tg). Microspheres obtained using sodium chloride and l-arginine demonstrated sustained plasma levels of TAK-029 for 3 weeks after subcutaneous injection in rats, while causing a slight increase of its plasma levels in 2–3 weeks.


mAbs | 2012

Characterization of a variety of neutralizing anti-heparin-binding epidermal growth factor-like growth factor monoclonal antibodies by different immunization methods.

Isamu Tsuji; Shuji Sato; Kentaro Otake; Takahiro Watanabe; Hiroko Kamada; Tomofumi Kurokawa

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a member of the epidermal growth factor family. The accumulated evidence on the tumor-progressing roles of HB-EGF has suggested that HB-EGF-targeted cancer therapy is expected to be promising. However, the generation of neutralizing anti-HB-EGF monoclonal antibodies (mAbs) has proved difficult. To overcome this difficulty, we performed a hybridoma approach using mice from different genetic backgrounds, as well as different types of HB-EGF immunogens. To increase the number of hybridoma clones to screen, we used an electrofusion system to generate hybridomas and a fluorometric microvolume assay technology to screen anti-HB-EGF mAbs. We succeeded in obtaining neutralizing anti-HB-EGF mAbs, primarily from BALB/c and CD1 mice, and these were classified into 7 epitope bins based on their competitive binding to the soluble form of HB-EGF (sHB-EGF). The mAbs showed several epitope bin-dependent characteristics, including neutralizing and binding activity to human sHB-EGF, cross-reactivity to mouse/rat sHB-EGF and binding activity to the precursor form of HB-EGF. The neutralizing activity was also validated in colony formation assays. Interestingly, we found that the populations of mAb bins and the production rates of the neutralizing mAbs were strikingly different by mouse strain and by immunogen type. We succeeded in generating a variety of neutralizing anti-HB-EGF mAbs, including potent sHB-EGF neutralizers that may have potential as therapeutic agents for treating HB-EGF-dependent cancers. Our results also suggest that immunization approaches using different mouse strains and immunogen types affect the biological activity of individual neutralizing antibodies.


Journal of Pharmacy and Pharmacology | 2003

A new animal model for evaluation of long-term growth rate over one month by rhGH/PLGA microcapsule formulations

Shigeyuki Takada; Tomofumi Kurokawa; Masafumi Misaki; Keiko Taira; Yutaka Yamagata

A new animal model to evaluate the long‐term growth rate produced by a sustained‐release formulation of recombinant human growth hormone (rhGH) over one month was developed and the usefulness of our microcapsule formulations was demonstrated in this model. Long‐term pharmacological effects by subcutaneous injection of microcapsules for sustained release of rhGH were evaluated in hypophysectomized (Hpx) rats treated with immunosuppressive agent along with hormone supplement. Copoly(dl‐lactic/glycolic)acid (PLGA) microcapsules for sustained release of rhGH, a two‐week sustained‐release formulation (rhGH‐SR‐2W) and a one‐month sustained‐release formulation (rhGH‐SR‐1M), were prepared by a solid‐in‐oil‐in‐water emulsion solvent evaporation technique. Body‐weight gain, body‐length gain and serum levels of rat insulin‐like growth factor‐l (rlGF‐l) induced by subcutaneous injection of rhGH‐SR were compared with those by daily injections of rhGH solution in Hpx rats for 35 days. Serum IGF‐l levels in Hpx rats after the injection of rhGH‐SR‐2W microcapsules were higher than those after daily injections of rhGH solution. Body‐length gain, a new parameter, after single injection of rhGH‐SR‐1M microcapsules demonstrated the higher growth rate than that after daily injections of rhGH solution for 35 days. Thus, single injection of rhGH‐SR microcapsules demonstrated long‐term pharmacological effects greater than those by daily injections of rhGH solution in a newly developed model, immunosuppressed Hpx rats.


Thrombosis Research | 1990

Enhancement of fibrinolysis by bispecific monoclonal antibodies reactive to fibrin and plasminogen activators

Tomofumi Kurokawa; Susumu Iwasa; Atsushi Kakinuma

Murine hybrid hybridomas secreting bispecific monoclonal antibodies (bs mAbs) were constructed by fusing hybridomas secreting an anti-tissue plasminogen activator (tPA) or anti-urokinase (UK) mAb with hybridomas secreting a mAb which binds to human fibrin but not to fibrinogen. The bs mAbs reactive to both fibrin and PA (tPA or UK) were purified by affinity chromatography employing the respective antigen-coupled columns and characterized by fibrin-binding, amidolytic and fibrinolytic assays. Immunochemical conjugation of PAs and the bs mAbs did not impair the catalytic activity of PAs at all and made it possible to concentrate PAs on fibrin clot. Pretreatment of fibrin with the bs mAbs enhanced the fibrin-binding of PAs and the subsequent fibrinolysis.


Journal of The Chemical Society-perkin Transactions 1 | 1999

Further studies on the chemical cleavage of an N-terminal extra methionine from recombinant methionylated proteins

Masato Suenaga; Hiroaki Ohmae; Norio Okutani; Tomofumi Kurokawa; Tsuneo Asano; Takao Yamada; Osamu Nishimura; Masahiko Fujino

An additional methionine residue of recombinant human growth hormone (hGH) was converted into an oxoacyl form with glyoxylic acid, copper(II) sulfate and pyridine, and then cleaved from the rest of the protein with 3,4-diaminobenzoic acid in the presence of 1 M AcOH and 2 M HCO2Na. The conditions for N-terminal methionine cleavage worked better than the previous conditions with 1,2-phenylenediamine. The same protocol was also applicable to the methionylated forms of recombinant human betacellulin (BTC), neurotrophin-3 (NT-3) and human interleukin-2 (IL-2). The conversion yield for hGH, BTC, NT-3 and IL-2 increased up to approximately 80, 70, 55 and 50%, respectively. These results indicate that non-methionylated recombinant proteins could be prepared from the methionylated derivatives by chemical methods.


Protein Expression and Purification | 2011

l-Cysteine-enhanced renaturation of bioactive soluble tumor necrosis factor ligand family member LIGHT from inclusion bodies in Escherichia coli

Isamu Tsuji; Hideki Mastui; Tatsuo Ito; Tomofumi Kurokawa; Yasushi Shintani

LIGHT is a membrane-bound protein that belongs to the tumor necrosis factor (TNF) superfamily ligands. In this study, we established an effective strategy for producing a bioactive soluble form of LIGHT (sLIGHT), an extracellular region (Ile⁸⁴-Val²⁴⁰) of human LIGHT. Because sLIGHT was expressed as inclusion bodies in Escherichia coli, we investigated reagents that enhance the renaturation of sLIGHT from the inclusion bodies. Interestingly, L-cysteine in the denaturation buffer containing 3.5 M guanidine hydrochloride significantly improved the renaturation efficiency of sLIGHT. The effect of L-cysteine was synergistically enhanced by L-arginine in the refolding buffer. The optimal concentrations of L-cysteine and L-arginine in the denaturation and refolding buffers were 8 mM and 0.8 M, respectively. With these buffers, approximately 90 mg of sLIGHT was purified from 200 g of frozen E. coli cells. sLIGHT thus obtained significantly induced apoptosis in the WiDr human colon adenocarcinoma cell line at nanomolar concentrations, the same amount of sLIGHT that was produced by Sf9 insect cells. These results suggest that L-cysteine in the denaturation buffer enhances the renaturation of recombinant proteins from inclusion bodies in E. coli.

Collaboration


Dive into the Tomofumi Kurokawa's collaboration.

Top Co-Authors

Avatar

Susumu Iwasa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Shigeyuki Takada

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tsutomu Oshima

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenjiro Tanoue

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shuji Sato

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yukio Toyoda

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsushi Kakinuma

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Isamu Tsuji

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Takao Yamada

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge