hosted by
publicationslist.org
    
Hiromitsu Jinno

jinno@sc.itc.keio.ac.jp

Journal articles

2007
 
PMID 
Tadashi Ikeda, Hiromitsu Jinno, Masatoshi Shirane (2007)  Chemosensitivity-related genes of breast cancer detected by DNA microarray.   Anticancer Res 27: 4C. 2649-2655 Jul/Aug  
Abstract: BACKGROUND: The feasibility of a preoperative docetaxel/5'-deoxy-5-fluorouridine (5'-DFUR) regimen for breast cancer patients was examined and the genes related to the response to it was investigated. PATIENTS AND METHODS: Women with advanced breast cancer were treated with docetaxel (60 mg/m2, day 1) and 5'-DFUR (800 mg/day, on days 1-14) q3 weeks by 4 cycles. Microarray analysis was carried out using preoperative core biopsy samples. Based on the mRNA expression levels, genes related to clinical and pathological responses were selected. RESULTS: The docetaxel/5'-DFUR regimen showed a 86% clinical response rate including 42% complete response, one pathological complete response and one ductal carcinoma in situ component. In microarray analysis, we identified 6 genes, including IGF-1, and derived a predictive formula with 67% accuracy. In addition, x2 analysis revealed a tendency for good response in ER-negative and Her2/neu-positive cases. CONCLUSION: Microarray analysis enabled us to predict the pathological response to docetaxel/5'-DFUR chemotherapy.
Notes:
 
DOI   
PMID 
N Chiba, M Ueda, T Shimada, H Jinno, J Watanabe, K Ishihara, M Kitajima (2007)  Novel immunosuppressant agents targeting activated lymphocytes by biocompatible MPC polymer conjugated with interleukin-2.   Eur Surg Res 39: 2. 103-110 02  
Abstract: The immunopharmacological profile of novel biocompatible water-soluble interleukin-2 (IL-2)-conjugated 2-methacryloyloxyethyl phosphorylcholine (MPC) polymer immunosuppressive agents was established. MPC-co-n- butyl methacrylate (BMA)-co-p-nitrophenylcarbonyloxyethyl methacrylate (NPMA) (PMBN) was prepared as a backbone for these novel agents. PMBN contained MPC as a biocompatible unit, BMA as a hydrophobic domain in water, and NPMA as an immobilizable unit with IL-2. This research showed that proliferation of cell lines with high-affinity IL-2 receptors derived from T cell malignancies were suppressed by the PMBN conjugated with IL-2 (PMBN-IL2 conjugate) incorporating paclitaxel (PTX) and cyclosporin A at lower concentrations than used conventionally. PMBN-IL2 conjugates incorporating PTX also inhibited the proliferation of responder cells in a human mixed lymphocyte culture at a lower concentration than unconjugated drug. However, PMBN-IL2 conjugates incorporating FK506 inhibited proliferation no more than FK506 alone. The PMBN-IL2 conjugate with PTX may therefore be useful for selectively eliminating activated lymphocytes that hyperproduce high-affinity IL-2 receptors. As an entirely human 'immunotoxin analogue' it may not be associated with the dose-limiting toxicity and immunogenicity of conventional immunotoxins.
Notes:
 
DOI   
PMID 
Naokazu Chiba, Masakazu Ueda, Toshiyuki Shimada, Hiromitsu Jinno, Junji Watanabe, Kazuhiko Ishihara, Masaki Kitajima (2007)  Development of gene vectors for pinpoint targeting to human hepatocytes by cationically modified polymer complexes.   Eur Surg Res 39: 1. 23-34 01  
Abstract: We developed a vector that might enable gene therapy of metabolic liver disease or hepatoma. Here we demonstrate the use of cationically modified biocompatible phospholipid polymer conjugated with hepatitis B surface (HBs) antigen for the specific transfer of genes into human hepatocytes. Poly(2-methacryloyloxyethyl phosphorylcholine (MPC)- co-N,N-dimethylaminoethyl methacrylate (DMAEMA)-co- p-nitrophenylcarbonyloxyethyl methacrylate(NPMA))(polyMDN) was prepared as a frame of vector. The specific expression of sFlt-1 or GFP by polyMDN conjugated with HBs containing plasmid (plasmid/polyMDN-HBs), polyMDN containing plasmid (plasmid/polyMDN), plasmid only and PBS were assessed in tumor cells (HepG2 or WiDr) in vitro and in vivo. The histological findings, organ weight changes, and degree of liver dysfunction were examined in the mice administered by several reagents. The sFlt-1 and GFP expression was observed only in the HepG2 cells transfected with sFlt-1 or GFP/polyMDN-HBs. None of the side effects mentioned above was observed. In conclusion, these results suggest that polyMDN-HBs is a human hepatocyte-specific gene delivery vector that might not have serious side effects.
Notes:
 
PMID 
Masahiro Wada, Hiromitsu Jinno, Masakazu Ueda, Tadashi Ikeda, Masaki Kitajima, Tomohiro Konno, Junji Watanabe, Kazuhiko Ishihara (2007)  Efficacy of an MPC-BMA co-polymer as a nanotransporter for paclitaxel.   Anticancer Res 27: 3B. 1431-1435 May/Jun  
Abstract: BACKGROUND: Paclitaxel (PTX) is administered as a solution in polyoxyethylated castor oil (CO) due to its low water solubility, but solvent-induced side-effects may be severe. MATERIALS AND METHODS: PMB30W is a co-polymer of 2-methacryloyloxyethyl phosphorylcholine (MPC) and butyl methacrylate (BMA). Cytotoxicities of PTX in PMB30W (PTX-PMB30W) were examined in cell culture and in vivo. RESULTS: PTX-PMB30W and PTX in dimethyl sulfoxide showed similar toxicity in breast cancer cell lines MCF-7, SK-BR-3 and MX-1. Antitumor efficacies of PTX-PMB30W and PTX in CO (PTX-CO) were similar following weekly intraperitoneal administration of 50 mg/kg PTX in nude mice transplanted with MX-1 cells. At 200 mg/kg PTX, all animals died within 1 minute of PTX-CO administration. However, all animals receiving PTX-PBM30W survived. Ulceration occurred following subcutaneous injection of PTX-CO, but injection of PTX-PMB30W did not cause skin changes. CONCLUSION: Our data suggest that PMB30W can act as an effective PTX nanotransporter.
Notes:
2006
 
PMID 
Sojun Hoshimoto, Masakazu Ueda, Hiromitsu Jinno, Masaki Kitajima, Junichiro Futami, Masaharu Seno (2006)  Mechanisms of the growth-inhibitory effect of the RNase-EGF fused protein against EGFR-overexpressing cells.   Anticancer Res 26: 2A. 857-863 Mar/Apr  
Abstract: BACKGROUND: We previously showed the usefulness of a fused protein of human pancreatic ribonuclease1 (hRNase1) with human epidermal growth factor (hEGF) for molecular targeting of EGF receptor (EGFR)-overexpressing cells. In this study, the mechanisms underlying the inhibition of cell growth by RNase-EGF fused proteins was confirmed. MATERIALS AND METHODS: Des.1-7 hRNase1 was genetically fused to hEGF. The fused proteins were expressed and isolated from Escherichia coli. The internalization of hRNase1-hEGF was confirmed by confocal fluorescence microscopy. The growth-inhibitory effect of the fused proteins was evaluated by MTT assay. RESULTS: FITC-labelled hRNase1-hEGF was internalized into EGFR-overexpressing A431 cells. The internalization was not observed in A431 cells pre-treated with hEGF and EGFR-deficient H69 cells. The growth-inhibitory effect of des.1-7 hRNase1-hEGF against A431 cells was statistically significantly more pronounced than that of hRNase1-hEGF. CONCLUSION: RNase-EGF fused proteins are internalized through EGFR and inhibit cell growth by exerting their ribonucleolytic activity in the cytosol.
Notes:
 
PMID 
Sota Asaga, Masakazu Ueda, Hiromitsu Jinno, Kiyoshi Kikuchi, Osamu Itano, Tadashi Ikeda, Masaki Kitajima (2006)  Identification of a new breast cancer-related gene by restriction landmark genomic scanning.   Anticancer Res 26: 1A. 35-42 Jan/Feb  
Abstract: BACKGROUND: Molecular genetic analyses have demonstrated the importance of the accumulation of genomic changes in the etiology of cancer and, additionally, have identified valuable genetic markers for certain cancers. Although several prognostic markers have already been identified for breast cancer, it is clear that others remain to be identified. MATERIALS AND METHODS: Fourteen breast cancer samples and non-cancerous counterparts were applied to restriction landmark genomic scanning (RLGS) and 6 breast cancer cell lines (MCF-7, MDA-MB-435, T-47D, MDA-MB-231, SK-BR-3 and BT-20) and 9 cancer tissue samples were applied to reverse transcnptionalpolymerase chain reaction (RT-PCR) for screening of novel genetic alterations. RESULTS: Two spots were identified on the RLGS profiles of cancerous tissue that differed from those of normal tissue. Nucleotide sequencing and homology search analysis showed that these spots represented the voltage-dependent calcium channel alpha1H subunit gene (CACNA1H gene) and a locus immediately downstream of the growth factor receptor-binding protein 7 (GRB7) gene. Expression of the CACNA1H gene was confirmed by RT-PCR. CONCLUSION: Two genes, Grb7 and CACNA1H, were identified by RLGS. The expression of CACNA1H in breast cancer was confirmed for the first time.
Notes:
 
DOI   
PMID 
Hiroshi Yagi, Masakazu Ueda, Hiromitsu Jinno, Koichi Aiura, Shuji Mikami, Hiroko Tada, Masaharu Seno, Hidenori Yamada, Masaki Kitajima (2006)  Anti-tumor effect in an in vivo model by human-derived pancreatic RNase with basic fibroblast growth factor insertional fusion protein through antiangiogenic properties.   Cancer Sci 97: 12. 1315-1320 Dec  
Abstract: It is thought that the export of angiogenic fibroblast growth factors (FGF) from tumors may be involved in the onset of tumor angiogenesis. To create a new active targeting drug that inhibits the tumor angiogenic process without toxicities to normal cells, human basic FGF (h-bFGF) was inserted genetically into the Gly89 position of cross-linked RNase1 (the ribonuclease inhibitor protein [RI] binding site of cross-linked human pancreatic RNase) to prevent stereospecific binding to RI. The resultant insertional-fusion protein (CL-RFN89) was active both as h-bFGF and as RNase1. Furthermore, it acquired an additional ability of evading RI through steric blockade of RI binding caused by the fused h-bFGF domain. In the present study, the effect of the resultant protein, CL-RFN89, on the antitumor response though its antiangiogenic properties was investigated in an in vivo model. Continuous systemic treatment with CL-RFN89 significantly inhibited the growth of human A431 squamous cell carcinomas in vivo. Seven days of treatment with CL-RFN89 resulted in a 58.2% inhibition of tumor growth compared with control mice (P < 0.0001). Furthermore, immunohistochemistry using a rat antimouse CD31 antibody showed that treatment with CL-RFN89 reduced tumor vascularization. These findings identify CL-RFN89 as a potent systemic inhibitor of tumor growth as a result of its antiangiogenic properties. This protein appears to be a new systemic antitumor agent.
Notes:
1999
 
PMID 
H Jinno, M G Steiner, R G Mehta, M P Osborne, N T Telang (1999)  Inhibition of aberrant proliferation and induction of apoptosis in HER-2/neu oncogene transformed human mammary epithelial cells by N-(4-hydroxyphenyl)retinamide.   Carcinogenesis 20: 2. 229-236 Feb  
Abstract: Epithelial cells from non-cancerous mammary tissue in response to exposure to chemical carcinogens or transfection with oncogenes exhibit hyperproliferation and hyperplasia prior to the development of cancer. Aberrant proliferation may, therefore, represent a modifiable early occurring preneoplastic event that is susceptible to chemoprevention of carcinogenesis. The synthetic retinoid N-(4-hydroxyphenyl)retinamide (HPR), has exhibited preventive efficacy in several in vitro and in vivo breast cancer models, and represents a promising chemopreventive compound for clinical trials. Clinically relevant biochemical and cellular mechanisms responsible for the chemopreventive effects of HPR, however, are not fully understood. Experiments were performed on preneoplastic human mammary epithelial 184-B5/HER cells derived from reduction mammoplasty and initiated for tumorigenic transformation by overexpression of HER-2/neu oncogene, to examine whether HPR inhibits aberrant proliferation of these cells and to identify the possible mechanism(s) responsible for the inhibitory effects of HPR. Continuous 7-day treatment with HPR produced a dose-dependent, reversible growth inhibition. Long-term (21 day) treatment of 184-B5/HER cells with HPR inhibited anchorage-dependent colony formation by approximately 80% (P < 0.01) relative to that observed in the solvent control. A 24 h treatment with cytostatic 400 nM HPR produced a 25% increase (P = 0.01) in G0/G1 phase, and a 36% decrease (P = 0.01) in S phase of the cell cycle. HPR treatment also induced a 10-fold increase (P = 0.02) in the sub-G0 (apoptotic) peak that was down-regulated in the presence of the antioxidant N-acetyl-L-cysteine. Treatment with HPR resulted in a 30% reduction of cellular immunoreactivity to tyrosine kinase, whereas immunoreactivity to p185HER remained essentially unaltered. HPR exposure resulted in time-dependent increase in cellular metabolism of the retinoid as evidenced by increased formation of the inert metabolite N-(4-methoxyphenyl)-retinamide (MPR) and progressive increase in apoptosis. Thus, HPR-induced inhibition of aberrant proliferation may be caused, in part, by its ability to inhibit HER-2/neu-mediated proliferative signal transduction, retard cell cycle progression and upregulate cellular apoptosis.
Notes:
1996
 
PMID 
H Jinno, M Ueda, S Ozawa, T Ikeda, K Enomoto, K Psarras, M Kitajima, H Yamada, M Seno (1996)  Epidermal growth factor receptor-dependent cytotoxicity for human squamous carcinoma cell lines of a conjugate composed of human EGF and RNase 1.   Life Sci 58: 21. 1901-1908  
Abstract: Recombinant human ribonuclease 1 (RNase 1) was chemically linked to recombinant human epidermal growth factor (EGF). The EGF-RNase conjugate showed dose-dependent cytotoxicity for EGF receptor-overexpressing A431 and TE-8 human squamous carcinoma cells with an IC50 of 2 x 10(-7)M and 10(-6)M, respectively, whereas the IC50 of RNase alone was almost 10(-4)M. An unconjugated mixture of EGF and RNase had no greater effect than RNase alone. The conjugate showed no detectable cytotoxicity against EGF receptor-deficient small cell lung cancer cells (H69). Addition of excess EGF in the medium protected A431 cells from the EGF-RNase conjugate cytotoxicity. The cytotoxicity of the EGF-RNase conjugate was positively correlated with the EGF receptor numbers of each cell line. The chimeric toxin composed of only human proteins might be a more useful anti-cancer agent with less immunogenicity than the conventional chimeric toxins.
Notes:
 
PMID 
H Jinno, M Ueda, K Enomoto, T Ikeda, P Kyriakos, M Kitajima (1996)  Effectiveness of an adriamycin immunoconjugate that recognizes the C-erbB-2 product on breast cancer cell lines.   Surg Today 26: 7. 501-507  
Abstract: Adriamycin (ADM) was chemically conjugated to a murine monoclonal antibody, A0011, which recognizes the c-erbB-2 product, via a disulfide bond using N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP) and 2-iminothiolane (2-IT). The molar ratio of ADM to the monoclonal antibody ranged from 15:1 to 25:1 and enzyme-linked immunosorbent assay (ELISA) showed that the binding activity of the conjugate was almost retained. We compared the efficacy of A0011 alone, ADM alone, the A0011-ADM conjugate, against the human breast cancer cell lines SK-BR-3, MDA-MB-361, MCF-7, and BT-20. The A0011-ADM conjugate was observed to be ten times more cytotoxic to the cell lines overexpressing the c-erbB-2 product, namely, SK-BR-3 and MDA-MB-361, than free ADM, but it showed weak cytotoxicity against the cell lines with a low level of c-erbB-2 product expression, namely, MCF-7 and BT-20. However, free A0011 and nonspecific murine IgM-ADM conjugate showed no cytotoxicity toward any of the four cell lines, while the addition of a tenfold molar excess of A0011 inhibited conjugate cytotoxicity. These data suggest that conjugate cytotoxicity is antibody-mediated. Moreover, conjugate cytotoxicity at 10(-6)M was correlated with antigen volume, and the data were fitted to the regression equation y = -11.63logX + 116.38 where the correlation coefficient = 0.950. Our results indicate that targeting therapy aiming at the c-erbB-2 product may be useful in the treatment of breast cancers overexpressing the c-erbB-2 product.
Notes:
 
PMID 
H Jinno, M Ueda, S Ozawa, K Kikuchi, T Ikeda, K Enomoto, M Kitajima (1996)  Epidermal growth factor receptor-dependent cytotoxic effect by an EGF-ribonuclease conjugate on human cancer cell lines--a trial for less immunogenic chimeric toxin.   Cancer Chemother Pharmacol 38: 4. 303-308  
Abstract: Mammalian pancreatic ribonuclease (RNase) was conjugated chemically via a disulfide bond to human or murine epidermal growth factor (EGF). The conjugation between EGF and RNase was ascertained by SDS-PAGE using reduced and nonreduced conjugates. The EGF-RNase conjugate retained potent RNase activity and competed with 125I-EGF for binding to EGFR to the same extent as unconjugated EGF. Both the human and murine EGF-RNase conjugates showed dose-dependent cytotoxicity against EGFR-overexpressing A431 human squamous carcinoma cells with IC50 values of 3 x 10(-7) M and 6 x 10(-7) M, respectively, whereas free RNase had an IC50 of 10(-4) M. Against the EGFR-deficient small-cell lung cancer cell line H69, the EGF-RNase conjugate had no cytotoxic effect. The Human EGF-RNase conjugate showed dose-dependent cytotoxicity against other squamous carcinoma cell lines (TE-5, TE-1) and breast cancer cell lines (BT-20, SK-BR-3, MCF-7) and the cytotoxicity of the conjugate correlated positively with the level of expression of EGFR by each cell line. An unconjugated mixture of EGF and RNase had no greater effect than RNase alone on any cell line. Excess free EGF blocked EGF-RNase conjugate cytotoxicity against A431 cells. These results suggest that the EGF-RNase conjugate may be a more effective anticancer agent with less immunogenicity than coventional chimeric toxins.
Notes:
Powered by publicationslist.org.