Network


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

Hotspot


Dive into the research topics where Eiko Shibata is active.

Publication


Featured researches published by Eiko Shibata.


Oncogene | 2000

The C3(1)/SV40 T-antigen transgenic mouse model of mammary cancer: ductal epithelial cell targeting with multistage progression to carcinoma.

Jeffrey E. Green; Masa-Aki Shibata; Katsuhide Yoshidome; Min-Ling Liu; Cheryl L. Jorcyk; Miriam R. Anver; Jon M. Wigginton; Robert H. Wiltrout; Eiko Shibata; Stanislaw J. Kaczmarczyk; Weili Wang; Zi-yao Liu; Alfonso Calvo; Christine Couldrey

The 5′ flanking region of the C3(1) component of the rat prostate steroid binding protein (PSBP) has been used to successfully target the expression of the SV40 large T-antigen (Tag) to the epithelium of both the mammary and prostate glands resulting in models of mammary and prostate cancers which histologically resemble the human diseases. Atypia of the mammary ductal epithelium develops at about 8 weeks of age, progressing to mammary intraepithelial neoplasia (resembling human ductal carcinoma in situ [DCIS]) at about 12 weeks of age with the development of invasive carcinomas at about 16 weeks of age in 100% of female mice. The carcinomas share features to what has been classified in human breast cancer as infiltrating ductal carcinomas. All FVB/N female mice carrying the transgene develop mammary cancer with about a 15% incidence of lung metastases. Approximately 10% of older male mice develop anaplastic mammary carcinomas. Unlike many other transgenic models in which hormones and pregnancy are used to induce a mammary phenotype, C3(1)/Tag mice develop mammary tumors in the mammary epithelium of virgin animals without hormone supplementation or pregnancy. Although mammary tumor development appears hormone-responsive at early stages, invasive carcinomas are hormone-independent, which corresponds to the loss of estrogen receptor-α expression during tumor progression. Molecular and biologic factors related to mammary tumor progression can be studied in this model since lesions evolve over a predictable time course. Genomic alterations have been identified during tumor progression, including an amplification of the distal portion of chromosome 6 containing ki-ras and loss of heterozygosity (LOH) in other chromosomal regions. We have demonstrated that stage specific alterations in the expression of genes which are critical regulators of the cell cycle and apoptosis are functionally important in vivo. C3(1)/Tag mice appear useful for testing particular therapies since growth of the mammary tumors can be reduced using chemopreventive agents, cytokines, and an anti-angiogenesis agent.


Cancer Gene Therapy | 2008

Combination therapy with short interfering RNA vectors against VEGF-C and VEGF-A suppresses lymph node and lung metastasis in a mouse immunocompetent mammary cancer model

Masa-Aki Shibata; Junji Morimoto; Eiko Shibata; Yoshinori Otsuki

Cancer metastasis contributes significantly to cancer mortality and is facilitated by lymphangiogenesis and angiogenesis. Vascular endothelial growth factor-C (VEGF-C) and VEGF-A are involved in lymphangiogenesis and angiogenesis. To inhibit metastasis, combination therapy with vector-based small interfering RNA (siRNA) against VEGF-C and/or VEGF-A was conducted on murine metastatic mammary cancer. Syngeneic, inoculated, metastatic mammary cancers received direct intratumoral injection of plasmid siRNA vector targeting VEGF-C (psiRNA-VEGF-C), VEGF-A (psiRNA-VEGF-A), both VEGF-C and VEGF-A (both psiRNA-VEGF-C and psiRNA-VEGF-A vectors injected, referred to as the psiRNA-VEGF-C+A group) or a scrambled sequence (psiRNA-SCR) as control, once a week for 8 weeks. Gene electrotransfer was performed on the tumors after each injection. Tumor volume was significantly lower in the psiRNA-VEGF-A and the psiRNA-VEGF-C+A groups throughout the study. Lymph node metastasis was significantly less frequent in all therapeutic groups, whereas the multiplicity of lung metastases was significantly lower in the psiRNA-VEGF-C+A group only. All siRNA therapeutic groups showed a significant reduction in the number of dilated lymphatic vessels containing intraluminal cancer cells and microvessel density. Our data suggest that specific silencing of the VEGF-C or VEGF-A gene alone can inhibit lymph node metastasis. However, combination siRNA therapy targeting both VEGF-C and VEGF-A inhibits both lymph node and lung metastasis, rendering this combined therapy more beneficial than either alone. The observed anti-metastatic activity of siRNA-expressing vectors targeting VEGF-C or VEGF-A may be of high clinical significance in the treatment of metastatic breast cancer.


Cancer Gene Therapy | 2001

Suppression of mammary carcinoma growth in vitro and in vivo by inducible expression of the Cdk inhibitor p21

Masa-Aki Shibata; Katsuhide Yoshidome; Eiko Shibata; Cheryl L. Jorcyk; Jeffrey E. Green

Mammary carcinomas that develop in C3(1)/SV40 T-antigen (TAg) transgenic mice have lost the p53-mediated induction of p21, leading to increased cellular proliferation and significant elevations of cyclins and Cdks. To test whether p21 could serve as a target for anticancer therapy for this mammary cancer model, a retroviral delivery system for the inducible expression of p21 was developed. We demonstrate that overexpression of p21 in C3(1)/TAg mammary tumor cells using the retroviral inducible p21 expression system results in increased apoptosis, reduced cell proliferation in vitro and reduced tumor growth in vivo associated with reduced expression of cyclins D1 and E, and Cdks 2, 4, and 6. Reciprocal changes in the expression of p21 and p27Kip1, another cell-cycle regulator, were also observed. Because reduced p21 expression occurs frequently in human breast cancer, restoration of the Cdk inhibitor p21 by gene therapy approaches may provide a method for inhibiting mammary tumor progression. Cancer Gene Therapy (2001) 8, 23–35


BMC Medicine | 2010

The endogenous soluble VEGF receptor-2 isoform suppresses lymph node metastasis in a mouse immunocompetent mammary cancer model

Masa-Aki Shibata; Jayakrishna Ambati; Eiko Shibata; Romulo Albuquerque; Junji Morimoto; Yuko Ito; Yoshinori Otsuki

BackgroundCancer metastasis contributes significantly to cancer mortality and is facilitated by lymphangiogenesis and angiogenesis. A new splicing variant, endogenous soluble vascular endothelial growth factor receptor-2 (esVEGFR-2) that we recently identified is an endogenous selective inhibitor of lymphangiogenesis. To evaluate the antimetastatic potential of esVEGFR-2, gene therapy with vector expressing esVEGFR-2 (pesVEGFR-2) or endostatin (pEndo) as a positive control was conducted on murine metastatic mammary cancer.MethodsSyngeneic inoculated metastatic mammary cancers received direct intratumoral injection of pesVEGFR-2, pEndo or pVec as control, once a week for six weeks. In vivo gene electrotransfer was performed on the tumors after each injection.ResultsDeaths from metastasis were much lower in the pesVEGFR-2 and pEndo groups than in those of the pVec. Tumor volume was significantly lower in the pesVEGFR-2 and the pEndo groups throughout the study. Multiplicity of lymph node and lung metastatic nodules was significantly suppressed in the pesVEGFR-2 and pEndo groups. Moreover, the total number of overall metastasis including the other organs was also decreased in these groups. However, pesVEGFR-2 was not able to decrease the number of lungs, ovaries, kidneys and adrenals with metastasis as counted by unilateral or bilateral metastasis. The number of CD34+/Lyve-1- blood microvessels was significantly decreased in the pEndo group, while the number of CD34-/Lyve-1+ lymphatic vessels was significantly decreased in the pesVEGFR-2 and pEndo groups. In addition, a significant reduction in the number of dilated lymphatic vessels containing intraluminal cancer cells was observed in the pesVEGFR-2 and pEndo groups. Levels of apoptosis were significantly increased in the pEndo group, whereas the rates of cell proliferation were significantly decreased in the pesVEGFR-2 and pEndo groups.ConclusionsOur data demonstrate that esVEGFR-2 can inhibit mainly lymph node metastasis. The antimetastatic activity of esVEGFR-2 may be of high clinical significance in the treatment of metastatic breast cancer because lymph node involvement is a most important prognostic factor in cancer patients.


BMC Cancer | 2010

Raloxifene inhibits tumor growth and lymph node metastasis in a xenograft model of metastatic mammary cancer

Masa-Aki Shibata; Junji Morimoto; Eiko Shibata; Hitomi Kurose; Kanako Akamatsu; Zhong-Lian Li; Moriaki Kusakabe; Masahide Ohmichi; Yoshinori Otsuki

BackgroundThe effects of raloxifene, a novel selective estrogen receptor modulator, were studied in a mouse metastatic mammary cancer model expressing cytoplasmic ERα.MethodsMammary tumors, induced by inoculation of syngeneic BALB/c mice with BJMC3879luc2 cells, were subsequently treated with raloxifene at 0, 18 and 27 mg/kg/day using mini-osmotic pumps.ResultsIn vitro study demonstrated that the ERα in BJMC3879luc2 cells was smaller (between 50 and 64 kDa) than the normal-sized ERα (66 kDa) and showed cytoplasmic localization. A statistically significant but weak estradiol response was observed in this cell line. When BJMC3879luc2 tumors were implanted into mice, the ERα mRNA levels were significantly higher in females than in males. In vitro studies showed that raloxifene induced mitochondria-mediated apoptosis and cell-cycle arrest in the G1-phase and a decrease in the cell population in the S-phase. In animal experiments, tumor volumes were significantly suppressed in the raloxifene-treated groups. The multiplicity of lymph node metastasis was significantly decreased in the 27 mg/kg group. Levels of apoptosis were significantly increased in the raloxifene-treated groups, whereas the levels of DNA synthesis were significantly decreased in these groups. No differences in microvessel density in tumors were observed between the control and raloxifene-treated groups. The numbers of dilated lymphatic vessels containing intraluminal tumor cells were significantly reduced in mammary tumors in the raloxifene-treated groups. The levels of ERα mRNA in mammary tumors tended to be decreased in the raloxifene-treated groups.ConclusionThese results suggest that the antimetastatic activity of raloxifene in mammary cancer expressing cytoplasmic ERα may be a crucial finding with clinical applications and that raloxifene may be useful as an adjuvant therapy and for the chemoprevention of breast cancer development.


Medical Molecular Morphology | 2012

Mammary cancer gene therapy targeting lymphangiogenesis: VEGF-C siRNA and soluble VEGF receptor-2, a splicing variant

Masa-Aki Shibata; Jayakrishna Ambati; Eiko Shibata; Katsuhide Yoshidome; Mariko Harada-Shiba

Metastasis contributes significantly to cancer mortality, and the most common pathway of initial dissemination is via the afferent ducts of the lymphatics. Overexpression of vascular endothelial growth factor (VEGF)-C has been associated with lymphangiogenesis and lymph node metastasis in a multitude of human neoplasms, including breast cancers. We recently reported that both VEGF-C siRNA and endogenous soluble vascular endothelial growth factor receptor-2 (esVEGFR-2, a new splicing variant) inhibit VEGF-C function and metastasis in a mouse model of metastatic mammary cancer. Here we briefly review our previous experimental work, specifically targeting tumor lymphangiogenesis, in which metastatic mouse mammary cancers received direct intratumoral injections of either expression vectors VEGF-C siRNA or esVEGFR-2, or the empty plasmid vector, once a week for 6 or 8 weeks, followed by in vivo gene electrotransfer of the injected tumors. Throughout our study, both tumor lymphangiogenesis and the multiplicity of lymph node metastasis were significantly inhibited, with an overall reduction in tumor growth, by both VEGF-C siRNA and esVEGFR-2; further, a significant reduction in the number of dilated lymphatic vessels containing intraluminal cancer cells was observed with both treatments. Thus, therapeutic strategies targeting lymphangiogenesis may have great clinical significance for the treatment of metastatic human breast cancer.


Archive | 2011

Inhibition of Tumor Growth and Metastasis by a Combination of Anti-VEGF-C and Enhanced IL-12 Therapy in an Immunocompetent Mouse Mammary Cancer Model

Masa-Aki Shibata; Junji Morimoto; Eiko Shibata; Mariko Harada-Shiba; Shigekazu Fujioka

Breast cancer represents a major health problem in women, with more than 1,000,000 new cases and 370,000 deaths yearly worldwide [1]. Perhaps more worrisome is an apparently increasing incidence of breast cancer among younger women under 40 years of age recently reported in many countries worldwide [2-4]. The lethality of breast cancer is largely due to metastasis, preferentially to the lymph nodes, lungs and bones [5]; in order to delay the progression of breast cancer and prolong patient life, more effective chemopreventive and antimetastatic treatments and less toxic chemotherapeutic agents are desperately required. Vascular endothelial growth factor-C (VEGF-C) is expressed in a variety of malignant tumors including mammary cancer [6] and over-expression of VEGF-C has been reported to be associated with lymph node metastasis and poor prognosis in breast cancer patients [7,8]. A number of animal studies using cell lines [9-11] and transgenic mice[12] have been conducted in an attempt to demonstrate that VEGF-C over-expression is able to promote cancer metastasis. Using a ‘RNA interference’ approach with an immunocompetent mouse mammary cancer model, we previously demonstrated that inhibition of VEGF-C or VEGF-A by gene silencing using vectors expressing short interfering RNA (siRNA) leads to suppression of lymphatic and/or hematogenous metastasis [13]. The cytokine interleukin-12 (IL-12), a heterodimer composed of p35 and p40 subunits, is produced primarily by dendritic cells, macrophages/monocytes, and neutrophils and functions in enhancing the activity of cytotoxic T lymphocytes and NK cells. Both subunits are necessary to exert biological activity [14]. IL-12 plays an important role in the induction of a cell-mediated immune response [15]. This cytokine is also involved in the differentiation of native T cells to the Th1 subset, and induces production of interferon- (IFN) in both T and NK cells. In addition, IL-12 has been shown to exert a potent anti-neoplastic effect in a


Carcinogenesis | 2003

Comparative effects of lovastatin on mammary and prostate oncogenesis in transgenic mouse models

Masa-Aki Shibata; Claudine Kavanaugh; Eiko Shibata; Hideaki Abe; Phuongmai Nguyen; Yoshinori Otsuki; Jane B. Trepel; Jeffrey E. Green


Cancer Research | 2001

2-difluoromethylornithine and dehydroepiandrosterone inhibit mammary tumor progression but not mammary or prostate tumor initiation in C3(1)/SV40 T/t-antigen transgenic mice.

Jeffrey E. Green; Masa-Aki Shibata; Eiko Shibata; Richard C. Moon; Miriam R. Anver; Gary J. Kelloff; Ronald A. Lubet


Anticancer Research | 2009

Panaxanthone isolated from pericarp of Garcinia mangostana L. suppresses tumor growth and metastasis of a mouse model of mammary cancer.

Hitoshi Doi; Masa-Aki Shibata; Eiko Shibata; Junji Morimoto; Yukihiro Akao; Munekazu Iinuma; Nobuhiko Tanigawa; Yoshinori Otsuki

Collaboration


Dive into the Eiko Shibata's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariko Harada-Shiba

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Jeffrey E. Green

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miriam R. Anver

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Hideaki Abe

Tokyo Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge