Network


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

Hotspot


Dive into the research topics where Eiichi Hara is active.

Publication


Featured researches published by Eiichi Hara.


Clinical Cancer Research | 2005

Dendritic Cells Fused with Allogeneic Colorectal Cancer Cell Line Present Multiple Colorectal Cancer–Specific Antigens and Induce Antitumor Immunity against Autologous Tumor Cells

Shigeo Koido; Eiichi Hara; Sadamu Homma; Akira Torii; Yoichi Toyama; Hidejiro Kawahara; Michiaki Watanabe; Katsuhiko Yanaga; Kiyotaka Fujise; Hisao Tajiri; Jianlin Gong; Gotaro Toda

The aim of antitumor immunotherapy is to induce CTL responses against autologous tumors. Previous work has shown that fusion of human dendritic cells and autologous tumor cells induce CTL responses against autologous tumor cells in vitro. However, in the clinical setting of patients with colorectal carcinoma, a major difficulty is the preparation of sufficient amounts of autologous tumor cells. In the present study, autologous dendritic cells from patients with colorectal carcinoma were fused to allogeneic colorectal tumor cell line, COLM-6 (HLA-A2−/HLA-24−), carcinoembryonic antigen (CEA)+, and MUC1+ as an alternative strategy to deliver shared colorectal carcinoma antigens to dendritic cells. Stimulation of autologous T cells by the fusion cells generated with autologous dendritic cells (HLA-A2+ and/or HLA-A24+) and allogeneic COLM-6 resulted in MHC class I– and MHC class II–restricted proliferation of CD4+ and CD8+ T cells, high levels of IFN-γ production in both CD4+ and CD8+ T cells, and the simultaneous induction of CEA- and MUC1-specific CTL responses restricted by HLA-A2 and/or HLA-A24. Finally, CTL induced by dendritic cell/allogeneic COLM-6 fusion cells were able to kill autologous colorectal carcinoma by HLA-A2- and/or HLA-A24-restricted mechanisms. The demonstration of CTL activity against shared tumor-associated antigens using an allogeneic tumor cell line, COLM-6, provides that the presence of alloantigens does not prevent the development of CTL with activity against autologous colorectal carcinoma cells. The fusion of allogeneic colorectal carcinoma cell line and autologous dendritic cells could have potential applicability to the field of antitumor immunotherapy through the cross-priming against shared tumor antigens and provides a platform for adoptive immunotherapy.


Journal of Immunology | 2007

Streptococcal Preparation OK-432 Promotes Fusion Efficiency and Enhances Induction of Antigen-Specific CTL by Fusions of Dendritic Cells and Colorectal Cancer Cells

Shigeo Koido; Eiichi Hara; Sadamu Homma; Akira Torii; Makoto Mitsunaga; Satoru Yanagisawa; Yoichi Toyama; Hidejiro Kawahara; Michiaki Watanabe; Seiya Yoshida; Susumu Kobayashi; Katsuhiko Yanaga; Kiyotaka Fujise; Hisao Tajiri

Dendritic/tumor fusion cell (FC) vaccine is an effective approach for various types of cancer but has not yet been standardized. Antitumor activity can be modulated by different mechanisms such as dendritic cell (DC) maturation state. This study addressed optimal strategies for FC preparations to enhance Ag-specific CTL activity. We have created three types of FC preparations by alternating fusion cell partners: 1) immature DCs fused with autologous colorectal carcinoma cells (Imm-FCs); 2) Imm-FCs followed by stimulation with penicillin-inactivated Streptococcus pyogenes (OK-432) (Imm-FCs/OK); and 3) OK-432-stimulated DCs directly fused to autologous colorectal carcinoma cells (OK-FCs). Both OK-FCs and Imm-FCs/OK coexpressed the CEA, MUC1, and significantly higher levels of CD86, CD83, and IL-12 than those obtained with Imm-FCs. Short-term culture of fusion cell preparations promoted the fusion efficiency. Interestingly, OK-FCs were more efficient in stimulating CD4+ and CD8+ T cells capable of high levels of IFN-γ production and cytolysis of autologous tumor or semiallogeneic targets. Moreover, OK-FCs are more effective inducer of CTL activation compared with Imm-FCs/OK on a per fusion cell basis. The pentameric assay confirmed that CEA- and MUC1-specific CTL was induced simultaneously by OK-FCs at high frequency. Furthermore, the cryopreserved OK-FCs retained stimulatory capacity for inducing antitumor immunity. These results suggest that OK-432 promotes fusion efficiency and induction of Ag-specific CTL by fusion cells. We conclude that DCs fused after stimulation by OK-432 may have the potential applicability to the field of antitumor immunotherapy and may provide a platform for adoptive immunotherapy in the clinical setting.


International Journal of Cancer | 2005

Induction of antigen-specific CD4- and CD8-mediated T-cell responses by fusions of autologous dendritic cells and metastatic colorectal cancer cells.

Shigeo Koido; Eiichi Hara; Akira Torii; Sadamu Homma; Yoichi Toyama; Hidejiro Kawahara; Masaichi Ogawa; Michiaki Watanabe; Katsuhiko Yanaga; Kiyotaka Fujise; Jianlin Gong; Gotaro Toda

Human metastatic colorectal carcinomas (CRCAs) express carcinoembryonic antigen (CEA) and/or MUC1 tumor‐associated antigens as potential targets for the induction of active specific immunity. In the present study, freshly isolated metastatic CRCA cells were successfully fused with immature autologous human monocyte‐derived dendritic cells (DCs). The created heterokaryons (DC/CRCA) coexpress the CRCA‐derived CEA and MUC1 antigens and DC‐derived MHC class II and costimulatory molecules. The fusion cells were functional in stimulating the proliferation of autologous T cells. In addition, both CD4+ and CD8+ T cells were activated by fusion cells, as demonstrated by the production of high levels of IFN‐γ. More importantly, coculture of fusion cells with patient‐derived peripheral blood mononuclear cells (PBMCs) resulted in the induction of antigen‐specific cytotoxic T lymphocytes (CTLs). CTLs were effective at lysis of not only autologous CRCA cells but also the CEA and/or MUC1‐positive and HLA partially matched target cells. Antigen‐specific CTL responses were confirmed by tetrameric analysis. Coculture of PBMCs with fusion cells resulted in increased frequency of CEA‐ and MUC1‐specific CTLs simultaneously. Taken together, these results indicate that freshly isolated human metastatic CRCA cells expressing the CEA and/or MUC1 may represent a potential partner for the creation of DC/tumor fusion cells targeting induction of antigen‐specific CTL responses. Our report demonstrates the simultaneous induction of CRCA‐specific CTL responses restricted by HLA‐A2 and ‐A24.


Journal of Immunology | 2007

Synergistic Induction of Antigen-Specific CTL by Fusions of TLR-Stimulated Dendritic Cells and Heat-Stressed Tumor Cells

Shigeo Koido; Eiichi Hara; Sadamu Homma; Makoto Mitsunaga; Akitaka Takahara; Eijiro Nagasaki; Hidejiro Kawahara; Michiaki Watanabe; Yoichi Toyama; Satoru Yanagisawa; Susumu Kobayashi; Katsuhiko Yanaga; Kiyotaka Fujise; Jianlin Gong; Hisao Tajiri

Dendritic cell (DC)/tumor cell fusion cells (FCs) can induce potent CTL responses. The therapeutic efficacy of a vaccine requires the improved immunogenicity of both DCs and tumor cells. The DCs stimulated with the TLR agonist penicillin-killed Streptococcus pyogenes (OK-432; OK-DCs) showed higher expression levels of MHC class I and II, CD80, CD86, CD83, IL-12, and heat shock proteins (HSPs) than did immature DCs. Moreover, heat-treated autologous tumor cells displayed a characteristic phenotype with increased expression of HSPs, carcinoembryonic Ag (CEA), MUC1, and MHC class I (HLA-A2 and/or A24). In this study, we have created four types of FC preparation by alternating fusion cell partners: 1) immature DCs fused with unheated tumor cells; 2) immature DCs fused with heat-treated tumor cells; 3) OK-DCs fused with unheated tumor cells; and 4) OK-DCs fused with heat-treated tumor cells. Although OK-DCs fused with unheated tumor cells efficiently enhanced CTL induction, OK-DCs fused with heat-treated tumor cells were most active, as demonstrated by: 1) up-regulation of multiple HSPs, MHC class I and II, CEA, CD80, CD86, CD83, and IL-12; 2) activation of CD4+ and CD8+ T cells able to produce IFN- γ at higher levels; 3) efficient induction of CTL activity specific for CEA or MUC1 or both against autologous tumor; and 4) superior abilities to induce CD107+IFN-γ+CD8+ T cells and CD154+ IFN-γ+CD4+ T cells. These results strongly suggest that synergism between OK-DCs and heat-treated tumor cells enhances the immunogenicity of FCs and provides a promising means of inducing therapeutic antitumor immunity.


Clinical & Developmental Immunology | 2009

Cancer Vaccine by Fusions of Dendritic and Cancer Cells

Shigeo Koido; Eiichi Hara; Sadamu Homma; Yoshihisa Namiki; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

Dendritic cells (DCs) are potent antigen-presenting cells and play a central role in the initiation and regulation of primary immune responses. Therefore, their use for the active immunotherapy against cancers has been studied with considerable interest. The fusion of DCs with whole tumor cells represents in many ways an ideal approach to deliver, process, and subsequently present a broad array of tumor-associated antigens, including those yet to be unidentified, in the context of DCs-derived costimulatory molecules. DCs/tumor fusion vaccine stimulates potent antitumor immunity in the animal tumor models. In the human studies, T cells stimulated by DC/tumor fusion cells are effective in lysis of tumor cells that are used as the fusion partner. In the clinical trials, clinical and immunological responses were observed in patients with advanced stage of malignant tumors after being vaccinated with DC/tumor fusion cells, although the antitumor effect is not as vigorous as in the animal tumor models. This review summarizes recent advances in concepts and techniques that are providing new impulses to DCs/tumor fusions-based cancer vaccination.


Clinical & Developmental Immunology | 2010

Regulation of Tumor Immunity by Tumor/Dendritic Cell Fusions

Shigeo Koido; Sadamu Homma; Eiichi Hara; Yoshihisa Namiki; Akitaka Takahara; Hideo Komita; Eijiro Nagasaki; Masaki Ito; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

The goal of cancer vaccines is to induce antitumor immunity that ultimately will reduce tumor burden in tumor environment. Several strategies involving dendritic cells- (DCs)- based vaccine incorporating different tumor-associated antigens to induce antitumor immune responses against tumors have been tested in clinical trials worldwide. Although DCs-based vaccine such as fusions of whole tumor cells and DCs has been proven to be clinically safe and is efficient to enhance antitumor immune responses for inducing effective immune response and for breaking T-cell tolerance to tumor-associated antigens (TAAs), only a limited success has occurred in clinical trials. This paper reviews tumor immune escape and current strategies employed in the field of tumor/DC fusions vaccine aimed at enhancing activation of TAAs-specific cytotoxic T cells in tumor microenvironment.


Archivum Immunologiae Et Therapiae Experimentalis | 2007

Dendritic/tumor fusion cell-based vaccination against cancer

Shigeo Koido; Eiichi Hara; Sadamu Homma; Kiyotaka Fujise; Jianlin Gong; Hisao Tajiri

A promising area of investigation is the use of cancer vaccines to eliminate residual tumor cells. Dendritic cells (DCs) are potent professional antigen-presenting cells able to induce primary immune responses. DCs capture and process antigens into peptides and present them to T cells and B cells through MHC class I and II molecules. An alternative approach to the induction of antitumor immunity is the use of fusions of DCs and tumor cells. In this approach, a broad spectrum of tumor-associated antigens, including those known and unidentified, are processed endogenously and presented by MHC class I and II pathways in the context of costimulatory signals. In animal studies, vaccination with DC/tumor fusion cells results in the elimination of established lung metastasis. Preclinical human studies have demonstrated that DC/tumor fusion cells induce antigen-specific polyclonal cytotoxic T-lymphocyte responses against autologous tumor in vitro. In clinical studies, vaccination of cancer patients with autologous DC/tumor fusion cells is associated with immunological and clinical responses in a subset of patients. Future studies should be investigated to improve the immunogenicity of DC/tumor fusion cell preparations. This review provides a general overview of the DC/tumor fusion cell-based vaccine and summarizes some of the recent advances in this field.


Clinical Immunology | 2010

Dendritic/pancreatic carcinoma fusions for clinical use: Comparative functional analysis of healthy- versus patient-derived fusions.

Shigeo Koido; Eiichi Hara; Sadamu Homma; Yoshihisa Namiki; Hideo Komita; Akitaka Takahara; Eijiro Nagasaki; Masaki Ito; Yukiko Sagawa; Makoto Mitsunaga; Kan Uchiyama; Kenichi Satoh; Seiji Arihiro; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

Fetal calf serum (FCS)-independent pancreatic cancer cells were established in plasma protein fraction (PPF)-supplemented medium that is an agent of good manufacturing practice (GMP) grade. Dendritic cells (DCs) were activated with the Toll-like receptor agonist, penicillin-inactivated Streptococcus pyogenes (OK-432) that is also a GMP grade agent. Therefore, sufficient amounts of FCS-independent fusions were successfully generated with decreased potential hazards of FCS. The FCS-independent fusions expressed tumor-associated antigens, HLA-DR, costimulatory molecules, IL-12, and IL-10. Stimulation of T cells with fusions from healthy donors resulted in proliferation of T cells with high expression levels of perforin/granzyme B and IFN-gamma and efficient induction of antigen-specific cytotoxic T lymphocytes (CTLs). Selection and expansion of T-cell clones were confirmed by TCR Vbeta analysis. However, fusions from patients with metastatic pancreatic cancer induced increased expression levels of TGF-beta1 in CD4+ CD25high T cells and low levels of CTLs with decreased IFN-gamma production.


Journal of Translational Medicine | 2008

In vitro generation of cytotoxic and regulatory T cells by fusions of human dendritic cells and hepatocellular carcinoma cells

Shigeo Koido; Sadamu Homma; Eiichi Hara; Makoto Mitsunaga; Yoshihisa Namiki; Akitaka Takahara; Eijiro Nagasaki; Hideo Komita; Yukiko Sagawa; Toshifumi Ohkusa; Kiyotaka Fujise; Jianlin Gong; Hisao Tajiri

BackgroundHuman hepatocellular carcinoma (HCC) cells express WT1 and/or carcinoembryonic antigen (CEA) as potential targets for the induction of antitumor immunity. In this study, generation of cytotoxic T lymphocytes (CTL) and regulatory T cells (Treg) by fusions of dendritic cells (DCs) and HCC cells was examined.MethodsHCC cells were fused to DCs either from healthy donors or the HCC patient and investigated whether supernatants derived from the HCC cell culture (HCCsp) influenced on the function of DCs/HCC fusion cells (FCs) and generation of CTL and Treg.ResultsFCs coexpressed the HCC cells-derived WT1 and CEA antigens and DCs-derived MHC class II and costimulatory molecules. In addition, FCs were effective in activating CD4+ and CD8+ T cells able to produce IFN-γ and inducing cytolysis of autologous tumor or semiallogeneic targets by a MHC class I-restricted mechanism. However, HCCsp induced functional impairment of DCs as demonstrated by the down-regulation of MHC class I and II, CD80, CD86, and CD83 molecules. Moreover, the HCCsp-exposed DCs failed to undergo full maturation upon stimulation with the Toll-like receptor 4 agonist penicillin-inactivated Streptococcus pyogenes. Interestingly, fusions of immature DCs generated in the presence of HCCsp and allogeneic HCC cells promoted the generation of CD4+ CD25high Foxp3+ Treg and inhibited CTL induction in the presence of HCCsp. Importantly, up-regulation of MHC class II, CD80, and CD83 on DCs was observed in the patient with advanced HCC after vaccination with autologous FCs. In addition, the FCs induced WT1- and CEA-specific CTL that were able to produce high levels of IFN-γ.ConclusionThe current study is one of the first demonstrating the induction of antigen-specific CTL and the generation of Treg by fusions of DCs and HCC cells. The local tumor-related factors may favor the generation of Treg through the inhibition of DCs maturation; however, fusion cell vaccination results in recovery of the DCs function and induction of antigen-specific CTL responses in vitro. The present study may shed new light about the mechanisms responsible for the generation of CTL and Treg by FCs.


Immunotherapy | 2009

Cancer immunotherapy by fusions of dendritic cells and tumor cells.

Shigeo Koido; Eiichi Hara; Sadamu Homma; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

Dendritic cells (DCs) are potent professional antigen-presenting cells and play a critical role in the induction of primary immune responses. DC-based vaccination represents a potentially powerful strategy for cancer immunotherapy. Thus, the use of cancer vaccines to eliminate residual tumor cells is a promising area of investigation. The immunotherapy of tumor antigen-loaded DCs has now been demonstrated in cancer patients and some clinical responses without any significant toxicity. Fusions of DCs and tumor cells represent an alternative but promising approach to overcome the inability of tumor antigens to induce a sustainable T-cell response. This review deals with recent progress in the immunotherapy of cancer with fusions of DCs and tumor cells.

Collaboration


Dive into the Eiichi Hara's collaboration.

Top Co-Authors

Avatar

Sadamu Homma

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shigeo Koido

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hisao Tajiri

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshifumi Ohkusa

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Akitaka Takahara

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kiyotaka Fujise

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yoshihisa Namiki

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Makoto Mitsunaga

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eijiro Nagasaki

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge