Network


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

Hotspot


Dive into the research topics where Akitaka Takahara is active.

Publication


Featured researches published by Akitaka Takahara.


Clinical & Developmental Immunology | 2011

Current Immunotherapeutic Approaches in Pancreatic Cancer

Shigeo Koido; Sadamu Homma; Akitaka Takahara; Yoshihisa Namiki; Shintaro Tsukinaga; Jimi Mitobe; Shunichi Odahara; Toyokazu Yukawa; Hiroshi Matsudaira; Keisuke Nagatsuma; Kan Uchiyama; Kenichi Satoh; Masaki Ito; Hideo Komita; Hiroshi Arakawa; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

Pancreatic cancer is a highly aggressive and notoriously difficult to treat. As the vast majority of patients are diagnosed at advanced stage of the disease, only a small population is curative by surgical resection. Although gemcitabine-based chemotherapy is typically offered as standard of care, most patients do not survive longer than 6 months. Thus, new therapeutic approaches are needed. Pancreatic cancer cells that develop gemcitabine resistance would still be suitable targets for immunotherapy. Therefore, one promising treatment approach may be immunotherapy that is designed to target pancreatic-cancer-associated antigens. In this paper, we detail recent work in immunotherapy and the advances in concept of combination therapy of immunotherapy and chemotherapy. We offer our perspective on how to increase the clinical efficacy of immunotherapies for pancreatic cancer.


Journal of Immunotherapy | 2014

Wilms tumor gene (WT1) peptide-based cancer vaccine combined with gemcitabine for patients with advanced pancreatic cancer.

Sumiyuki Nishida; Shigeo Koido; Yutaka Takeda; Sadamu Homma; Hideo Komita; Akitaka Takahara; Satoshi Morita; Toshinori Ito; Soyoko Morimoto; Kazuma Hara; Akihiro Tsuboi; Yoshihiro Oka; Satoru Yanagisawa; Yoichi Toyama; Masahiro Ikegami; Toru Kitagawa; Hidetoshi Eguchi; Hiroshi Wada; Hiroaki Nagano; Jun Nakata; Yoshiki Nakae; Naoki Hosen; Yusuke Oji; Toshio Tanaka; Ichiro Kawase; Atsushi Kumanogoh; Junichi Sakamoto; Yuichiro Doki; Masaki Mori; Toshifumi Ohkusa

Wilms tumor gene (WT1) protein is an attractive target for cancer immunotherapy. We aimed to investigate the feasibility of a combination therapy consisting of gemcitabine and WT1 peptide–based vaccine for patients with advanced pancreatic cancer and to make initial assessments of its clinical efficacy and immunologic response. Thirty-two HLA-A*24:02+ patients with advanced pancreatic cancer were enrolled. Patients received HLA-A*24:02-restricted, modified 9-mer WT1 peptide (3 mg/body) emulsified with Montanide ISA51 adjuvant (WT1 vaccine) intradermally biweekly and gemcitabine (1000 mg/m2) on days 1, 8, and 15 of a 28-day cycle. This combination therapy was well tolerated. The frequencies of grade 3–4 adverse events for this combination therapy were similar to those for gemcitabine alone. Objective response rate was 20.0% (6/30 evaluable patients). Median survival time and 1-year survival rate were 8.1 months and 29%, respectively. The association between longer survival and positive delayed-type hypersensitivity to WT1 peptide was statistically significant, and longer survivors featured a higher frequency of memory-phenotype WT1-specific cytotoxic T lymphocytes both before and after treatment. WT1 vaccine in combination with gemcitabine was well tolerated for patients with advanced pancreatic cancer. Delayed-type hypersensitivity-positivity to WT1 peptide and a higher frequency of memory-phenotype WT1-specific cytotoxic T lymphocytes could be useful prognostic markers for survival in the combination therapy with gemcitabine and WT1 vaccine. Further clinical investigation is warranted to determine the effectiveness of this combination therapy.


Cancer Immunology, Immunotherapy | 2011

Gemcitabine enhances Wilms’ tumor gene WT1 expression and sensitizes human pancreatic cancer cells with WT1-specific T-cell-mediated antitumor immune response

Akitaka Takahara; Shigeo Koido; Masaki Ito; Eijiro Nagasaki; Yukiko Sagawa; Takeo Iwamoto; Hideo Komita; Toshiki Ochi; Hiroshi Fujiwara; Masaki Yasukawa; Junichi Mineno; Hiroshi Shiku; Sumiyuki Nishida; Haruo Sugiyama; Hisao Tajiri; Sadamu Homma

Wilms’ tumor gene (WT1), which is expressed in human pancreatic cancer (PC), is a unique tumor antigen recognized by T-cell-mediated antitumor immune response. Gemcitabine (GEM), a standard therapeutic drug for PC, was examined for the regulation of WT1 expression and the sensitizing effect on PC cells with WT1-specific antitumor immune response. Expression of WT1 was examined by quantitative PCR, immunoblot analysis, and confocal microscopy. Antigenic peptide of WT1 presented on HLA class I molecules was detected by mass spectrometry. WT1-specific T-cell receptor gene–transduced human T cells were used as effecter T cells for the analysis of cytotoxic activity. GEM treatment of human MIAPaCa2 PC cells enhanced WT1 mRNA levels, and this increase is associated with nuclear factor kappa B activation. Tumor tissue from GEM-treated MIAPaCa2-bearing SCID mice also showed an increase in WT1 mRNA. Some human PC cell lines other than MIAPaCa2 showed up-regulation of WT1 mRNA levels following GEM treatment. GEM treatment shifted WT1 protein from the nucleus to the cytoplasm, which may promote proteasomal processing of WT1 protein and generation of antigenic peptide. In fact, presentation of HLA-A*2402-restricted antigenic peptide of WT1 (CMTWNQMNL) increased in GEM-treated MIAPaCa2 cells relative to untreated cells. WT1-specific cytotoxic T cells killed MIAPaCa2 cells treated with an optimal dose of GEM more efficiently than untreated MIAPaCa2 cells. GEM enhanced WT1 expression in human PC cells and sensitized PC cells with WT1-specific T-cell-mediated antitumor immune response.


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 | 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.


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.


PLOS ONE | 2013

Combined TLR2/4-Activated Dendritic/Tumor Cell Fusions Induce Augmented Cytotoxic T Lymphocytes

Shigeo Koido; Sadamu Homma; Masato Okamoto; Yoshihisa Namiki; Kazuki Takakura; Akitaka Takahara; Shunichi Odahara; Shintaro Tsukinaga; Toyokazu Yukawa; Jimi Mitobe; Hiroshi Matsudaira; Keisuke Nagatsuma; Kan Uchiyama; Mikio Kajihara; Seiji Arihiro; Hiroo Imazu; Hiroshi Arakawa; Shin Kan; Hideo Komita; Masaki Ito; Toshifumi Ohkusa; Jianlin Gong; Hisao Tajiri

Induction of antitumor immunity by dendritic cell (DC)-tumor fusion cells (DC/tumor) can be modulated by their activation status. In this study, to address optimal status of DC/tumor to induce efficient antigen-specific cytotoxic T lymphocytes (CTLs), we have created various types of DC/tumor: 1) un-activated DC/tumor; 2) penicillin-killed Streptococcus pyogenes (OK-432; TLR4 agonist)-activated DC/tumor; 3) protein-bound polysaccharides isolated from Coriolus versicolor (PSK; TLR2 agonist)-activated DC/tumor; and 4) Combined OK-432- and PSK-activated DC/tumor. Moreover, we assessed the effects of TGF-β1 derived from DC/tumor on the induction of MUC1-specific CTLs. Combined TLR2- and TLR4-activated DC/tumor overcame immune-suppressive effect of TGF-β1 in comparison to those single activated or un-activated DC/tumor as demonstrated by: 1) up-regulation of MHC class II and CD86 expression on DC/tumor; 2) increased fusion efficiency; 3) increased production of fusions derived IL-12p70; 4) activation of CD4+ and CD8+ T cells that produce high levels of IFN-γ; 5) augmented induction of CTL activity specific for MUC1; and 6) superior efficacy in inhibiting CD4+CD25+Foxp3+ T cell generation. However, DC/tumor-derived TGF-β1 reduced the efficacy of DC/tumor vaccine in vitro. Incorporating combined TLRs-activation and TGF-β1-blockade of DC/tumor may enhance the effectiveness of DC/tumor-based cancer vaccines and have the potential applicability to the field of adoptive immunotherapy.


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 | 2012

Immunotherapy synergizes with chemotherapy targeting pancreatic cancer

Shigeo Koido; Sadamu Homma; Akitaka Takahara; Yoshihisa Namiki; Hideo Komita; Kan Uchiyama; Masaki Ito; Jianlin Gong; Toshifumi Ohkusa; Hisao Tajiri

Pancreatic adenocarcinoma is a devastating disease and has an extremely poor prognosis. Despite aggressive treatment approaches, including surgery, chemotherapy and radiation, the overall 5‐year survival rate is less than 5%. Therefore, the development of therapeutic strategies for advanced pancreatic cancer has traditionally been considered particularly challenging to improve clinical outcomes. Although immunotherapy that is designed to target tumor-associated antigens (TAAs) is a promising treatment approach, immunotherapy alone is limited by the number of cytotoxic T lymphocytes (CTLs) able to penetrate the large established pancreatic tumor. Even if large numbers of antigen-specific polyclonal CTLs were generated in vitro and injected into the patients, CTLs cannot penetrate into tumor sites because of stroma cells such as cancer-associated fibroblasts, tolerogenic dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), immunosuppressive tumor-associated macrophages and Tregs. Moreover, these cells produce immunosuppressive cytokines such as IL-10 and TGF-β; thus, clinical responses by immunotherapy alone cannot induce efficient antitumor immunity in patients with advanced pancreatic cancer. On the other hand, cytotoxic chemotherapy is well known to blunt immune responses, because of its toxicity for dividing cells including peripheral lymphoid tissue as well as the bone marrow. However, increasing evidence has been mounting to suggest that immunotherapy has the possibility of achieving better success when used in combination with chemotherapy [1]. For example, necrotic or apoptotic tumor cells induced by chemotherapy can be phagocytosed by DCs that are potent antigen-presenting cells, processed and presented to immune lymphocytes, followed by induction of antitumor immune responses. Different chemotherapeutic agents may kill tumor cells through an apparently homogeneous apoptotic pathway. Of note, treatment of pancreatic cancer cells with a standard cytotoxic agent for pancreatic cancer, gemcitabine, results in enhanced cross-presentation of TAAs by DCs and CTL induction [2]. Moreover, gemcitabine can also inhibit Tregs, B cells and MDSCs [2,3], but induce the proliferation of DCs [4]. These phenomena suggest that gemcitabine induces efficient CTL responses, improves the penetration of CTLs into the tumor parenchyma, and enhances tumor cell sensitivity to lyze antigen-specific CTLs. Thus, immunotherapy may mediate a potent antitumor effect when c ombined with chemotherapy.


Journal of Immunotherapy | 2010

Combined treatment with dendritic cells and 5-fluorouracil elicits augmented NK cell-mediated antitumor activity through the tumor necrosis factor-alpha pathway.

Eijiro Nagasaki; Akitaka Takahara; Shigeo Koido; Yukiko Sagawa; Keisuke Aiba; Hisao Tajiri; Hideo Yagita; Sadamu Homma

Antitumor effects and mechanism of combined therapy with a dendritic cell (DC) vaccine and fluorouracil (5-FU) were investigated. Cytotoxic activity against MC38 cells, untreated or pretreated with 5-FU, was examined in splenocytes from mice inoculated with DCs: DCs pulsed with MC38 lysate or treated with LPS or both and untreated DCs. Inoculation with all types of DCs induced the significant cytotoxic activity of splenocytes, and pretreatment of MC38 cells with 5-FU significantly enhanced the cytotoxic activity of splenocytes. Depletion of natural killer (NK) cells, but not of CD8+ or CD4+ T cells, in the splenocytes from DC (without MC38 lysate-pulse or LPS treatment thereafter)-inoculated mice decreased the cytotoxic activity. The cytotoxic effect was eliminated by treatment with a monoclonal antibody (mAb) against tumor necrosis factor (TNF)-α and was partially inhibited by concanamycin A. Inoculation of mice with DCs upregulated TNFα expression on NK cells. MC38 cells pretreated with 5-FU exhibited enhanced expression of procaspase 8 and efficiently underwent apoptosis by TNF-α with activation of caspase 8. Although treatment with 5-FU upregulated Rae-1 expression on MC38 cells, the NK-cell–mediated cytotoxic activity was not suppressed by treatment with an anti–Rae-1 mAb or an antinatural killer group 2D mAb or both. These results indicate that combined therapy with a DC vaccine and 5-FU is a promising strategy for cancer treatment mediated by the tumoricidal activity of NK cells through the TNF-α pathway.

Collaboration


Dive into the Akitaka Takahara's collaboration.

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

Sadamu Homma

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Toshifumi Ohkusa

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kan Uchiyama

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hideo Komita

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eijiro Nagasaki

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge