Network


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

Hotspot


Dive into the research topics where Satoshi Takashima is active.

Publication


Featured researches published by Satoshi Takashima.


Cancer Science | 2006

Wilms’ tumor gene WT1 17AA(–)/KTS(–) isoform induces morphological changes and promotes cell migration and invasion in vitro

Tanyarat Jomgeow; Yusuke Oji; Naoko Tsuji; Yoko Ikeda; Ken Ito; Asako Tsuda; Tsutomu Nakazawa; Naoya Tatsumi; Nao Sakaguchi; Satoshi Takashima; Toshiaki Shirakata; Sumiyuki Nishida; Naoki Hosen; Manabu Kawakami; Akihiro Tsuboi; Yoshihiro Oka; Kazuyuki Itoh; Haruo Sugiyama

The wild‐type Wilms’ tumor gene WT1 is overexpressed in human primary leukemia and in a wide variety of solid cancers. All of the four WT1 isoforms are expressed in primary cancers and each is considered to have a different function. However, the functions of each of the WT1 isoforms in cancer cells remain unclear. The present study demonstrated that constitutive expression of the WT1 17AA(–)/KTS(–) isoform induces morphological changes characterized by a small‐sized cell shape in TYK‐nu.CP‐r (TYK) ovarian cancer cells. In the WT1 17AA(–)/KTS(–) isoform‐transduced TYK cells, cell–substratum adhesion was suppressed, and cell migration and in vitro invasion were enhanced compared to that in mock vector‐transduced TYK cells. Constitutive expression of the WT1 17AA(–)/KTS(–) isoform also induced morphological changes in five (one gastric, one esophageal, two breast and one fibrosarcoma) of eight cancer cell lines examined. No WT1 isoforms other than the WT1 17AA(–)/KTS(–) isoform induced the phenotypic changes. A decrease in α‐actinin 1 and cofilin expression and an increase in gelsolin expression were observed in WT1 17AA(–)/KTS(–) isoform‐transduced TYK cells. In contrast, co‐expression of α‐actinin 1 and cofilin or knockdown of gelsolin expression by small interfering RNA restored WT1 17AA(–)/KTS(–) isoform‐transduced TYK cells to a phenotype that was comparable to that of the parent TYK cells. These results indicated that the WT1 17AA(–)/KTS(–) isoform exerted its oncogenic functions through modulation of cytoskeletal dynamics. The present results may provide a novel insight into the signaling pathway of the WT1 gene for its oncogenic functions. (Cancer Sci 2006; 97: 259–270)


Blood Cancer Journal | 2013

Maintenance of complete remission after allogeneic stem cell transplantation in leukemia patients treated with Wilms tumor 1 peptide vaccine

Tetsuo Maeda; Naoki Hosen; Kentaro Fukushima; Akihiro Tsuboi; Soyoko Morimoto; Toshimitsu Matsui; Hiroshi Sata; Jiro Fujita; Kana Hasegawa; Sumiyuki Nishida; Jun Nakata; Yoshiki Nakae; Satoshi Takashima; Hiroko Nakajima; Fumihiro Fujiki; Naoya Tatsumi; T Kondo; Masayuki Hino; Yusuke Oji; Yoshihiro Oka; Yuzuru Kanakura; Atsushi Kumanogoh; Haruo Sugiyama

The prognosis of patients after allogeneic hematopoietic stem cell transplantation (HSCT) is still not satisfactory because, while treatment-related mortalities have decreased, relapse after HSCT remains a major concern. The effectiveness of allogeneic HSCT for hematological malignancies is the result of immunologic rejection of recipient leukemia cells by donor T cells, known as the graft-versus-leukemia (GVL) effect.1 It is thus obviously important to be able to exploit the GVL effect while minimizing graft-versus-host disease (GVHD). A targeted anti-leukemic immunotherapy, such as use of a leukemia vaccine,2 is a promising strategy to boost the GVL effect. Wilms tumor1 (WT1) protein is one of the best targets for leukemia vaccines. Overexpression of the wild-type WT1 gene has been detected in all types of human leukemia.3, 4, 5 We performed a phase I clinical study of immunotherapy targeting the WT1 protein in patients with leukemia, and were able to show that WT1 vaccination was safe and could induce WT1-specific cytotoxic T lymphocyte (CTL).6 Furthermore, reduction of minimal residual disease and long-lasting complete remission (CR) was observed in some leukemia patients who were given the WT1 vaccine.7 This report presents the results of phase I clinical study of WT1 vaccination for HLA-A*2402-positivie post-HSCT patients who were at high risk of relapse (HSCT in non-CR and 2nd HSCT for post-transplant relapse) or had already relapsed. The HLA-A*2402-restricted modified 9-mer WT1 peptide (amino acids 235–243 CYTWNQMNL)8 was emulsified with Montanide ISA51 adjuvant. Patients were intradermally injected with 1.0 mg (three patients: UPNs 1, 4 and 6) or 3.0 mg (other six patients) of WT1 peptide four times weekly. When no adverse effects and no obvious disease progression were observed after the fourth injection, further WT1 vaccinations at 2-week intervals were administered. Nine patients (five with acute myeloid leukemia (AML), one each with acute lymphoblastic leukemia, chronic myelomonocytic leukemia, multiple myeloma and T-cell lymphoblastic lymphoma) were enrolled in this study (Supplementary Tables 1 and 2). Local inflammatory response was observed at the vaccine injection sites of all patients. One patient (UPN5) suffered mild hypoxia (PaO2 65 mm Hg at room air) and restrictive pulmonary dysfunction (FEV1.0 40%) 65 days after the start of WT1 vaccination (day 199 after HSCT; Figure 1a). He was diagnosed with bronchioleitis obliterans (BO), which was a symptom of chronic GVHD. The patient recovered soon after administration of inhaled steroids. While early and sudden discontinuation of prednisolone and tacrolimus (day 103 after HSCT) were considered to be the reason for development of BO, the possibility of an association between BO and WT1 vaccination cannot be entirely ruled out. In other eight patients, no severe toxicities related to WT1 vaccine were observed (Table1). Figure 1 Clinical course of patients who attained CR after the start of WT1 peptide vaccination. (a) Clinical course of UPN5 who achieved CR after administration of WT1 vaccine but stopped vaccination because of the development of bronchioleitis obliterans. ( ... Table 1 Patient outcomes Three AML patients (UPN1–3), who had undergone HSCT in non-CR, started WT1 vaccine in CR (Supplementary Tables 1 and 2). They started WT1 vaccination on post-HSCT days 141, 76 and 93 and have remained in CR for 1038, 973 and 662 days, respectively (as of 8 April 2013; Table1), suggesting the potential of WT1 vaccination as a maintenance therapy after HSCT. Six patients started WT1 vaccination in non-CR and two of them became CR after WT1 vaccination. One B-ALL patient (UPN4) with MLL-AF4 underwent bone marrow transplantation from an HLA-matched unrelated donor during the first CR. On post-HSCT day 111, MLL-AF4 and WT1 mRNA in peripheral blood (PB) had increased to 16 000 and 15 000 copies/μg RNA, indicating that the disease had relapsed. Tacrolimus and prednisolone doses were tapered off to induce GVL effects. The expression levels of MLL-AF4 and WT1 mRNA in PB had decreased to 2700 and 190 copies/μg RNA by day 132, and WT1 vaccination was started on day 133. MLL-AF4 mRNA had become undetectable by day 146, and had never appeared until post-HSCT day 1312 (day 1179 after the start of WT1 vaccination as of 8 April 2013; Figure 1b). Skin tumors appeared in UPN5 (AML-M5) on post-HSCT day 103 and was diagnosed by biopsy as leukemia relapse. Tacrolimus was discontinued on day103, and WT1 vaccination was started on day 130. Cutaneous tumors had regressed 2 weeks after the start of WT1 vaccination, but vaccination was terminated after the second injection because of the development of BO as described earlier (Figure 1a). This patient has been remained in CR until post-HSCT day 972 (day 842 after the start of WT1 vaccination at 8 April 2013). While the exact contribution of the vaccination effect to the disease remission in addition to the GVL effect was unclear, the fact that both of these two patients still have remained in CR until now is encouraging to continue this trial. In the following phase II trials, the enumeration of WT1-specific CTLs should be performed more frequently after the start of vaccination to clarify the relationship between the effect of WT1 peptide vaccination and leukemia regression. WT1 (a natural 9-mer WT1 peptide) HLA-A*2402 tetramer assays could be performed with peripheral blood mononuclear cell in seven of the nine patients to determine whether WT1235 peptide-specific CD8+ T cells had increased after WT1 vaccination. The gates for WT1 tetramer+ cells were drawn as <0.1% of CD8+ T cells were included in the tetramer-positive gate in multiple healthy individuals (Supplementary Figure 1A). WT1235 tetramer+ cells increased after the start of vaccination in three (UPNs1, 2 and 4) of the four patients who have remained in CR (Figure 1b and Supplementary Figure 1B). In the cases with progressive disease, continuous increase in the frequencies of WT1235 tetramer+ cells was not observed (Supplementary Figure 1B). Our results suggest that WT1 vaccination should be started when the leukemia burden is minimal. The timing of the start of WT1 vaccination may be also important. For the cases with good outcomes, WT1 vaccination was started 76–140 days after transplantation (UPNs1–5), and at later times (days 299–1815) for PD cases (UPNs 6–9). A lymphopenic environment a few months after transplantation may be favorable for rapid and extensive expansion of tumor antigen-specific CTLs. In summary, this report suggests that WT1 vaccine can be safely administrated for post-HSCT patients with hematological malignancies and has potential as a maintenance therapy. Clinical benefit of WT1 vaccination for post-HSCT patients will be evaluated in the subsequent phase II trials.


Expert Review of Hematology | 2014

Vaccination strategies to improve outcome of hematopoietic stem cell transplant in leukemia patients: early evidence and future prospects

Naoki Hosen; Tetsuo Maeda; Yoshiko Hashii; Akihiro Tsuboi; Sumiyuki Nishida; Jun Nakata; Yoshiki Nakae; Satoshi Takashima; Yusuke Oji; Yoshihiro Oka; Atsushi Kumanogoh; Haruo Sugiyama

Allogeneic hematopoietic stem cell transplantation (HSCT) has largely improved the prognosis of leukemia patients. However, relapse is still a major concern. One promising option for the prevention of relapse is vaccination therapy. The post allogeneic HSCT period provides a unique platform for vaccination, because tumor burden is minimal, lymphopenic condition allows for rapid expansion of cytotoxic T cells (CTLs), donor-derived CTLs are not exhausted and inflammatory condition is caused by allo reactions. Tumor cells, dendritic cells and peptides have been used as vaccines targeting leukemia-associated antigens or minor histocompatibility antigens. Clinical trials with several types of vaccines for post-HSCT patients showed that the vaccination induced immunological response and might benefit patients with minimal residual disease, while their effect in patients with advanced disease were limited. To enhance the effect, vaccination in combination with other immune-modulatory drugs such as checkpoint antibodies is now being considered.


International Journal of Cancer | 2016

Association of WT1 IgG antibody against WT1 peptide with prolonged survival in glioblastoma multiforme patients vaccinated with WT1 peptide.

Yusuke Oji; Naoya Hashimoto; Akihiro Tsuboi; Yui Murakami; Miki Iwai; Naoki Kagawa; Yasuyoshi Chiba; Shuichi Izumoto; Olga A. Elisseeva; Ryo Ichinohasama; Junichi Sakamoto; Satoshi Morita; Hiroko Nakajima; Satoshi Takashima; Yoshiki Nakae; Jun Nakata; Manabu Kawakami; Sumiyuki Nishida; Naoki Hosen; Fumihiro Fujiki; Soyoko Morimoto; Mayuko Adachi; Masahiro Iwamoto; Yoshihiro Oka; Toshiki Yoshimine; Haruo Sugiyama

We previously evaluated Wilms’ tumor gene 1 (WT1) peptide vaccination in a large number of patients with leukemia or solid tumors and have reported that HLA‐A*24:02 restricted, 9‐mer WT1‐235 peptide (CYTWNQMNL) vaccine induces cellular immune responses and elicits WT1‐235‐specific cytotoxic T lymphocytes (CTLs). However, whether this vaccine induces humoral immune responses to produce WT1 antibody remains unknown. Thus, we measured IgG antibody levels against the WT1‐235 peptide (WT1‐235 IgG antibody) in patients with glioblastoma multiforme (GBM) receiving the WT1 peptide vaccine. The WT1‐235 IgG antibody, which was undetectable before vaccination, became detectable in 30 (50.8%) of a total of 59 patients during 3 months of WT1 peptide vaccination. The dominant WT1‐235 IgG antibody subclass was Th1‐type, IgG1 and IgG3. WT1‐235 IgG antibody production was significantly and positively correlated with both progression‐free survival (PFS) and overall survival (OS). Importantly, the combination of WT1‐235 IgG antibody production and positive delayed type‐hypersensitivity (DTH) to the WT1‐235 peptide was a better prognostic marker for long‐term OS than either parameter alone. These results suggested that WT1‐235 peptide vaccination induces not only WT1‐235‐specific CTLs as previously described but also WT1‐235‐specific humoral immune responses associated with antitumor cellular immune response. Our results indicate that the WT1 IgG antibody against the WT1 peptide may be a useful predictive marker, with better predictive performance in combination with DTH to WT1 peptide, and provide a new insight into the antitumor immune response induction in WT1 peptide vaccine‐treated patients.


PLOS ONE | 2015

An Immunocompetent Mouse Model for MLL/AF9 Leukemia Reveals the Potential of Spontaneous Cytotoxic T-Cell Response to an Antigen Expressed in Leukemia Cells

Kana Hasegawa; Satomi Tanaka; Fumihiro Fujiki; Soyoko Morimoto; Hiroko Nakajima; Naoya Tatsumi; Jun Nakata; Satoshi Takashima; Sumiyuki Nishida; Akihiro Tsuboi; Yoshihiro Oka; Yusuke Oji; Atsushi Kumanogoh; Haruo Sugiyama; Naoki Hosen

Leukemia differs substantially with respect to stromal milieu from tumors that progress locally as solid masses, and the physiological importance of immunosurveillance in leukemia remains unclear. However, currently available mouse leukemia models have critical limitations in the context of analyzing immunological regulation of leukemia development. In this study, we transferred mouse MLL/AF9 leukemia-initiating cells into immunocompetent recipient mice without any pre-conditioning such as irradiation, and then analyzed the spontaneous T cell response to an immunogenic antigen expressed in leukemia cells. When the minimum numbers of leukemia-initiating cells for engraftment were transferred, leukemia cells were eradicated by the adaptive immune response in most, if not all, wild-type mice, but not in Rag2 -/- recipient mice, which lack adaptive immunity. By contrast, mice transplanted with larger numbers of leukemia cells always developed leukemia. In mice with advanced leukemia, antigen-specific CTLs were also expanded, but were unresponsive to antigen stimulation and expressed high levels of PD-1 and LAG-3. These results provide the first clear demonstration that the spontaneous CTL response to a tumor-cell antigen has the potential to eradicate leukemia, whereas antigen-specific CTLs are exhausted in animals with advanced leukemia. This immunocompetent mouse leukemia model provides a useful platform for developing effective immunotherapies against leukemia.


Future Science OA | 2016

Syndecan-4 as a biomarker to predict clinical outcome for glioblastoma multiforme treated with WT1 peptide vaccine

Satoshi Takashima; Yoshihiro Oka; Fumihiro Fujiki; Soyoko Morimoto; Hiroko Nakajima; Yoshiki Nakae; Jun Nakata; Sumiyuki Nishida; Naoki Hosen; Naoya Tatsumi; Kenji Mizuguchi; Naoya Hashimoto; Yusuke Oji; Akihiro Tsuboi; Atsushi Kumanogoh; Haruo Sugiyama

Aim: In cancer immunotherapy, biomarkers are important for identification of responsive patients. This study was aimed to find biomarkers that predict clinical outcome of WT1 peptide vaccination. Materials & methods: Candidate genes that were expressed differentially between long- and short-term survivors were identified by cDNA microarray analysis of peripheral blood mononuclear cells that were extracted from 30 glioblastoma patients (discovery set) prior to vaccination and validated by quantitative RT-PCR using discovery set and different 23 patients (validation set). Results: SDC-4 mRNA expression levels distinguished between the long- and short-term survivors: 1-year survival rates were 64.0 and 18.5% in SDC4-low and -high patients, respectively. Conclusion: SDC-4 is a novel predictive biomarker for the efficacy of WT1 peptide vaccine.


International Journal of Cancer | 2018

WT1 peptide-based immunotherapy for advanced thymic epithelial malignancies: WT1 peptide vaccine for thymic malignancies

Yusuke Oji; Masayoshi Inoue; Yoshito Takeda; Naoki Hosen; Yasushi Shintani; Manabu Kawakami; Takuya Harada; Yui Murakami; Miki Iwai; Mari Fukuda; Sumiyuki Nishida; Jun Nakata; Yoshiki Nakae; Satoshi Takashima; Toshiaki Shirakata; Hiroko Nakajima; Kana Hasegawa; Hiroshi Kida; Takashi Kijima; Soyoko Morimoto; Fumihiro Fujiki; Akihiro Tsuboi; Eiichi Morii; Satoshi Morita; Junichi Sakamoto; Atsushi Kumanogoh; Yoshihiro Oka; Meinoshin Okumura; Haruo Sugiyama

Thymic epithelial tumors are rare malignancies, and no optimal therapeutic regimen has been defined for patients with advanced disease. Patients with advanced thymic epithelial tumors, which were resistant or intolerable to prior therapies, were eligible for this study. Patients received 9 mer‐WT1‐derived peptide emulsified with Montanide ISA51 adjuvant via intradermal administration once a week as a monotherapy. After the 3‐month‐protocol treatment, the treatment was continued mostly at intervals of 2–4 weeks until disease progression or intolerable adverse events occurred. Of the 15 patients enrolled, 11 had thymic carcinoma (TC) and 4 had invasive thymoma (IT). Median period from diagnosis to the start of treatment was 13.3 and 65.5 months for TC and IT, respectively. No patients achieved a complete or partial response. Of the 8 evaluable TC patients, 6 (75.0%) had stable disease (SD) and 2 had progressive disease (PD). Of the 4 evaluable IT patients, 3 (75.0%) had SD and 1 (25.0%) had PD. Median period of monotherapy treatment was 133 and 683 days in TC and IT patients, respectively. No severe adverse events occurred during the 3‐month‐protocol treatment. As adverse events in long responders, thymoma‐related autoimmune complications, pure red cell aplasia and myasthenia gravis occurred in two IT patients. Cerebellar hemorrhage developed in a TC patient complicated with Von Willebrand disease. Induction of WT1‐specific immune responses was observed in the majority of the patients. WT1 peptide vaccine immunotherapy may have antitumor potential against thymic malignancies.


PLOS ONE | 2016

Glycosylation Status of CD43 Protein Is Associated with Resistance of Leukemia Cells to CTL-Mediated Cytolysis

Kana Hasegawa; Satomi Tanaka; Fumihiro Fujiki; Soyoko Morimoto; Katsuhiko Nakano; Hiroko Kinoshita; Atsushi Okumura; Yuka Fujioka; Rika Urakawa; Hiroko Nakajima; Naoya Tatsumi; Jun Nakata; Satoshi Takashima; Sumiyuki Nishida; Akihiro Tsuboi; Yoshihiro Oka; Yusuke Oji; Eiji Miyoshi; Takako Hirata; Atsushi Kumanogoh; Haruo Sugiyama; Naoki Hosen

To improve cancer immunotherapy, it is important to understand how tumor cells counteract immune-surveillance. In this study, we sought to identify cell-surface molecules associated with resistance of leukemia cells to cytotoxic T cell (CTL)-mediated cytolysis. To this end, we first established thousands of monoclonal antibodies (mAbs) that react with MLL/AF9 mouse leukemia cells. Only two of these mAbs, designated R54 and B2, bound preferentially to leukemia cells resistant to cytolysis by a tumor cell antigen–specific CTLs. The antigens recognized by these mAbs were identified by expression cloning as the same protein, CD43, although their binding patterns to subsets of hematopoietic cells differed significantly from each other and from a pre-existing pan-CD43 mAb, S11. The epitopes of R54 and B2, but not S11, were sialidase-sensitive and expressed at various levels on leukemia cells, suggesting that binding of R54 or B2 is associated with the glycosylation status of CD43. R54high leukemia cells, which are likely to express sialic acid-rich CD43, were highly resistant to CTL-mediated cytolysis. In addition, loss of CD43 in leukemia cells or neuraminidase treatment of leukemia cells sensitized leukemia cells to CTL-mediated cell lysis. These results suggest that sialic acid-rich CD43, which harbors multiple sialic acid residues that impart a net negative surface charge, protects leukemia cells from CTL-mediated cell lysis. Furthermore, R54high or B2high leukemia cells preferentially survived in vivo in the presence of adaptive immunity. Taken together, these results suggest that the glycosylation status of CD43 on leukemia is associated with sensitivity to CTL-mediated cytolysis in vitro and in vivo. Thus, regulation of CD43 glycosylation is a potential strategy for enhancing CTL-mediated immunotherapy.


Archive | 2016

WT1 Peptide Vaccine for the Treatment of Malignancies: Its Development, Recent Progress, and Future Perspectives

Yoshihiro Oka; Akihiro Tsuboi; Hiroko Nakajima; Fumihiro Fujiki; Sumiyuki Nishida; Soyoko Morimoto; Jun Nakata; Yoshiki Nakae; Satoshi Takashima; Naoya Tatsumi; Naoki Hosen; Yusuke Oji; Haruo Sugiyama

Wilms’ tumor gene (WT1) possesses oncogenic functions and is expressed in various hematological malignancies and solid cancers, and the gene product WT1 protein is highly immunogenic, which indicates that WT1 should be a promising target antigen for cancer immunotherapy. The identification of human WT1 cytotoxic T lymphocyte (CTL) epitopes and the demonstration using a mouse model that WT1 could serve in vivo as a target antigen for cancer immunotherapy were reported in 2000. Based on these findings, clinical trials for WT1 peptide vaccine were started. The early clinical trials demonstrated that the WT1 peptide vaccination could induce WT1-specific immunological response such as an increase in WT1-specific CTL frequency, resulting in occurrence of clinical response such as a decrease in leukemia/solid tumor load, which strongly suggested the therapeutic potential of the WT1 peptide vaccine for the treatment of malignancies. A review article published in 2009 in a prestigious journal gave WT1 the highest ranking as a target antigen for cancer immunotherapy. Now, cases which showed immunological and/or clinical responses with treatment by WT1 peptide vaccine are being accumulated. Some of the recent clinical trials showed noteworthy results, such as the demonstration that WT1 peptide vaccination may lead acute myeloid leukemia patients with minimal residual disease to a cure and that the vaccination may prevent relapse of patients with hematological malignancies who have received allogeneic hematopoietic stem cell transplantation but are at high risk of relapse. In addition, clinical usefulness of the WT1 peptide vaccine combined with chemotherapy drugs or molecular target-based drugs was also suggested. Continuing progress of WT1-targeting immunotherapy, a translational research based on basic research, should lead to innovative development of cancer immunotherapy. Furthermore, comprehensive analysis of the samples obtained from the patients treated with the WT1-targeting immunotherapy, a reverse-translational research, should contribute to the elucidation of cancer immunity mechanisms.


Cancer Science | 2004

Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma

Yusuke Oji; Shoji Nakamori; Masahiro Fujikawa; Shin-ichi Nakatsuka; Asumi Yokota; Naoya Tatsumi; Sakie Abeno; Ai Ikeba; Satoshi Takashima; Masanori Tsujie; Hirofumi Yamamoto; Masato Sakon; Riichiro Nezu; Kiyoshi Kawano; Sumiyuki Nishida; Kazuhiro Ikegame; Manabu Kawakami; Akihiro Tsuboi; Yoshihiro Oka; Kiyoshi Yoshikawa; Katsuyuki Aozasa; Morito Monden; Haruo Sugiyama

Collaboration


Dive into the Satoshi Takashima's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge