研究者業績

冨澤 聡史

トミザワ サトシ  (Satoshi Tomizawa)

基本情報

所属
千葉大学 大学院医学研究院臓器制御外科学講座 大学院生

研究者番号
80895839
J-GLOBAL ID
202201006818774558
researchmap会員ID
R000040828

論文

 9
  • Satoshi Tomizawa, Shigetsugu Takano, Ryotaro Eto, Tsukasa Takayashiki, Satoshi Kuboki, Masayuki Ohtsuka
    Cancer cell international 23(1) 155-155 2023年8月3日  
    PURPOSE: Semaphorins, axon guidance cues in neuronal network formation, have been implicated in cancer progression. We previously identified semaphorin 3 C (SEMA3C) as a secreted protein overexpressed in pancreatic ductal adenocarcinoma (PDAC). We, therefore, hypothesized that SEMA3C supports PDAC progression. In this study, we aimed to investigate the clinical features of SEMA3C, especially its association with chemo-resistance and peritoneal dissemination. METHODS: In resected PDAC tissues, we assessed the relationship between SEMA3C expression and clinicopathological features by immunohistochemistry. In vitro studies, we have shown invasion assay, pancreatosphere formation assay, colony formation assay, cytotoxicity assay, and activation of SEMA3C downstream targets (c-Met, Akt, mTOR). In vivo, we performed a preclinical trial to confirm the efficacy of SEMA3C shRNA knockdown and Gemcitabine and nab-Paclitaxel (GnP) in an orthotopic transplantation mouse model and in peritoneal dissemination mouse model. RESULTS: In resected PDAC tissues, SEMA3C expression correlated with invasion and peritoneal dissemination after surgery. SEMA3C promoted cell invasion, self-renewal, and colony formation in vitro. We further demonstrated that SEMA3C knockdown increased Gem-induced cytotoxicity by suppressing the activation of the Akt/mTOR pathway via the c-Met receptor. Combination therapy with SEMA3C knockdown and GnP reduced tumor growth and peritoneal dissemination. CONCLUSIONS: SEMA3C enhances peritoneal dissemination by regulating putative cancer stemness and Gem resistance and activates phosphorylation of the Akt/mTOR pathway via c-Met. Our findings provide a new avenue for therapeutic strategies in regulating peritoneal dissemination during PDAC progression.
  • 冨澤 聡史, 小西 孝宜, 高野 重紹, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 細川 勇, 三島 敬, 西野 仁恵, 仲田 真一郎, 大塚 将之
    膵臓 38(3) A403-A403 2023年7月  
  • 冨澤 聡史, 高野 重紹, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 細川 勇, 三島 敬, 小西 孝宜, 西野 仁恵, 仲田 真一郎, 大塚 将之
    日本外科学会定期学術集会抄録集 123回 SF-3 2023年4月  
  • Yoji Miyahara, Shigetsugu Takano, Kazuyuki Sogawa, Satoshi Tomizawa, Katsunori Furukawa, Tsukasa Takayashiki, Satoshi Kuboki, Masayuki Ohtsuka
    Cancer science 113(8) 2548-2559 2022年5月28日  
    Glycoproteins produced by tumor cells are involved in cancer progression, metastasis, and the immune response, and serve as possible therapeutic targets. Considering the dismal outcomes of pancreatic ductal adenocarcinoma (PDAC) due to its unique tumor microenvironment, which is characterized by low antitumor T-cell infiltration, we hypothesized that tumor-derived glycoproteins may serve as regulating the tumor microenvironment. We used glycoproteomics with tandem mass tag labeling to investigate the culture media of three human PDAC cell lines, and attempted to identify the key secreted proteins from PDAC cells. Among the identified glycoproteins, prosaposin (PSAP) was investigated for its functional contribution to PDAC progression. PSAP is highly expressed in various PDAC cell lines; however, knockdown of intrinsic PSAP expression did not affect the proliferation and migration capacities. Based on the immunohistochemistry of resected human PDAC tissues, high PSAP expression was associated with poor prognosis in patients with PDAC. Notably, tumors with high PSAP expression showed significantly lower CD8+ T-cell infiltration than those with low PSAP expression. Furthermore, PSAP stimulation decreased the proportion of CD8+ T cells in peripheral blood monocytes. Finally, in an orthotopic transplantation model, the number of CD8+ T cells in the PSAP shRNA groups was significantly increased, resulting in a decreased tumor volume compared with that in the control shRNA group. PSAP suppresses CD8+ T-cell infiltration, leading to the promotion of PDAC progression. However, further studies are warranted to determine whether this study contributes to the development of a novel immunomodulating therapy for PDAC.
  • 冨澤 聡史, 高野 重紹, 古川 勝規, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 細川 勇, 三島 敬, 小西 孝宜, 西野 仁恵
    日本外科学会定期学術集会抄録集 122回 SF-2 2022年4月  
  • 冨澤 聡史, 高野 重紹, 賀川 真吾, 古川 勝規, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 細川 勇, 三島 敬, 小西 孝宜, 大塚 将之
    膵臓 36(3) A279-A279 2021年8月  
  • Kosuke Sasaki, Shigetsugu Takano, Satoshi Tomizawa, Yoji Miyahara, Katsunori Furukawa, Tsukasa Takayashiki, Satoshi Kuboki, Mamoru Takada, Masayuki Ohtsuka
    Journal of experimental & clinical cancer research : CR 40(1) 212-212 2021年6月24日  
    BACKGROUND: Recent studies indicate that complement plays pivotal roles in promoting or suppressing cancer progression. We have previously identified C4b-binding protein α-chain (C4BPA) as a serum biomarker for the early detection of pancreatic ductal adenocarcinoma (PDAC). However, its mechanism of action remains unclear. Here, we elucidated the functional roles of C4BPA in PDAC cells and the tumor microenvironment. METHODS: We assessed stromal C4BPA, the C4BPA binding partner CD40, and the number of CD8+ tumor-infiltrating lymphocytes in resected human PDAC tissues via immunohistochemical staining. The biological functions of C4BPA were investigated in peripheral blood mononuclear cells (PBMCs) and human PDAC cell lines. Mouse C4BPA (mC4BPA) peptide, which is composed of 30 amino acids from the C-terminus and binds to CD40, was designed for further in vitro and in vivo experiments. In a preclinical experiment, we assessed the efficacy of gemcitabine plus nab-paclitaxel (GnP), dual immune checkpoint blockades (ICBs), and mC4BPA peptide in a mouse orthotopic transplantation model. RESULTS: Immunohistochemical analysis revealed that high stromal C4BPA and CD40 was associated with favorable PDAC prognosis (P=0.0005). Stromal C4BPA strongly correlated with the number of CD8+ tumor-infiltrating lymphocytes (P=0.001). In in vitro experiments, flow cytometry revealed that recombinant human C4BPA (rhC4BPA) stimulation increased CD4+ and CD8+ T cell numbers in PBMCs. rhC4BPA also promoted the proliferation of CD40-expressing PDAC cells. By contrast, combined treatment with gemcitabine and rhC4BPA increased PDAC cell apoptosis rate. mC4BPA peptide increased the number of murine T lymphocytes in vitro and the number of CD8+ tumor-infiltrating lymphocytes surrounding PDAC tumors in vivo. In a preclinical study, GnP/ICBs/mC4BPA peptide treatment, but not GnP treatment, led to the accumulation of a greater number of CD8+ T cells in the periphery of PDAC tumors and to greater tumor regression than did control treatment. CONCLUSIONS: These findings demonstrate that the combination of GnP therapy with C4BPA inhibits PDAC progression by promoting antitumor T cell accumulation in the tumor microenvironment.
  • 冨澤 聡史, 高野 重紹, 賀川 真吾, 古川 勝規, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 細川 勇, 三島 敬, 大塚 将之
    日本臨床外科学会雑誌 81(12) 2553-2553 2020年12月  
  • 冨澤 聡史, 高野 重紹, 吉富 秀幸, 賀川 真吾, 古川 勝規, 高屋敷 吏, 久保木 知, 鈴木 大亮, 酒井 望, 三島 敬, 中臺 英里
    膵臓 35(3) A342-A342 2020年7月  

MISC

 2