研究者業績

本橋 新一郎

モトハシ シンイチロウ  (Shinichiro Motohashi)

基本情報

所属
千葉大学 大学院医学研究院免疫細胞医学 教授
学位
博士(医学)(千葉大学)

研究者番号
60345022
J-GLOBAL ID
200901096314072885
researchmap会員ID
1000284756

外部リンク

学歴

 2

論文

 179
  • Ami Aoki, Chiaki Iwamura, Masahiro Kiuchi, Kaori Tsuji, Atsushi Sasaki, Takahisa Hishiya, Rui Hirasawa, Kota Kokubo, Sachiko Kuriyama, Atsushi Onodera, Tadanaga Shimada, Tetsutaro Nagaoka, Satoru Ishikawa, Akira Kojima, Haruki Mito, Ryota Hase, Yasunori Kasahara, Naohide Kuriyama, Sukeyuki Nakamura, Takashi Urushibara, Satoru Kaneda, Seiichiro Sakao, Osamu Nishida, Kazuhisa Takahashi, Motoko Y Kimura, Shinichiro Motohashi, Hidetoshi Igari, Yuzuru Ikehara, Hiroshi Nakajima, Takuji Suzuki, Hideki Hanaoka, Taka-Aki Nakada, Toshiaki Kikuchi, Toshinori Nakayama, Koutaro Yokote, Kiyoshi Hirahara
    Journal of clinical immunology 44(4) 104-104 2024年4月22日  
    PURPOSE: Auto-antibodies (auto-abs) to type I interferons (IFNs) have been identified in patients with life-threatening coronavirus disease 2019 (COVID-19), suggesting that the presence of auto-abs may be a risk factor for disease severity. We therefore investigated the mechanism underlying COVID-19 exacerbation induced by auto-abs to type I IFNs. METHODS: We evaluated plasma from 123 patients with COVID-19 to measure auto-abs to type I IFNs. We performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells from the patients with auto-abs and conducted epitope mapping of the auto-abs. RESULTS: Three of 19 severe and 4 of 42 critical COVID-19 patients had neutralizing auto-abs to type I IFNs. Patients with auto-abs to type I IFNs showed no characteristic clinical features. scRNA-seq from 38 patients with COVID-19 revealed that IFN signaling in conventional dendritic cells and canonical monocytes was attenuated, and SARS-CoV-2-specific BCR repertoires were decreased in patients with auto-abs. Furthermore, auto-abs to IFN-α2 from COVID-19 patients with auto-abs recognized characteristic epitopes of IFN-α2, which binds to the receptor. CONCLUSION: Auto-abs to type I IFN found in COVID-19 patients inhibited IFN signaling in dendritic cells and monocytes by blocking the binding of type I IFN to its receptor. The failure to properly induce production of an antibody to SARS-CoV-2 may be a causative factor of COVID-19 severity.
  • Mariko Takami, Takahiro Aoki, Katsuhiro Nishimura, Hidekazu Tanaka, Atsushi Onodera, Shinichiro Motohashi
    Cancer research communications 4(2) 446-459 2024年2月19日  査読有り
    UNLABELLED: Invariant natural killer T (iNKT) cells play an essential role in antitumor immunity by exerting cytotoxicity and producing massive amounts of cytokines. iNKT cells express invariant T-cell receptors (TCR) to recognize their cognate glycolipid antigens such as α-galactosylceramide (α-GalCer) presented on CD1d. We recently reported that iNKT cells recognize CD1d-negative leukemia cell line K562 in a TCR-dependent manner. However, it remains controversial how iNKT cells use TCRs to recognize and exhibit cytotoxic activity toward CD1d-negative tumors cells without CD1d restriction. Here, we report that iNKT cells exerted cytotoxicity toward K562 cells via a carried over anti-Vα24 TCR mAb from positive selection by magnetic bead sorting. We found that addition of the anti-Vα24Jα18 TCR mAb (6B11 mAb) rendered iNKT cells cytotoxic to K562 cells in an FcγRII (CD32)-dependent manner. Moreover, iNKT cells treated with 6B11 mAb became cytotoxic to other CD32+ cell lines (U937 and Daudi). In addition, iNKT cells treated with 6B11 mAb suppressed K562 cell growth in a murine xenograft model in vivo. These data suggest that anti-iNKT TCR mAb treatment of iNKT cells can be applied as a therapeutic strategy to treat CD32+ cancers such as leukemia, lymphoma, and lung cancer. SIGNIFICANCE: Our findings unveiled that iNKT cells recognize and kill CD1d-negative target tumors via the anti-iNKT TCR mAb bound to CD32 at the tumor site, thereby bridging iNKT cells and CD1d-negative tumors. These findings shed light on the therapeutic potential of anti-iNKT TCR mAbs in NKT cell-based immunotherapy to treat CD1d-negative CD32+ cancers.
  • Takahiro Aoki, Shinichiro Motohashi
    Critical reviews in oncogenesis 29(1) 1-9 2024年  査読有り
    Immune cell therapy has received attention in the clinical setting. However, current chimeric antigen receptor T cell therapies require individualized manufacturing based on patient cells, resulting in high costs and long processing times. Allogeneic immune cell therapy, which involves the use of immune cells from other donors, is emerging as a promising alternative that offers multiple advantages, including off-the-shelf availability, standardized manufacturing, and potentially stronger effector functions. Natural killer T (NKT) cells are a type of T cell that can be activated without being restricted by HLA, indicating their potential use in allogeneic cell immunotherapy. They exhibit cytotoxic activity against various cancer targets. However, their low frequency in blood limits their use in ex vivo amplification for treatment. This has led researchers to focus on allogeneic NKT cells as a potential treatment agent. In this study, we review the research on NKT cell-based immunotherapy and focus on the recent progress in clinical trials related to NKT cell-based immunotherapy worldwide. NKT cell-based therapy is not limited to specific cancer types and has been investigated in many ways worldwide over the past decade. Some clinical trials targeting NKT cells have shown promising results; however, the number of trials is low compared to those using T and natural killer cells. The use of allogeneic NKT cells may revolutionize the treatment of cancer and other diseases. However, further research and clinical trials are necessary to fully understand their efficacy, safety, and long-term benefits.
  • Hiroki Matsumoto, Hidemi Suzuki, Takahiro Yamanaka, Taisuke Kaiho, Atsushi Hata, Terunaga Inage, Takamasa Ito, Toshiko Kamata, Kazuhisa Tanaka, Yuichi Sakairi, Shinichiro Motohashi, Ichiro Yoshino
    Life (Basel, Switzerland) 13(10) 2023年10月11日  
    Antibody-mediated rejection (AMR) is a risk factor for chronic lung allograft dysfunction, which impedes long-term survival after lung transplantation. There are no reports evaluating the efficacy of the single use of anti-CD20 antibodies (aCD20s) in addition to calcineurin inhibitors in preventing AMR. Thus, this study aimed to evaluate the efficacy of aCD20 treatment in a murine orthotopic lung transplantation model. Murine left lung transplantation was performed using a major alloantigen strain mismatch model (BALBc (H-2d) → C57BL/6 (BL/6) (H-2b)). There were four groups: isograft (BL/6→BL/6) (Iso control), no-medication (Allo control), cyclosporine A (CyA) treated, and CyA plus murine aCD20 (CyA+aCD20) treated groups. Severe neutrophil capillaritis, arteritis, and positive lung C4d staining were observed in the allograft model and CyA-only-treated groups. These findings were significantly improved in the CyA+aCD20 group compared with those in the Allo control and CyA groups. The B cell population in the spleen, lymph node, and graft lung as well as the levels of serum donor-specific IgM and interferon γ were significantly lower in the CyA+aCD20 group than in the CyA group. Calcineurin inhibitor-mediated immunosuppression combined with aCD20 therapy effectively suppressed AMR in lung transplantation by reducing donor-specific antibodies and complement activation.
  • 飯沼 智久, 本橋 新一郎
    炎症と免疫 31(4) 302-305 2023年6月  
    NKT細胞はHLA拘束性をもたず,直接的にがん細胞に対して抗腫瘍効果を発揮するほか,IFN-γを産生して周囲の細胞傷害活性を増強する.筆者らはNKT細胞を活性化する糖脂質抗原であるアルファガラクトシルセラミドを使用して,がん免疫治療の検討をおこなってきた.しかし,免疫能の低下したがん患者のNKT細胞の利用には,その数の少なさや機能の低下など問題がある.その問題点を解決するため,理化学研究所でiPS細胞技術を用いて作成したiPS-NKT細胞を用いて,他家移植としての免疫細胞療法を2020年に医師主導治験として開始した.開発には造腫瘍試験やGMPグレードの達成など,さまざまな非臨床試験をおこなうことでハードルを乗り越えてきた.(著者抄録)
  • Ryo Koyama-Nasu, Motoko Y Kimura, Masahiro Kiuchi, Ami Aoki, Yangsong Wang, Yukiyoshi Mita, Ichita Hasegawa, Yukihiro Endo, Atsushi Onodera, Kiyoshi Hirahara, Shinichiro Motohashi, Toshinori Nakayama
    Cancer immunology research 2023年5月22日  査読有り
    Tumor-specific CD8+ T cells play a pivotal role in anti-tumor immunity and are a key target of immunotherapeutic approaches. Intratumoral CD8+ T cells are heterogeneous; Tcf1+ stem-like CD8+ T cells give rise to their cytotoxic progeny - Tim-3+ terminally differentiated CD8+ T cells. However, where and how this differentiation process occurs has not been elucidated. We herein show that terminally differentiated CD8+ T cells can be generated within tumor-draining lymph nodes (TDLNs) and that CD69 expression on tumor-specific CD8+ T cells controls its differentiation process through regulating the expression of the transcription factor TOX. In TDLNs, CD69 deficiency diminished TOX expression in tumor-specific CD8+ T cells, and consequently promoted generation of functional terminally differentiated CD8+ T cells. Anti-CD69 administration promoted the generation of terminally differentiated CD8+ T cells, and the combined use of anti-CD69 and anti-PD-1 showed an efficient anti-tumor effect. Thus, CD69 is an attractive target for cancer immunotherapy that synergizes with immune checkpoint blockade.
  • Takahiro Aoki, Shinichiro Motohashi, Haruhiko Koseki
    Inflammation and regeneration 43(1) 27-27 2023年5月12日  査読有り
    Invariant natural killer T (iNKT) cells are a subset of innate-like T cells restricted by a major histocompatibility complex (MHC) class I-like molecule, CD1d. iNKT cells express an invariant T cell receptor (TCR) encoded by Vα14 Jα18 in mice and Vα24 Jα18 in humans and are activated by recognizing glycolipid antigens, such as α-galactosylceramide (αGalCer), presented by CD1d. iNKT cells exhibit anti-tumor activity via their NK-like cytotoxicity and adjuvant activity. Although iNKT cell-targeted immunotherapy is a conceptually promising approach, we still found a technical hurdle for its clinical implementation which is mainly due to the low frequency of iNKT cells, particularly in humans. To compensate for this, we proposed to generate adequate numbers of clinically competent NKT cells from induced pluripotent stem cells (iPSCs) for cancer immunotherapy. Toward this goal, we first obtained the proof of concept (POC) for this approach in mice. We developed a technology to differentiate iPSCs into iNKT cells (iPSC-iNKT cells) and found iPSC-iNKT cells efficiently rejected a syngeneic experimental thymoma by inducing antigen-specific CD8 T cells. After achieving the POC in mice, we developed human iPSC-iNKT cells, which had a high correlation in their gene expression profiles with parental iNKT cells. Human iPSC-iNKT cells also exhibited anti-tumor activity and adjuvant activity for human NK cells in vivo. Based on this supporting evidence for the anti-tumor activity of human iPSC-iNKT cells, we began to generate good manufacturing practice (GMP)-grade iPSC-iNKT cells. As of now, the first-in-human clinical trial of iPSC-iNKT cell therapy is ongoing as a single-agent, dose-escalation study for patients with advanced head and neck cancer. Demonstration of the safety of iPSC-iNKT cell therapy may allow us to improve the strategy by further reinforcing the therapeutic activity of iPSC-iNKT, cells either by gene-editing or combinatorial use with other immune cell products such as dendritic cells. Sixteen years after the establishment of the iPSC technology, we are reaching the first checkpoint to evaluate the clinical efficacy of iPSC-derived immune cells.
  • KEITA OGAWA, TETSUHIRO CHIBA, MASATO NAKAMURA, JUN ARAI, JIAQI ZHANG, YAOJIA MA, NA QIANG, JUNJIE AO, SAE YUMITA, TAKAMASA ISHINO, MOTOYASU KAN, TERUNAO IWANAGA, MIYUKI NAKAGAWA, KISAKO FUJIWARA, TAKAFUMI SAKUMA, HIROAKI KANZAKI, KEISUKE KOROKI, YUKO KUSAKABE, KAZUFUMI KOBAYASHI, NAOYA KANOGAWA, SOICHIRO KIYONO, TAKAYUKI KONDO, RYO NAKAGAWA, SADAHISA OGASAWARA, RYOSUKE MUROYAMA, SHINGO NAKAMOTO, TATSUO KANDA, HITOSHI MARUYAMA, JUN KATO, SHOJI MATSUMOTO, TAKAYOSHI ARAI, SHINICHIRO MOTOHASHI, NAOYA KATO
    Anticancer Research 43(3) 1043-1052 2023年2月28日  査読有り
  • Taisuke Kaiho, Hidemi Suzuki, Atsushi Hata, Hiroki Matsumoto, Kazuhisa Tanaka, Yuichi Sakairi, Shinichiro Motohashi, Ichiro Yoshino
    Frontiers in pharmacology 14 1298085-1298085 2023年  
    Immune checkpoint molecules such as programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) have revolutionized the field of lung cancer treatment. As part of our study, we examined the role of these proteins in acute rejection in a mouse model of heterotopic tracheal transplantation. Recipient mice were untreated (Allo group) or treated with anti-PD-L1 (aPDL1 group) or PD-L1 Fc recombinant protein (PD-L1 Fc group). A further group of C57BL/6 mice received isografts (Iso group). The occlusion rate was significantly higher in the Allo group than in the Iso group (p = 0.0075), and also higher in the aPD-L1 group (p = 0.0066) and lower in the PD-L1 Fc group (p = 0.030) than in the Allo group. PD-L1 Fc recombinant protein treatment significantly decreased interleukin-6 and interferon-γ levels and reduced the CD4+/CD8+ T cell ratio, without increasing PD-1 and T-cell immunoglobulin mucin 3 expression in CD4+ T cells. These data suggest that PD-L1 Fc recombinant protein decreases the levels of inflammatory cytokines and the proportion of CD4+ T cells without exhaustion. The PD-L1-mediated immune checkpoint mechanism was associated with rejection in the murine tracheal transplant model, suggesting a potential novel target for immunotherapy in lung transplantation.
  • Masataka Ikeda, Tomomi Ide, Shouji Matsushima, Soichiro Ikeda, Kosuke Okabe, Akihito Ishikita, Tomonori Tadokoro, Masashi Sada, Ko Abe, Midori Sato, Akiko Hanada, Shinobu Arai, Kisho Ohtani, Atsushi Nonami, Shinichi Mizuno, Sachio Morimoto, Shinichiro Motohashi, Koichi Akashi, Masaru Taniguchi, Hiroyuki Tsutsui
    Circulation. Heart failure 15(12) e009366 2022年12月  査読有り
    BACKGROUND: Dilated cardiomyopathy (DCM) is a life-threatening disease, resulting in refractory heart failure. An immune disorder underlies the pathophysiology associated with heart failure progression. Invariant natural killer T (iNKT) cell activation is a prospective therapeutic strategy for ischemic heart disease. However, its efficacy in nonischemic cardiomyopathy, such as DCM, remains to be elucidated, and the feasible modality for iNKT cell activation in humans is yet to be validated. METHODS: Dendritic cells isolated from human volunteers were pulsed with α-galactosylceramide ex vivo, which were used as α-galactosylceramide-pulsed dendritic cells (αGCDCs). We treated DCM mice harboring mutated troponin TΔK210/ΔK210 with αGCDCs and evaluated the efficacy of iNKT cell activation on heart failure in DCM mice. Furthermore, we investigated the molecular basis underlying its therapeutic effects in these mice and analyzed primary cardiac cells under iNKT cell-secreted cytokines. RESULTS: The number of iNKT cells in the spleens of DCM mice was reduced compared with that in wild-type mice, whereas αGCDC treatment activated iNKT cells, prolonged survival of DCM mice, and prevented decline in the left ventricular ejection fraction for 4 weeks, accompanied by suppressed interstitial fibrosis. Mechanistically, αGCDC treatment suppressed TGF (transforming growth factor)-β signaling and expression of fibrotic genes and restored vasculature that was impaired in DCM hearts by upregulating angiopoietin 1 (Angpt1) expression. Consistently, IFNγ (interferon gamma) suppressed TGF-β-induced Smad2/3 signaling and the expression of fibrotic genes in cardiac fibroblasts and upregulated Angpt1 expression in cardiomyocytes via Stat1. CONCLUSIONS: Immunomodulatory cell therapy with αGCDCs is a novel therapeutic strategy for heart failure in DCM.
  • 松本 寛樹, 鈴木 秀海, 山中 崇寛, 西井 開, 太枝 帆高, 伊藤 祐輝, 海寳 大輔, 畑 敦, 伊藤 貴正, 田中 教久, 坂入 祐一, 本橋 新一郎, 吉野 一郎
    移植 57(総会臨時) 340-340 2022年10月  
  • Yuki Shiina, Hidemi Suzuki, Atsushi Hata, Taisuke Kaiho, Hiroki Matsumoto, Takahide Toyoda, Yuichi Sakairi, Hironobu Wada, Shinichiro Motohashi, Ichiro Yoshino
    General Thoracic and Cardiovascular Surgery 2022年6月29日  査読有り
    OBJECTIVE: Antibody-mediated rejection (AMR) could induce acute or chronic graft failure during organ transplantation. Several reports have shown that anti-C5 antibodies are effective against AMR after kidney transplantation. However, few reports have assessed the efficacy of anti-C5 antibodies against AMR after lung transplantation. Therefore, this study aimed to evaluate the efficacy of this novel therapy against AMR after lung transplantation. METHODS: BALB/c and C57BL/6 mice were used as donors and recipients. One group was pre-sensitized (PS) by skin transplantation 14 days before lung transplantation. The other group was non-sensitized (NS). Orthotopic left-lung transplantation was performed in both groups. Animals were killed at 2 or 7 days after lung transplantation and evaluated for histopathology, C4d immunostaining, and serum donor-specific antibodies (DSAs) (n = 5 per group). Isograft (IS) models with C57BL/6 mice were used as controls. To evaluate the efficacy of C5 inhibition, other animals, which received similar treatments to those in the PS group, were treated with anti-C5 antibodies, cyclosporine/methylprednisolone, anti-C5 antibodies/cyclosporine/methylprednisolone, or isotype-matched irrelevant control monoclonal antibodies (n = 5 per group). RESULTS: Two days after lung transplantation, the NS group exhibited mild, localized graft-rejection features (rejection score: 0.45 ± 0.08, p = 0.107). The PS group exhibited AMR features with a significantly higher rejection score (2.29 ± 0.42, p = 0.001), C4d vascular-endothelium deposition, and substantial presence of serum DSA. On day 7 after lung transplantation, both groups showed extensive graft alveolar wall destruction, and high acute-rejection scores. Mice receiving anti-C5 antibodies or anti-C5/antibodies/cyclosporine/methylprednisolone demonstrated significantly lower acute-rejection scores (0.63 ± 0.23, p = 0.002; 0.59 ± 0.22, p = 0.001, respectively) than those receiving isotype control antibodies. CONCLUSIONS: Murine orthotopic allograft lung transplant models met the clinical diagnosis and pathogenesis classification criteria of AMR. In these models, anti-C5 antibodies suppressed AMR. Therefore, anti-C5 therapy may be effective against AMR after lung transplantation.
  • 松本 寛樹, 鈴木 秀海, 山中 崇寛, 西井 開, 伊藤 祐輝, 海寳 大輔, 畑 敦, 田中 教久, 坂入 祐一, 芳野 充, 本橋 新一郎, 吉野 一郎
    日本呼吸器外科学会雑誌 36(Suppl.) O76-2 2022年5月  
  • 鈴木 秀海, 畑 敦, 山中 崇寛, 松本 寛樹, 海寳 大輔, 椎名 裕樹, 豊田 行英, 伊藤 貴正, 稲毛 輝長, 田中 教久, 坂入 祐一, 和田 啓伸, 岩田 剛和, 本橋 新一郎, 吉田 成利, 吉野 一郎
    胸部外科 75(4) 244-251 2022年4月  
  • Hidemi Suzuki, Atsushi Hata, Takahiro Yamanaka, Hiroki Matsumoto, Taisuke Kaiho, Yuki Shiina, Takahide Toyoda, Takamasa Ito, Terunaga Inage, Kazuhisa Tanaka, Yuichi Sakairi, Hironobu Wada, Takekazu Iwata, Shinichiro Motohashi, Shigetoshi Yoshida, Ichiro Yoshino
    Kyobu geka. The Japanese journal of thoracic surgery 75(4) 244-251 2022年4月  
    Lung transplantation has become popular in Japan, showing better survival rate than other countries. However, the results are still not satisfactory compared with other solid organ transplantation. One of the reasons for this might be that knowledge on donor-specific antibodies or antibody-related rejection, which has been attracting attention these days, is less than that of kidney or liver transplantation. Our laboratory has continued basic research in this field using rodent lung transplantation model. We have previously shown that type V collagen is associated in chronic rejection as an autoimmune, and that oral administration of type V collagen induces tolerance. The murine chronic rejection model of the minor antigen mismatch was developed, and involvement of the humoral immunity and role of the complement activation were shown. We are now studying the effects of immune checkpoint molecules, which play a central role in the field of cancer therapy, on rejection after lung transplantation. We are also working to verify the effects of anti-complement drugs and molecular targeted drugs in the future treatment on rejection.
  • Keita Ogawa, Hiroaki Kanzaki, Tetsuhiro Chiba, Junjie Ao, Na Qiang, Yaojia Ma, Jiaqi Zhang, Sae Yumita, Takamasa Ishino, Hidemi Unozawa, Motoyasu Kan, Terunao Iwanaga, Miyuki Nakagawa, Kisako Fujiwara, Naoto Fujita, Takafumi Sakuma, Keisuke Koroki, Yuko Kusakabe, Kazufumi Kobayashi, Naoya Kanogawa, Soichiro Kiyono, Masato Nakamura, Takayuki Kondo, Tomoko Saito, Ryo Nakagawa, Sadahisa Ogasawara, Eiichiro Suzuki, Shingo Nakamoto, Ryosuke Muroyama, Tatsuo Kanda, Hitoshi Maruyama, Naoya Mimura, Jun Kato, Shinichiro Motohashi, Naoya Kato
    Journal of Cancer 13(8) 2656-2661 2022年  査読有り
    Atezolizumab plus bevacizumab (ATZ/BV) treatment is a combined immunotherapy consisting of immune checkpoint inhibitor (ICI) and anti-vascular endothelial growth factor monoclonal antibody, which has brought a major paradigm shift in the treatment of unresectable hepatocellular carcinoma (HCC). Gain-of-function mutation of CTNNB1 contributes to resistance of ICI monotherapy through the framework of non-T-cell-inflamed tumor microenvironment. However, whether CTNNB1 mutation renders resistance to ATZ/BV similar to ICI monotherapy remains to be elucidated. In this study, a liquid biopsy sample in plasma of 33 patients with HCC treated with ATZ/BV was subjected to droplet digital PCR for detecting hotspot mutations at the exon 3 of CTNNB1 locus. A total of eight patients (24.2%) exhibited at least one CTNNB1 mutation. The objective response rate (ORR) in patients with wild-type (WT) and mutant (MT) CTNNB1 was 8.0% and 12.5%, respectively, and the disease control rate (DCR) was 68.0% and 87.5%, respectively. No significant difference in both ORR and DCR has been observed between the two groups. The median progression-free survival in patients with WT and MT CTNNB1 was 6.6 and 7.6 months, respectively (not statistically significant). Similarly, no significant difference in overall survival has been observed between patients with WT and MT CTNNB1 (13.6 vs. 12.3 months). In conclusion, the treatment effect of ATZ/BV in patients with HCC with MT CTNNB1 was comparable to those patients with WT CTNNB1. These results implicate that BV added to ATZ might improve immunosuppressive tumor microenvironment caused by CTNNB1 mutation.
  • 西井 開, 本橋 新一郎, 苅田 涼, 平井 有紀, 太枝 帆高, 伊藤 祐輝, 小野里 優希, 松本 寛樹, 海寳 大輔, 大橋 康太, 豊田 行英, 稲毛 輝長, 田中 教久, 坂入 祐一, 鈴木 秀海, 中島 崇裕, 吉野 一郎
    千葉医学雑誌 97(6) 122-122 2021年12月  
  • 海寳 大輔, 鈴木 秀海, 松本 寛樹, 豊田 行英, 稲毛 輝長, 田中 教久, 坂入 祐一, 中島 崇裕, 吉野 一郎, 本橋 新一郎, 木内 政宏, 中山 俊憲
    千葉医学雑誌 97(6) 122-122 2021年12月  
  • 萩谷 政明, 田中 健, 會田 啓介, 白神 梨沙, 伊原 史英, 石井 絢菜, 高見 真理子, 本間 敏明, 和田 祐一, 本橋 新一郎
    調査研究ジャーナル 10(1) 27-33 2021年10月  査読有り
    NKT細胞特異的リガンドであるαガラクトシルセラミドを提示させた抗原提示細胞(以下APC)の投与によって、体内で活性化されたNKT細胞は強力な抗腫瘍効果を発揮する。進行・再発非小細胞肺癌の標準治療後に対してAPC投与を行い、長期生存が得られる可能性を臨床研究で示してきた。今回、未治療の進行非小細胞肺癌患者2症例に対する初回治療として、APCの静脈内投与を2回行い、その後プラチナ製剤を含む抗癌剤治療を2コース行った。その結果、APC投与に関連する重篤な有害事象を認めなかった。APC投与後の抗癌剤投与により、末梢血NKT細胞数は2例ともに減少したが、NKT細胞により活性化されるNK細胞数やCD8+T細胞、αガラクトシルセラミド特異的インターフェロン-γ産生細胞数は1例で増加し、もう1例では治療前値レベルを維持した。NKT細胞により誘導される抗腫瘍免疫応答は、細胞傷害性抗癌剤治療後も持続する可能性が示唆された。(著者抄録)
  • Toshinori Nakayama, Kiyoshi Hirahara, Motoko Y Kimura, Chiaki Iwamura, Masahiro Kiuchi, Kota Kokubo, Atsushi Onodera, Kahoko Hashimoto, Shinichiro Motohashi
    International Immunology 2021年8月24日  査読有り
    CD4 + T cells direct immune responses against infectious microorganisms but are also involved in the pathogenesis of inflammatory diseases. In the last two to three decades, various researchers have identified and characterized several functional CD4 + T cell subsets, including T-helper 1 (Th1), Th2, Th9 and Th17 cells and regulatory T (Treg) cells. In this mini-review, we introduce the concept of pathogenic Th cells that induce inflammatory diseases with a model of disease induction by a population of pathogenic Th cells; "pathogenic Th population disease-induction model". We will focus on Th2 cells that induce allergic airway inflammation-pathogenic Th2 cells (Tpath2 cells)-and discuss the nature of Tpath2 cells that shape the pathology of chronic inflammatory diseases. Various Tpath2 cell subsets have been identified and their unique features are summarized in mouse and human systems. Second, we will discuss how Th cells migrate and are maintained in chronic inflammatory lesions. We propose a model known as the "CD69-Myl9 system". CD69 is a cell surface molecule expressed on activated T cells and interaction with its ligand myosin light chain 9 (Myl9) is required for the induction of inflammatory diseases. Myl9 molecules in the small vessels of inflamed lungs may play a crucial role in the migration of activated T cells into inflammatory lesions. Emerging evidence may provide new insight into the pathogenesis of chronic inflammatory diseases and contribute to the development of new therapeutic strategies for intractable inflammatory disorders.
  • Reiri Shimazaki, Shigetsugu Takano, Mamoru Satoh, Mamoru Takada, Yoji Miyahara, Kosuke Sasaki, Hideyuki Yoshitomi, Shingo Kagawa, Katsunori Furukawa, Tsukasa Takayashiki, Satoshi Kuboki, Kazuyuki Sogawa, Shinichiro Motohashi, Fumio Nomura, Masaru Miyazaki, Masayuki Ohtsuka
    Cellular Oncology (Dordrecht) 44(4) 937-950 2021年8月  査読有り
    BACKGROUND: The interplay between cancer cells and stromal components, including soluble mediators released from cancer cells, contributes to the progression of pancreatic ductal adenocarcinoma (PDAC). Here, we set out to identify key secreted proteins involved in PDAC progression. METHODS: We performed secretome analyses of culture media of mouse pancreatic intraepithelial neoplasia (PanIN) and PDAC cells using Stable Isotope Labeling by Amino acid in Cell culture (SILAC) with click chemistry and liquid chromatography-mass spectrometry (LC-MS/MS). The results obtained were verified in primary PDAC tissue samples and cell line models. RESULTS: Complement factor B (CFB) was identified as one of the robustly upregulated proteins, and found to exhibit elevated expression in PDAC cells compared to PanIN cells. Endogenous CFB knockdown by a specific siRNA dramatically decreased the proliferation of PDAC cells, PANC-1 and MIA PaCa-II. CFB knockdown induced increases in the number of senescence-associated-β-galactosidase (SA-β-gal) positive cells exhibiting p21 expression upregulation, which promotes cellular senescence with cyclinD1 accumulation. Furthermore, CFB knockdown facilitated downregulation of proliferating cell nuclear antigen and led to cell cycle arrest in the G1 phase in PDAC cells. Using immunohistochemistry, we found that high stromal CFB expression was associated with unfavorable clinical outcomes with hematogenous dissemination after surgery in human PDAC patients. Despite the presence of enriched CD8+ tumor infiltrating lymphocytes in the PDAC tumor microenvironments, patients with a high stromal CFB expression exhibited a significantly poorer prognosis compared to those with a low stromal CFB expression. Immunofluorescence staining revealed a correlation between stromal CFB expression in the tumor microenvironment and an enrichment of immunosuppressive regulatory T-cells (Tregs), myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). We also found that high stromal CFB expression showed a positive correlation with high CD8+/Foxp3+ Tregs populations in PDAC tissues. CONCLUSIONS: Our data indicate that CFB, a key secreted protein, promotes proliferation by preventing cellular senescence and is associated with immunological tumor promotion in PDAC. These findings suggest that CFB may be a potential target for the treatment of PDAC.
  • Ayaka Hara, Ryo Koyama-Nasu, Mariko Takami, Takahide Toyoda, Takahiro Aoki, Fumie Ihara, Masayoshi Kobayashi, Seiichiro Hirono, Tomoo Matsutani, Toshinori Nakayama, Yasuo Iwadate, Shinichiro Motohashi
    Cancer Immunology, Immunotherapy : CII 2020年10月31日  査読有り
    Glioblastoma is the most common and aggressive type of brain tumor with high recurrence and fatality rates. Although various therapeutic strategies have been explored, there is currently no effective treatment for glioblastoma. Recently, the number of immunotherapeutic strategies has been tested for malignant brain tumors. Invariant natural killer T (iNKT) cells play an important role in anti-tumor immunity. To address if iNKT cells can target glioblastoma to exert anti-tumor activity, we assessed the expression of CD1d, an antigen-presenting molecule for iNKT cells, on glioblastoma cells. Glioblastoma cells from 10 of 15 patients expressed CD1d, and CD1d-positive glioblastoma cells pulsed with glycolipid ligand induced iNKT cell-mediated cytotoxicity in vitro. Although CD1d expression was low on glioblastoma stem-like cells, retinoic acid, which is the most common differentiating agent, upregulated CD1d expression in these cells and induced iNKT cell-mediated cytotoxicity. Moreover, intracranial administration of human iNKT cells induced tumor regression of CD1d-positive glioblastoma in orthotopic xenografts in NOD/Shi-scid IL-2RγKO (NOG) mice. Thus, CD1d expression represents a novel target for NKT cell-based immunotherapy for glioblastoma patients.
  • Akane S. Suzuki, Ryoji Yagi, Motoko Y. Kimura, Chiaki Iwamura, Kenta Shinoda, Atsushi Onodera, Kiyoshi Hirahara, Damon J. Tumes, Ryo Koyama-Nasu, Siiri E. Iismaa, Robert M. Graham, Shinichiro Motohashi, Toshinori Nakayama
    Frontiers in Immunology 11 2020年7月21日  査読有り
  • Atsushi Hata, Hidemi Suzuki, Takahiro Nakajima, Taiki Fujiwara, Yuki Shiina, Taisuke Kaiho, Takahide Toyoda, Terunaga Inage, Takamasa Ito, Yuichi Sakairi, Hajime Tamura, Hironobu Wada, Yoshito Yamada, Masako Chiyo, Keisuke Matsusaka, Masaki Fukuyo, Ken-ichi Shinohara, Sakae Itoga, Shinichiro Motohashi, Kazuyuki Matsushita, Atsushi Kaneda, Ichiro Yoshino
    PLOS ONE 15(5) e0232884-e0232884 2020年5月8日  査読有り
  • Fumihiro Ishibashi, Yuichi Sakairi, Takekazu Iwata, Yasumitsu Moriya, Teruaki Mizobuchi, Hidehisa Hoshino, Shigetoshi Yoshida, Hideki Hanaoka, Ichiro Yoshino, Shinichiro Motohashi
    Clinical Immunology (Orlando, Fla.) 215 108457-108457 2020年5月6日  査読有り
    We conducted a phase I study of the trans-bronchial injection of α-galactosylceramide (αGalCer)-pulsed antigen presenting cells (APCs) to evaluate their safety, immune responses, and anti-tumor activities. Patients with advanced or recurrent non-small cell lung cancer (NSCLC) refractory to standard treatments were eligible. αGalCer-pulsed APCs were administered intratumorally or intranodally by bronchoscopy. Twenty-one patients were enrolled in this study. No severe adverse events related to the cell therapy were observed during this study in any patient. After αGalCer-pulsed APCs were administrated, increased iNKT cell numbers were observed in PBMCs from eight cases, and IFN-γ producing cells were increased in the peripheral blood of 10 cases. Regarding clinical responses, one case exhibited a partial response and eight were classified as stable disease. In the tumor microenvironment, IFN-γ expression was upregulated after treatment in partial response or stable disease cases and TGF-β was upregulated in progressive disease cases.
  • Aoki, T, Takami, M, Takatani, T, Motoyoshi, K, Ishii, A, Hara, A, Toyoda, T, Okada, R, Hino, M, Koyama-Nasu, R, Kiuchi, M, Hirahara, K, Kimura, M, Nakayama, T, Shimojo, N, Motohashi, S
    Cancer Science 111(7) 2223-2233 2020年4月23日  査読有り
  • Takahide Toyoda, Toshiko Kamata, Kazuhisa Tanaka, Fumie Ihara, Mariko Takami, Hidemi Suzuki, Takahiro Nakajima, Takayuki Ikeuchi, Yohei Kawasaki, Hideki Hanaoka, Toshinori Nakayama, Ichiro Yoshino, Shinichiro Motohashi
    Journal for Immunotherapy of Cancer 8(1) 2020年3月  査読有り
    BACKGROUND: Invariant natural killer T (iNKT) cells produce copious amounts of cytokines in response to specific glycolipid antigens such as α-galactosylceramide (αGalCer) presented by CD1d-expressing antigen-presenting cells (APCs), thus orchestrating other immune cells to fight tumors. Because of their ability to induce strong antitumor responses activated by αGalCer, iNKT cells have been studied for their application in cancer immunotherapy. In our previous phase I/II trial in non-small cell lung cancer (NSCLC) patients who had completed the standard treatment, we showed a relatively long median survival time without severe treatment-related adverse events. Based on these results, we performed a phase II trial to evaluate clinical responses, safety profiles and immune responses as a second-line treatment for advanced NSCLC. METHODS: Patients with advanced or recurrent NSCLC refractory to first-line chemotherapy were eligible. αGalCer-pulsed APCs were intravenously administered four times. Overall survival time was evaluated as the primary endpoint. The safety profile and immune responses after APC injection were also monitored. This study was an open label, single-arm, phase II clinical trial performed at Chiba University Hospital, Japan. RESULTS: Thirty-five patients were enrolled in this study, of which 32 (91.4%) completed the trial. No severe adverse events related to the treatment were observed. The estimated median survival time of the 35 cases was 21.9 months (95% CI, 14.8 to 26.0). One case (2.9%) showed a partial response, 14 cases (40.0%) remained as stable disease, and 19 cases (54.3%) were evaluated as progressive disease. The geometric mean number of iNKT cells in all cases was significantly decreased and the mean numbers of natural killer (NK) cells, interferon-γ-producing cells in response to αGalCer, and effector CD8+ T cells were significantly increased after the administration of αGalCer-pulsed APCs. CONCLUSIONS: The intravenous administration of αGalCer-pulsed APCs was well-tolerated and was accompanied by prolonged overall survival. These results are encouraging and warrant further evaluation in a randomized phase III trial to demonstrate the survival benefit of this immunotherapy. TRIAL REGISTRATION NUMBER: UMIN000007321.
  • Toshiko Kamata, Shigetoshi Yoshida, Mariko Takami, Fumie Ihara, Hiroko Yoshizawa, Takahide Toyoda, Yuichiro Takeshita, Seiichi Nobuyama, Yukiko Kanetsuna, Tetsuo Sato, Ichiro Yoshino, Shinichiro Motohashi
    Cancer science 111(1) 288-296 2020年1月  査読有り
    The role of immune checkpoint inhibitors in metastatic lung cancer has been established in recent years and the pretherapeutic profiles of the tumor microenvironment in responders have been increasingly reported. The role of salvage surgery and the immune profiles of the posttherapeutic specimens in patients achieving an objective response have rarely been studied. We report a case of metastatic lung cancer treated by anti-programmed death-1 Ab followed by surgical resection. The immune status of the tumor was assessed, showing germinal center formation, memory B cell infiltration, and a high frequency of interferon gamma -secreting T cells.
  • Fumie Ihara, Daiju Sakurai, Mariko Takami, Toshiko Kamata, Naoki Kunii, Kazuki Yamasaki, Tomohisa Iinuma, Toshinori Nakayama, Shinichiro Motohashi, Yoshitaka Okamoto
    Cancer immunology, immunotherapy : CII 68(12) 1935-1947 2019年12月  査読有り
    BACKGROUND: Due to the strong tumoricidal activities of activated natural killer T (NKT) cells, invariant NKT cell-based immunotherapy has shown promising clinical efficacy. However, suppressive factors, such as regulatory T cells (Tregs), may be obstacles in the use of NKT cell-based cancer immunotherapy for advanced cancer patients. Here, we investigated the suppressive effects of Tregs on NKT cells and the underlying mechanisms with the aim to improve the antitumor activities of NKT cells. METHODS: Peripheral blood samples were obtained from healthy donors, patients with benign tumors, and patients with head and neck squamous cell carcinoma (HNSCC). NKT cells, induced with α-galactosylceramide (α-GalCer), and monocyte-derived dendritic cells (DCs) were co-cultured with naïve CD4+ T cell-derived Tregs to investigate the mechanism of the Treg suppressive effect on NKT cell cytotoxic function. The functions and phenotypes of NKT cells were evaluated with flow cytometry and cytometric bead array. RESULTS: Treg suppression on NKT cell function required cell-to-cell contact and was mediated via impaired DC maturation. NKT cells cultured under Treg-enriched conditions showed a decrease in CD4- NKT cell frequency, which exert strong tumoricidal responsiveness upon α-GalCer stimulation. The same results were observed in HNSCC patients with significantly increased effector Tregs. CONCLUSION: Tregs exert suppressive effects on NKT cell tumoricidal function by inducing more CD4- NKT cell anergy and less CD4+ NKT cell anergy. Both Treg depletion and NKT cell recovery from the anergy state may be important for improving the clinical efficacy of NKT cell-based immunotherapy in patients with advanced cancers.
  • Takahiro Aoki, Mariko Takami, Tomozumi Takatani, Kiwamu Motoyoshi, Ayana Ishii, Ayaka Hara, Takahide Toyoda, Reona Okada, Moeko Hino, Ryo Koyama-Nasu, Masahiro Kiuchi, Kiyoshi Hirahara, Toshinori Nakayama, Naoki Shimojo, Shinichiro Motohashi
    Blood 134(Supplement_1) 3225-3225 2019年11月13日  
    Background: Invariant natural killer T (iNKT) cells are known as CD1d-restricted T cells that express the invariant T-cell receptors (TCR) Vα24 and Vβ11 in humans and specifically recognize glycolipid antigens such as α-galactosylceramide (αGalCer) presented by CD1d. iNKT cells show direct cytotoxicity toward CD1d-positive tumor cells presenting glycolipid antigens and indirect cytotoxicity by activating other cytotoxic immune cells or regulating CD1d-positive immunosuppressive cells in the tumor microenvironment. Although we previously reported that αGalCer-activated NKT cells exert a potent perforin-dependent cytotoxic activity against a wide variety of human tumor cell lines, the direct recognition of CD1d-negative tumors is controversial and the mechanism is unknown. Here we clarify whether iNKT cells recognize and exhibit cytotoxicity toward leukemia cells in a CD1d-independent manner and identify the molecule that recognizes CD1d-negative leukemia cells. Methods: Purified iNKT cells were generated from peripheral blood mononuclear cells (PBMCs) of healthy adult volunteer donors. PBMCs were cultured in complete RPMI 1640 medium for 9-14 days in the presence of 100 U/mL of recombinant human IL-2 and 200 ng/mL of αGalCer. The iNKT cells were then isolated with an autoMACS Pro separator using FITC-labeled anti-Vα24 antibody (clone, C15) and anti-FITC microbeads. We evaluated the cytotoxic activity of iNKT cells toward CD1d-negative leukemia cells within four days after isolation using a CD107a assay for degranulation, cytometric bead array for cytokine production, and cytotoxicity assay in vitro and in vivo. For in vivo cytotoxicity assays, NOG mice were inoculated with 1 × 106 K562-luc cells on day 0 and with 4 × 106 human iNKT cells on day 1. Gene knock-out (KO) was performed using a CRISPR/Cas9 system. T-cell or NK receptor-KO iNKT cells were used for experiments three or four days after electroporation of the Cas9 protein and guide RNA CRISPR ribonucleoprotein complex. Patient-derived leukemia cells were obtained from PBMCs or bone marrow mononuclear cells of pre-treatment pediatric patients. All studies were approved by the institutional review board and the Animal Care and Use Committee of Chiba University. Results: We observed that iNKT cells degranulated and released Th1 cytokines when co-cultured with CD1d-negative leukemia cells (K562, HL-60, REH, and CD1d-KO U937) as well as αGalCer-loaded CD1d-positive leukemia cells (Jurkat), and showed in vitro cytotoxicity toward these CD1d-negative leukemia cells. This CD1d-independent degranulation decreased over time after isolation and was not restored with re-stimulation by αGalCer. The cytotoxicity of iNKT cells toward K562 cells was confirmed in vivo by comparsion with survival curves of K562-inoculated NOG mice given iNKT cells or PBS alone (log-rank, p= 0.016). To identify the receptors contributing to the CD1d-independent recognition and cytotoxicity against CD1d-negative leukemia cells, we first focused on costimulatory receptors, which are also known as activating NK receptors and are expressed on iNKT cells such as NKG2D, DNAM-1, 2B4, LFA-1, and CD2, and analyzed cytotoxicity after blocking these receptors with antibodies. We found that all costimulatory receptors that we assessed contributed to cytotoxicity toward CD1d-negative leukemia cells. Next, we analyzed cytotoxicity of TCR-KO iNKT cells toward CD1d-negative leukemia cells to confirm the contribution of TCR to CD1d-independent recognition. Notably, TCR-KO iNKT cells showed decreased degranulation, Th1 cytokine release, and cytotoxicity toward K562 cells more so than iNKT cells with KO of NK receptors such as LFA-1(CD11a) or CD2. To assess the clinical application potential of adoptive iNKT cell immunotherapy for leukemia treatment, we analyzed degranulation of iNKT cells using patient-derived leukemia cells. We found iNKT cells degranulation using cells from four out of five myeloid leukemia cases, but only one out of eight BCP-ALL cases (p = 0.032). Conclusion: Primary iNKT cells activated by αGalCer can recognize and show anti-tumor effects toward leukemia cells in an unrestricted manner via CD1d. The TCR also has an important role in recognizing CD1d-negative leukemia cells and multiple NK receptors assist in cytotoxicity. Adoptive iNKT cell immunotherapy may be effective in treating myeloid leukemia. Disclosures No relevant conflicts of interest to declare.
  • Kazuaki Harada, Fumie Ihara, Mariko Takami, Toshiko Kamata, Naoko Mise, Hiroko Yoshizawa, Tomoro Hishiki, Takeshi Saito, Keita Terui, Mitsuyuki Nakata, Shugo Komatsu, Takayuki Ikeuchi, Toshinori Nakayama, Hideo Yoshida, Shinichiro Motohashi
    Cancer science 110(3) 888-902 2019年3月  査読有り
    Dendritic cells (DC) play a key role in the initiation of both antitumor immunity and immunological tolerance. It has been demonstrated that exposure to soluble factors produced by tumor cells modulates DC functions and induces tolerogenic DC differentiation. In this study, we investigated the effects of neuroblastoma cell line-derived soluble factors on DC differentiation. Monocytes isolated from healthy volunteers were incubated with interleukin (IL)-4 and granulocyte-macrophage colony-stimulating factor in the presence of culture supernatants from neuroblastoma cell lines. The culture supernatants from neuroblastoma cell lines, such as NLF and GOTO, partially blocked both downregulation of CD14 and upregulation of CD1a, and dramatically decreased IL-12 and tumor necrosis factor (TNF)-α production from mature DC, while no effect of SH-SY5Y cell supernatant was noted. In addition, IL-6 and IL-10 production from monocytes was increased by the supernatants of NLF and GOTO cells at 24 hours after incubation. Furthermore, we evaluated DC functions through stimulation of invariant natural killer T (iNKT) cells. α-Galactosylceramide-pulsed DC co-cultured with supernatants of NLF cells were unable to sufficiently stimulate iNKT cells. The decreased ability of iNKT cells to produce interferon (IFN)-γ after stimulation with neuroblastoma cell line supernatant-cultured DC was reversed by addition of IL-12. CD40 expression and IL-12 production in NLF-sup-treated DC were increased by addition of exogenous IFN-γ. These results indicate that tolerogenic DC are induced in the neuroblastoma tumor microenvironment and attenuate the antitumor effects of iNKT cells. Interactions between iNKT cells and αGalCer-pulsed DC have the potential to restore the immunosuppression of tolerogenic DC through IFN-γ production.
  • Makita Y, Kunii N, Sakurai D, Ihara F, Motohashi S, Suzuki A, Nakayama T, Okamoto Y
    BMC cancer 18(1) 1254 2018年12月  査読有り
  • Takami M, Cunha C, Motohashi S, Nakayama T, Iwashima M
    European journal of immunology 48(12) 1938-1943 2018年12月  査読有り
  • Tanaka K, Kanesaka Y, Takami M, Suzuki A, Hosokawa H, Onodera A, Kamata T, Nagato K, Nakayama T, Yoshino I, Motohashi S
    Biochemical and biophysical research communications 506(1) 27-32 2018年11月  査読有り
  • Takami M, Iwashima M, Motohashi S
    調査研究ジャーナル 7(2) 80-87 2018年10月  査読有り
  • Kimura MY, Igi A, Hayashizaki K, Mita Y, Shinzawa M, Kadakia T, Endo Y, Ogawa S, Yagi R, Motohashi S, Singer A, Nakayama T
    Nature communications 9(1) 3749-3749 2018年9月14日  査読有り
  • Takami M, Ihara F, Motohashi S
    Frontiers in immunology 9 2021 2018年9月  査読有り
  • Mita Y, Kimura MY, Hayashizaki K, Koyama-Nasu R, Ito T, Motohashi S, Okamoto Y, Nakayama T
    International immunology 30(12) 559-567 2018年8月  査読有り
  • Sakurai D, Uchida R, Ihara F, Kunii N, Nakagawa T, Chazono H, Hanazawa T, Motohashi S, Okamoto Y
    BMC research notes 11(1) 479 2018年7月  査読有り
  • Tomoro Hishiki, Naoko Mise, Kazuaki Harada, Fumie Ihara, Mariko Takami, Takeshi Saito, Keita Terui, Mitsuyuki Nakata, Shugo Komatsu, Hideo Yoshida, Shinichiro Motohashi
    Pediatric surgery international 34(2) 169-176 2018年2月  査読有り
    BACKGROUND: Invariant natural killer T (iNKT) cells play an important role in tumor immunity, enhancing both innate and acquired immunity. We have previously shown the enhancement of antibody-dependent cellular cytotoxicity against neuroblastoma by activated iNKT cells. As a first step towards clinical application, we studied the frequency and proliferative response of circulating iNKT cells in children with and without cancer. METHODS: Blood samples were collected from 10 patients with pediatric malignant solid tumors and 11 patients with non-neoplastic diseases (control). The frequency of circulating iNKT cells was quantified by flow cytometry. Whole peripheral blood mononuclear cells were then stimulated with α-galactosylceramide (α-GalCer) for 7 days, and the expansion rate of the iNKT-cell fraction was assessed. RESULTS: The frequency of iNKT cells in the patients of the cancer and control group did not differ to a statistically significant extent. The iNKT-cell population increased after α-GalCer stimulation in all cases. The iNKT cells of patients who had undergone intensive chemotherapy also had the potential to expand in vitro. CONCLUSIONS: Unlike adult cancer patients, the numbers of circulating iNKT cells were not decreased in pediatric cancer patients. α-GalCer stimulation induced a proliferative response in all of the patients.
  • Tomoro Hishiki, Naoko Mise, Kazuaki Harada, Fumie Ihara, Mariko Takami, Takeshi Saito, Keita Terui, Mitsuyuki Nakata, Shugo Komatsu, Hideo Yoshida, Shinichiro Motohashi
    Pediatric surgery international 34(2) 195-201 2018年2月  査読有り
    BACKGROUND: Tumor immunity has been suggested to play a key role in clinical and biological behavior of neuroblastomas. Given that CD1-restricted invariant natural killer T (iNKT) cells enhance both innate and acquired tumor immunity, we investigated the expression of the iNKT-cell-specific T-cell receptor Vα24-Jα18 in neuroblastoma tissues and its correlation with clinical and biological characteristics. METHODS: Using real- time quantitative PCR, we quantified the expression of Vα24-Jα18 in untreated tumor samples from 107 neuroblastoma cases followed in our institution and analyzed the correlation between the presence of infiltrated iNKT cells and clinical characteristics or patients' outcome. RESULTS: Vα24-Jα18 receptor was detected in 62 untreated cases (57.9%). The expression was significantly higher in stages 1, 2, 3, or 4S (P = 0.0099), in tumors with low or intermediate risk (P = 0.0050), with high TrkA expression (P = 0.0229), with favorable histology (P = 0.0026), with aneuploidy (P = 0.0348), and in younger patients (P = 0.0036). The overall survival rate was significantly higher in patients with iNKT-cell infiltration (log-rank; P = 0.0089). CONCLUSIONS: Since tumor-infiltrating iNKT cells were predominantly observed in neuroblastomas undergoing spontaneous differentiation and/or regression, we suggest that iNKT cells might play a key role in these processes.
  • Kawashima T, Ikari N, Watanabe Y, Kubota Y, Yoshio S, Kanto T, Motohashi S, Shimojo N, Tsuji NM
    Frontiers in immunology 9 27 2018年1月23日  査読有り
  • Fumie Ihara, Daiju Sakurai, Atsushi Horinaka, Yuji Makita, Akira Fujikawa, Toshioki Sakurai, Kazuki Yamasaki, Naoki Kunii, Shinichiro Motohashi, Toshinori Nakayama, Yoshitaka Okamoto
    CANCER IMMUNOLOGY IMMUNOTHERAPY 66(10) 1275-1285 2017年10月  査読有り
    Background Although regulatory T cells (Tregs) are thought to play an important role in immune suppression, their clinical significance in head and neck squamous cell carcinoma (HNSCC) is unclear. A recent study reported Tregs could be divided into functional subsets based on the expression of CD45RA and Foxp3. Method The frequency of circulating Treg subsets was analyzed in patients with HNSCC and compared with the frequency in patients with benign tumors. The association of Treg subsets with the frequency of lymphocyte subsets, status of progression, clinical course, and prognosis were also examined. Results The frequency of CD4(+)Foxp3(+) Tregs was comparable between HNSCC patients and age-matched benign tumor patients; however, CD45RA(-)Foxp3(high) Tregs were significantly increased in HNSCC patients, in particular those with advanced stage tumors. The high frequency of CD45RA(-)Foxp3(high) Tregs correlated with a poor prognosis and the low frequency of CD45RA(-)Foxp3(high) Tregs before treatment showed a better clinical outcome, even in patients with advanced stage tumors. CD45RA(-)Foxp3(high) Treg numbers were decreased after intensive treatments; however, Treg numbers recovered in the early stages of recurrent cases, even before the clinical manifestation. Conclusion CD45RA(-)Foxp3(high) Tregs are associated with the clinical course of HNSCC and might be a new target for treatment and an early marker of tumor recurrence in HNSCC patients.
  • Motoko Y. Kimura, Koji Hayashizaki, Koji Tokoyoda, Shiki Takamura, Shinichiro Motohashi, Toshinori Nakayama
    IMMUNOLOGICAL REVIEWS 278(1) 87-100 2017年7月  査読有り
    CD69 has been known as an early activation marker of lymphocytes; whereas, recent studies demonstrate that CD69 also has critical functions in immune responses. Early studies using human samples revealed the involvement of CD69 in various inflammatory diseases including asthma. Moreover, murine disease models using Cd69(-/-) mice and/or anti-CD69 antibody (Ab) treatment have revealed crucial roles for CD69 in inflammatory responses. However, it had not been clear how the CD69 molecule contributes to the pathogenesis of inflammatory diseases. We recently elucidated a novel mechanism, in which the interaction between CD69 and its ligands, myosin light chain 9, 12a and 12b (Myl9/12) play a critical role in the recruitment of activated T cells into the inflammatory lung. In this review, we first summarize CD69 function based on its structure and then introduce the evidence for the involvement of CD69 in human diseases and murine disease models. Then, we will describe how we discovered CD69 ligands, Myl9 and Myl12, and how the CD69-Myl9 system regulates airway inflammation. Finally, we will discuss possible therapeutic usages of the blocking Ab to the CD69-Myl9 system.
  • Toshiko Kamata, Akane Suzuki, Naoko Mise, Fumie Ihara, Mariko Takami, Yuji Makita, Atsushi Horinaka, Kazuaki Harada, Naoki Kunii, Shigetoshi Yoshida, Ichiro Yoshino, Toshinori Nakayama, Shinichiro Motohashi
    CANCER IMMUNOLOGY IMMUNOTHERAPY 65(12) 1477-1489 2016年12月  査読有り
    The role of invariant natural killer T (iNKT) cells in antitumor immunity has been studied extensively, and clinical trials in patients with advanced cancer have revealed a prolonged survival in some cases. In recent years, humanized blocking antibodies against co-stimulatory molecules such as PD-1 have been developed. The enhancement of T cell function is reported to improve antitumor immunity, leading to positive clinical effects. However, there are limited data on the role of PD-1/programmed death ligand (PDL) molecules in human iNKT cells. In this study, we investigated the interaction between PD-1 on iNKT cells and PDL on antigen-presenting cells (APCs) in the context of iNKT cell stimulation. The blockade of PDL1 at the time of stimulation resulted in increased release of helper T cell (Th) 1 cytokines from iNKT cells, leading to the activation of NK cells. The direct antitumor function of iNKT cells was also enhanced after stimulation with anti-PDL1 antibody-treated APCs. According to these results, we conclude that the co-administration of anti-PDL1 antibody and alpha-galactosylceramide (alpha GalCer)-pulsed APCs enhances iNKT cell-mediated antitumor immunity.
  • Koji Hayashizaki, Motoko Y. Kimura, Koji Tokoyoda, Hiroyuki Hosokawa, Kenta Shinoda, Kiyoshi Hirahara, Tomomi Ichikawa, Atsushi Onodera, Asami Hanazawa, Chiaki Iwamura, Jungo Kakuta, Kenzo Muramoto, Shinichiro Motohashi, Damon J. Tumes, Tomohisa Iinuma, Heizaburo Yamamoto, Yuzuru Ikehara, Yoshitaka Okamoto, Toshinori Nakayama
    Science Immunology 1(3) eaaf9154-eaaf9154 2016年9月  査読有り
  • Takahiro Aoki, Moeko Hino, Katsuyoshi Koh, Masashi Kyushiki, Hiroshi Kishimoto, Yuki Arakawa, Ryoji Hanada, Hiroshi Kawashima, Jun Kurihara, Naoki Shimojo, Shinichiro Motohashi
    PEDIATRIC BLOOD & CANCER 63(8) 1461-1464 2016年8月  査読有り
    Programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway blockade has become a promising therapeutic target in adult cancers. We evaluated PD-L1 expression and tumor-infiltrating CD8(+) T cells in formalin-fixed, paraffin-embedded tumor specimens from 53 untreated pediatric patients with eight cancer types: neuroblastoma, extracranial malignant germ cell tumor, hepatoblastoma, germinoma, medulloblastoma, renal tumor, rhabdomyosarcoma, and atypical teratoid/rhabdoid tumor. One rhabdomyosarcoma with the shortest survival exhibited membranous PD-L1 expression and germinoma contained abundant tumor-infiltrating CD8+ T cells and PD-L1-positive macrophages. The PD-1/PD-L1 pathway tended to be inactive in pediatric cancers. (C) 2016 The Authors. Pediatric Blood & Cancer, published by Wiley Periodicals, Inc.
  • Hosokawa H, Tanaka T, Endo Y, Kato M, Shinoda K, Suzuki A, Motohashi S, Matsumoto M, Nakayama KI, Nakayama T
    Nature communications 7 11289 2016年4月  査読有り
  • 田中 教久, 本橋 新一郎, 高見 真理子, 和田 啓伸, 藤原 大樹, 鈴木 秀海, 中島 崇裕, 岩田 剛和, 吉田 成利, 吉野 一郎
    日本呼吸器外科学会雑誌 30(3) P75-5 2016年4月  
  • Atsushi Horinaka, Daiju Sakurai, Fumie Ihara, Yuji Makita, Naoki Kunii, Shinichiro Motohashi, Toshinori Nakayama, Yoshitaka Okamoto
    CANCER SCIENCE 107(3) 207-216 2016年3月  査読有り
    Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature and progenitor myeloid cells with an immunosuppressive role in various types of cancer, including head and neck squamous cell carcinoma (HNSCC). However, the effect on the host immune system, especially on invariant NKT (iNKT) cells with potent anti-tumor activity, remains unclear. In this study, we investigated the effects of circulating MDSC subsets on the peripheral lymphocytes of patients with head and neck tumors. A significant accumulation of CD15(+) granulocytic MDSC (G-MDSC) and CD14(+) monocytic MDSC (M-MDSC) was demonstrated in HNSCC patients. The percentage of G-MDSC showed an inverse correlation with the percentage of T cells in the peripheral blood. The increased G-MDSC was significantly associated with advanced clinical stage and poor prognosis of HNSCC patients. The proliferation and viability of T cells were suppressed by CD15(+) cells, and the suppression was reversed by adding the hydrogen peroxide scavenger catalase. However, iNKT cell activation upon -galactosylceramide (GalCer) stimulation was not affected by the presence or absence of CD15(+) G-MDSC. These results indicate that increased G-MDSC negatively affects peripheral T cell immunity, but not iNKT cells, in HNSCC patients, and that T cells are more sensitive to hydrogen peroxide produced by G-MDSC than iNKT cells. Cancer immunotherapy designed to enhance the antitumor activity of iNKT cells by stimulation with GalCer may remain effective in the presence of G-MDSC.

MISC

 199

書籍等出版物

 13

講演・口頭発表等

 187

共同研究・競争的資金等の研究課題

 31