籠谷 勇紀 (カゴヤ ユウキ)

Kagoya, Yuki

写真a

所属(所属キャンパス)

医学部 先端医科学研究所(がん免疫) (信濃町)

職名

教授

メールアドレス

メールアドレス

HP

研究室住所

東京都新宿区信濃町35

研究室電話番号

03-5843-6174

研究室FAX番号

03-5843-6177

プロフィール 【 表示 / 非表示

  • 2007年東京大学医学部卒業。臨床研修を経て、2009-2013年 東京大学大学院医学系研究科・内科学専攻博士課程、博士(医学)取得。2013-2014年 東京大学医学部附属病院血液・腫瘍内科助教を経て、2014-2018年にPrincess Margaret Cancer Centre (カナダ・トロント)において、リサーチ・フェローとしてがん免疫療法の基礎研究に従事。2018-2019年 東京大学医学部附属病院無菌治療部講師。2019年10月より愛知県がんセンター研究所 腫瘍免疫応答研究分野・分野長として研究室を主宰。2020年4月より名古屋大学大学院医学系研究科がん先端診断・治療開発学講座 細胞腫瘍学分野連携教授 (兼任)。2023年1月より現職。

経歴 【 表示 / 非表示

  • 2007年04月
    -
    2009年03月

    関東労災病院, 内科

  • 2013年04月
    -
    2013年09月

    東京大学, 医学部附属病院 血液・腫瘍内科, 特任研究員

  • 2013年10月
    -
    2014年05月

    東京大学, 医学部附属病院 血液・腫瘍内科, 助教

  • 2014年06月
    -
    2018年05月

    プリンセス・マーガレットがんセンター, Tumor Immunotherapy Program, Research Fellow

  • 2018年06月
    -
    2019年09月

    東京大学, 医学部附属病院, 講師

全件表示 >>

学歴 【 表示 / 非表示

  • 2001年04月
    -
    2007年03月

    東京大学, 医学部, 医学科

  • 2009年04月
    -
    2013年03月

    東京大学, 大学院医学系研究科

学位 【 表示 / 非表示

  • 博士(医学), 東京大学, 課程, 2013年03月

 

研究分野 【 表示 / 非表示

  • ライフサイエンス / 免疫学 (腫瘍免疫、がん免疫療法、細胞療法、分子細胞生物学、血液・腫瘍内科学)

 

論文 【 表示 / 非表示

  • CD83 expression characterizes precursor exhausted T cell population

    Zhiwen Wu, Toshiaki Yoshikawa, Satoshi Inoue, Yusuke Ito, Hitomi Kasuya, Takahiro Nakashima, Haosong Zhang, Saki Kotaka, Waki Hosoda, Shiro Suzuki, Yuki Kagoya

    Communications Biology 6 ( 1 ) 258 2023年03月

    研究論文(学術雑誌), 最終著者, 責任著者, 査読有り

     概要を見る

    T cell exhaustion is a main obstacle against effective cancer immunotherapy. Exhausted T cells include a subpopulation that maintains proliferative capacity, referred to as precursor exhausted T cells (TPEX). While functionally distinct and important for antitumor immunity, TPEX possess some overlapping phenotypic features with the other T-cell subsets within the heterogeneous tumor-infiltrating T-lymphocytes (TIL). Here we explore surface marker profiles unique to TPEX using the tumor models treated by chimeric antigen receptor (CAR)-engineered T cells. We find that CD83 is predominantly expressed in the CCR7+PD1+ intratumoral CAR-T cells compared with the CCR7-PD1+ (terminally differentiated) and CAR-negative (bystander) T cells. The CD83+CCR7+ CAR-T cells exhibit superior antigen-induced proliferation and IL-2 production compared with the CD83- T cells. Moreover, we confirm selective expression of CD83 in the CCR7+PD1+ T-cell population in primary TIL samples. Our findings identify CD83 as a marker to discriminate TPEX from terminally exhausted and bystander TIL.

  • Genetic ablation of PRDM1 in antitumor T cells enhances therapeutic efficacy of adoptive immunotherapy

    Toshiaki Yoshikawa, Zhiwen Wu, Satoshi Inoue, Hitomi Kasuya, Hirokazu Matsushita, Yusuke Takahashi, Hiroaki Kuroda, Waki Hosoda, Shiro Suzuki, Yuki Kagoya

    Blood (American Society of Hematology)  139 ( 14 ) 2156 - 2172 2022年04月

    最終著者, 責任著者, 査読有り,  ISSN  0006-4971

     概要を見る

    Adoptive cancer immunotherapy can induce objective clinical efficacy in patients with advanced cancer; however, a sustained response is achieved in a minority of cases. The persistence of infused T cells is an essential determinant of a durable therapeutic response. Antitumor T cells undergo a genome-wide remodeling of the epigenetic architecture upon repeated antigen encounters, which inevitably induces progressive T-cell differentiation and the loss of longevity. In this study, we identified PR domain zinc finger protein 1 (PRDM1) ie, Blimp-1, as a key epigenetic gene associated with terminal T-cell differentiation. The genetic knockout of PRDM1 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) supported the maintenance of an early memory phenotype and polyfunctional cytokine secretion in repeatedly stimulated chimeric antigen receptor (CAR)-engineered T cells. PRDM1 disruption promoted the expansion of less differentiated memory CAR-T cells in vivo, which enhanced T-cell persistence and improved therapeutic efficacy in multiple tumor models. Mechanistically, PRDM1-ablated T cells displayed enhanced chromatin accessibility of the genes that regulate memory formation, thereby leading to the acquisition of gene expression profiles representative of early memory T cells. PRDM1 knockout also facilitated maintaining an early memory phenotype and cytokine polyfunctionality in T-cell receptor-engineered T cells as well as tumor-infiltrating lymphocytes. In other words, targeting PRDM1 enabled the generation of superior antitumor T cells, which is potentially applicable to a wide range of adoptive cancer immunotherapies.

  • CAMK2G is identified as a novel therapeutic target for myelofibrosis

    Masashi Miyauchi, Ken Sasaki, Yuki Kagoya, Kazuki Taoka, Yosuke Masamoto, Sho Yamazaki, Shunya Arai, Hideaki Mizuno, Mineo Kurokawa

    Blood Advances (American Society of Hematology)  6 ( 5 ) 1585 - 1597 2022年03月

    査読有り,  ISSN  2473-9529

     概要を見る

    Although JAK1/2 inhibition is effective in alleviating symptoms of myelofibrosis (MF), it does not result in the eradication of MF clones, which can lead to inhibitor-resistant clones emerging during the treatment. Here, we established induced pluripotent stem cells (iPSCs) derived from MF patient samples (MF-iPSCs) harboring JAK2 V617F, CALR type 1, or CALR type 2 mutations. We demonstrated that these cells faithfully recapitulate the drug sensitivity of the disease. These cells were used for chemical screening, and calcium/calmodulin-dependent protein kinase 2 (CAMK2) was identified as a promising therapeutic target. MF model cells and mice induced by MPL W515L, another type of mutation recurrently detected in MF patients, were used to elucidate the therapeutic potential of CAMK2 inhibition. CAMK2 inhibition was effective against JAK2 inhibitor-sensitive and JAK2 inhibitor-resistant cells. Further research revealed CAMK2 γ subtype was important in MF model cells induced by MPL W515L. We showed that CAMK2G hetero knockout in the primary bone marrow cells expressing MPL W515L decreased colony-forming capacity. CAMK2G inhibition with berbamine, a CAMK2G inhibitor, significantly prolonged survival and reduced disease phenotypes, such as splenomegaly and leukocytosis in a MF mouse model induced by MPL W515L. We investigated the molecular mechanisms underlying the therapeutic effect of CAMK2G inhibition and found that CAMK2G is activated by MPL signaling in MF model cells and is an effector in the MPL-JAK2 signaling pathway in these cells. These results indicate CAMK2G plays an important role in MF, and CAMK2G inhibition may be a novel therapeutic strategy that overcomes resistance to JAK1/2 inhibition.

  • CD62L expression level determines the cell fate of myeloid progenitors

    Yusuke Ito, Fumio Nakahara, Yuki Kagoya, Mineo Kurokawa

    Stem Cell Reports (Elsevier BV)  16 ( 12 ) 2871 - 2886 2021年12月

    査読有り,  ISSN  2213-6711

     概要を見る

    Hematopoietic cells differentiate through several progenitors in a hierarchical manner, and recent single-cell analyses have revealed substantial heterogeneity within each progenitor. Although common myeloid progenitors (CMPs) are defined as a multipotent cell population that can differentiate into granulocyte-monocyte progenitors (GMPs) and megakaryocyte-erythrocyte progenitors (MEPs), and GMPs generate neutrophils and monocytes, these myeloid progenitors must contain some lineage-committed progenitors. Through gene expression analysis at single-cell levels, we identified CD62L as a marker to reveal the heterogeneity. We confirmed that CD62L-negative CMPs represent "bona fide" CMPs, whereas CD62L-high CMPs are mostly restricted to GMP potentials both in mice and humans. In addition, we identified CD62L-negative GMPs as the most immature subsets in GMPs and Ly6C+CD62L-intermediate and Ly6C+CD62L-high GMPs are skewed to neutrophil and monocyte differentiation in mice, respectively. Our findings contribute to more profound understanding about the mechanism of myeloid differentiation.

  • Evi1 upregulates Fbp1 and supports progression of acute myeloid leukemia through pentose phosphate pathway activation

    Hideaki Mizuno, Junji Koya, Yosuke Masamoto, Yuki Kagoya, Mineo Kurokawa

    Cancer Science (Wiley)  112 ( 10 ) 4112 - 4126 2021年10月

    査読有り,  ISSN  1347-9032

     概要を見る

    Evi1 is a transcription factor essential for the development as well as progression of acute myeloid leukemia (AML) and high Evi1 AML is associated with extremely poor clinical outcome. Since targeting metabolic vulnerability is the emerging therapeutic strategy of cancer, we herein investigated a novel therapeutic target of Evi1 by analyzing transcriptomic, epigenetic, and metabolomic profiling of mouse high Evi1 leukemia cells. We revealed that Evi1 overexpression and Evi1-driven leukemic transformation upregulate transcription of gluconeogenesis enzyme Fbp1 and other pentose phosphate enzymes with interaction between Evi1 and the enhancer region of these genes. Metabolome analysis using Evi1-overexpressing leukemia cells uncovered pentose phosphate pathway upregulation by Evi1 overexpression. Suppression of Fbp1 as well as pentose phosphate pathway enzymes by shRNA-mediated knockdown selectively decreased Evi1-driven leukemogenesis in vitro. Moreover, pharmacological or shRNA-mediated Fbp1 inhibition in secondarily transplanted Evi1-overexpressing leukemia mouse significantly decreased leukemia cell burden. Collectively, targeting FBP1 is a promising therapeutic strategy of high Evi1 AML.

全件表示 >>

総説・解説等 【 表示 / 非表示

  • Epigenetic engineering for optimal chimeric antigen receptor T cell therapy

    Yusuke Ito, Yuki Kagoya

    Cancer Science 113 ( 11 ) 3664 - 3671 2022年11月

    責任著者

     概要を見る

    Recent advancements in cancer immunotherapy, such as chimeric antigen receptor (CAR)-engineered T cell therapy and immune checkpoint therapy, have significantly improved the clinical outcomes of patients with several types of cancer. To broaden its applicability further and induce durable therapeutic efficacy, it is imperative to understand how antitumor T cells elicit cytotoxic functions, survive as memory T cells, or are impaired in their effector functions (exhausted) at the molecular level. T cell properties are regulated by their gene expression profiles, which are further controlled by epigenetic architectures, such as DNA methylation and histone modifications. Multiple studies have elucidated specific epigenetic genes associated with T-cell phenotypic changes. Conversely, exogenous modification of these key epigenetic factors can significantly alter T cell functions by extensively altering the transcription network, which can be applied in cancer immunotherapy by improving T cell persistence or augmenting effector functions. As CAR-T cell therapy involves a genetic engineering step during the preparation of the infusion products, it would be a feasible strategy to additionally modulate specific epigenetic genes in CAR-T cells to improve their quality. Here, we review recent studies investigating how individual epigenetic factors play a crucial role in T-cell biology. We further discuss future directions to integrate these findings for optimal cancer immunotherapy.

  • Dissecting the heterogeneity of exhausted T cells at the molecular level.

    Yuki Kagoya

    International Immunolology 34 ( 11 ) 547 - 553 2022年10月

    筆頭著者, 最終著者, 責任著者

     概要を見る

    Our understanding of mechanisms underlying T-cell exhaustion has been refined by analysis of exhausted T cells at the molecular level. The development and functions of exhausted T cells are regulated by a number of transcription factors, epigenetic factors and metabolic enzymes. In addition, recent work to dissect exhausted T cells at the single-cell level has enabled us to discover a precursor exhausted T-cell subset equipped with long-term survival capacity. Starting from the analysis of mouse models, the existence of precursor exhausted T cells has also been documented in human T cells in the context of chronic virus infections or tumors. Clinical data suggest that evaluating the quality of exhausted T cells on the basis of their differentiation status may be helpful to predict the therapeutic response to inhibition of programmed death 1 (PD1). Moreover, beyond immune-checkpoint blockade, novel therapeutic approaches to re-invigorate exhausted T cells have been explored based on molecular insights into T-cell exhaustion. Here I will discuss key molecular profiles associated with the development, maintenance and differentiation of exhausted T cells and how these findings can be applicable in the field of cancer immunotherapy.

  • NF-κBシグナルによる白血病幹細胞制御

    籠谷 勇紀

    血液内科 (科学評論社)  70 ( 3 ) 373 - 378 2015年03月

    ISSN  2185-582X

研究発表 【 表示 / 非表示

  • 次世代型CAR-T細胞療法の開発

    籠谷 勇紀

    第6回バイオ医薬EXPO, 

    2022年07月

    口頭発表(招待・特別)

  • CAR-T 細胞療法の適用拡大に向けた研究開発

    籠谷 勇紀

    第7回日本がんサポーティブケア学会学術集会, 

    2022年06月

    口頭発表(招待・特別)

  • エピジェネティクス改変による長期生存型CAR-T細胞の製造

    籠谷 勇紀

    第14回日本血液疾患免疫療法学会, 

    2022年06月

    口頭発表(招待・特別)

  • エピジェネティクス修飾によるCAR-T細胞の改良

    籠谷 勇紀

    第3回東京理科大学総合研究院合成生物学研究部門シンポジウム, 

    2022年03月

    口頭発表(招待・特別)

  • Genetic modification of antitumor T cells for optimal adoptive cancer immunotherapy

    Franco-Japanese immuno-oncology webinar series, 

    2022年03月

    口頭発表(招待・特別)

全件表示 >>

競争的研究費の研究課題 【 表示 / 非表示

  • 遺伝子改変による長期生存型CAR-NK細胞療法の開発

    2023年04月
    -
    2026年03月

    籠谷 勇紀, 基盤研究(B), 補助金,  研究代表者

 

担当授業科目 【 表示 / 非表示

  • 先端医科学演習

    2023年度

  • 先端医科学実習

    2023年度

  • 先端医科学特論

    2023年度

  • 先端医科学

    2023年度