HUSCAP logo Hokkaido Univ. logo

Hokkaido University Collection of Scholarly and Academic Papers >
Graduate School of Medicine / Faculty of Medicine >
Peer-reviewed Journal Articles, etc >

Vaccine immunotherapy with ARNAX induces tumor-specific memory T cells and durable anti-tumor immunity in mouse models

This item is licensed under:Creative Commons Attribution-NonCommercial 4.0 International

Files in This Item:
Takeda_et_al-2018-Cancer_Science.pdf866.79 kBPDFView/Open
cas13649-sup-0001-Supinfo.docSupporting Information1.12 MBMicrosoft WordView/Open
Please use this identifier to cite or link to this item:http://hdl.handle.net/2115/71370

Title: Vaccine immunotherapy with ARNAX induces tumor-specific memory T cells and durable anti-tumor immunity in mouse models
Authors: Takeda, Yohei Browse this author
Yoshida, Sumito Browse this author
Takashima, Ken Browse this author
Ishii-Mugikura, Noriko Browse this author
Shime, Hiroaki Browse this author →KAKEN DB
Seya, Tsukasa Browse this author →KAKEN DB
Matsumoto, Misako Browse this author →KAKEN DB
Keywords: cancer immunotherapy
memory CD8+ T cell
PD-L1 blockade
Toll-like receptor 3 adjuvant
tumor microenvironment
Issue Date: Jul-2018
Publisher: John Wiley & Sons
Journal Title: Cancer science
Volume: 109
Issue: 7
Start Page: 2119
End Page: 2129
Publisher DOI: 10.1111/cas.13649
Abstract: Immunological checkpoint blockade therapies benefit a limited population of cancer patients. We have previously shown that vaccine immunotherapy with Toll-like receptor (TLR)3-adjuvant and tumor antigen overcomes anti-programmed death ligand-1 (PD-L1) resistance in mouse tumor models. In the present study, 4 different ovalbumin (OVA)-expressing tumor cell lines were implanted into syngeneic mice and subjected to anti-tumor immunotherapy using ARNAX and whole OVA protein. ARNAX is a TLR3-specific agonist that does not activate the mitochondrial antiviral-signaling protein (MAVS) pathway, and thus does not induce systemic inflammation. Dendritic cell priming and proliferative CTL were induced by ARNAX + OVA, but complete remission was achieved only in a PD-L1-low cell line of EG7. Addition of anti-PD-L1 antibody to the ARNAX + OVA therapy brought complete remission to another PD-L1-high subline of EG7. Tumor shrinkage but not remission was observed in MO5 in that regimen. We analyzed tumor cells and tumor-infiltrating immune cells to identify factors associated with successful ARNAX vaccine therapy. Tumors that responded to ARNAX therapy expressed high levels of MHC class I and low levels of PD-L1. The tumor-infiltrating immune cells in ARNAX-susceptible tumors contained fewer immunosuppressive myeloid cells with low PD-L1 expression. Combination with anti-PD-L1 antibody functioned not only within tumor sites but also within lymphoid tissues, augmenting the therapeutic efficacy of the ARNAX vaccine. Notably, ARNAX therapy induced memory CD8+ T cells and rejection of reimplanted tumors. Thus, ARNAX vaccine + anti-PD-L1 therapy enabled permanent remission against some tumors that stably present antigens.
Rights: http://creativecommons.org/licenses/by-nc/4.0/
Type: article
URI: http://hdl.handle.net/2115/71370
Appears in Collections:医学院・医学研究院 (Graduate School of Medicine / Faculty of Medicine) > 雑誌発表論文等 (Peer-reviewed Journal Articles, etc)

Submitter: 松本 美佐子

Export metadata:

OAI-PMH ( junii2 , jpcoar_1.0 )

MathJax is now OFF:


 

 - Hokkaido University