3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Non–clinical efficacy, safety and stable clinical cell processing of induced pluripotent stem cell‐derived anti–glypican‐3 chimeric antigen receptor‐expressing natural killer/innate lymphoid cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The use of allogeneic, pluripotent stem‐cell‐derived immune cells for cancer immunotherapy has been the subject of recent clinical trials. In Japan, investigator‐initiated clinical trials will soon begin for ovarian cancer treatment using human leukocyte antigen (HLA)‐homozygous‐induced pluripotent stem cell (iPSC)‐derived anti–glypican‐3 (GPC3) chimeric antigen receptor (CAR)‐expressing natural killer/innate lymphoid cells (NK/ILC). Using pluripotent stem cells as the source for allogeneic immune cells facilitates stringent quality control of the final product, in terms of efficacy, safety and producibility. In this paper, we describe our methods for the stable, feeder‐free production of CAR‐expressing NK/ILC cells from CAR‐transduced iPSC with clinically relevant scale and materials. The average number of cells that could be differentiated from 1.8‐3.6 × 10 6 iPSC within 7 weeks was 1.8‐4.0 × 10 9. These cells showed stable CD45/CD7/CAR expression, effector functions of cytotoxicity and interferon gamma (IFN‐γ) production against GPC3‐expressing tumor cells. When the CAR‐NK/ILC cells were injected into a GPC3‐positive, ovarian‐tumor‐bearing, immunodeficient mouse model, we observed a significant therapeutic effect that prolonged the survival of the animals. When the cells were injected into immunodeficient mice during non–clinical safety tests, no acute systemic toxicity or tumorigenicity of the final product or residual iPSC was observed. In addition, our test results for the CAR‐NK/ILC cells generated with clinical manufacturing standards are encouraging, and these methods should accelerate the development of allogeneic pluripotent stem cell‐based immune cell cancer therapies.

          Abstract

          This translational study aimed to develop anti–GPC3 CAR‐expressing NK/ILC cells derived from HLA‐homozygous iPSC clone as an effective cell therapy against disseminated ovarian tumors and to assess the clinical cell manufacturing and pre–clinical aspects of the therapy, including safety and efficacy. Those aspects of the therapy clarified in the study provide perspective for the planned clinical trial.

          Related collections

          Most cited references29

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells

          Summary Checkpoint inhibitors have revolutionized cancer treatment. However, only a minority of patients respond to these immunotherapies. Here, we report that blocking the inhibitory NKG2A receptor enhances tumor immunity by promoting both natural killer (NK) and CD8+ T cell effector functions in mice and humans. Monalizumab, a humanized anti-NKG2A antibody, enhanced NK cell activity against various tumor cells and rescued CD8+ T cell function in combination with PD-x axis blockade. Monalizumab also stimulated NK cell activity against antibody-coated target cells. Interim results of a phase II trial of monalizumab plus cetuximab in previously treated squamous cell carcinoma of the head and neck showed a 31% objective response rate. Most common adverse events were fatigue (17%), pyrexia (13%), and headache (10%). NKG2A targeting with monalizumab is thus a novel checkpoint inhibitory mechanism promoting anti-tumor immunity by enhancing the activity of both T and NK cells, which may complement first-generation immunotherapies against cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity

            Chimeric antigen receptors (CARs) significantly enhance anti-tumor activity of immune effector cells. While most studies have evaluated CAR-expression in T cells, here we evaluate different CAR constructs that improve natural killer (NK) cell-mediated killing. We identified a CAR containing the transmembrane domain of NKG2D, the 2B4 co-stimulatory domain, and the CD3ζ signaling domain to mediate strong antigen-specific NK cell signaling. NK cells derived from human iPSCs that express this CAR (NK-CAR-iPSC-NK cells) have a typical NK cell phenotype and demonstrate improved anti-tumor activity compared to T-CAR expressing iPSC-derived NK cells (T-CAR-iPSC-NK cells) and non-CAR expressing cells. Using an ovarian cancer xenograft model, NK-CAR-iPSC-NK cells significantly inhibited tumor growth and prolonged survival compared to PB-NK cells, iPSC-NK cells, or T-CAR-iPSC-NK cells. Additionally, NK-CAR-iPSC-NK cells demonstrate similar in vivo activity as T-CAR-expressing T cells, though with less toxicity. These NK-CAR-iPSC-NK cells now provide standardized, targeted “off the shelf” lymphocytes for anti-cancer immunotherapy. Natural killer (NK) cells are a key part of the immune system’s ability to mediate anti-cancer activity. Kaufman and colleagues utilize human iPSCs to produce NK cells with novel chimeric antigen receptors that specifically target cancer cells in an antigen-specific manner to improve survival in an ovarian cancer xenograft model.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade

              Checkpoint blockade immunotherapy targeting the PD-1/PD-L1 inhibitory axis has produced remarkable results in the treatment of several types of cancer. Whereas cytotoxic T cells are known to provide important antitumor effects during checkpoint blockade, certain cancers with low MHC expression are responsive to therapy, suggesting that other immune cell types may also play a role. Here, we employed several mouse models of cancer to investigate the effect of PD-1/PD-L1 blockade on NK cells, a population of cytotoxic innate lymphocytes that also mediate antitumor immunity. We discovered that PD-1 and PD-L1 blockade elicited a strong NK cell response that was indispensable for the full therapeutic effect of immunotherapy. PD-1 was expressed on NK cells within transplantable, spontaneous, and genetically induced mouse tumor models, and PD-L1 expression in cancer cells resulted in reduced NK cell responses and generation of more aggressive tumors in vivo. PD-1 expression was more abundant on NK cells with an activated and more responsive phenotype and did not mark NK cells with an exhausted phenotype. These results demonstrate the importance of the PD-1/PD-L1 axis in inhibiting NK cell responses in vivo and reveal that NK cells, in addition to T cells, mediate the effect of PD-1/PD-L1 blockade immunotherapy.
                Bookmark

                Author and article information

                Contributors
                kaneko.shin@cira.kyoto-u.ac.jp
                Journal
                Cancer Sci
                Cancer Sci
                10.1111/(ISSN)1349-7006
                CAS
                Cancer Science
                John Wiley and Sons Inc. (Hoboken )
                1347-9032
                1349-7006
                31 March 2020
                May 2020
                : 111
                : 5 ( doiID: 10.1111/cas.v111.5 )
                : 1478-1490
                Affiliations
                [ 1 ] Shin Kaneko Laboratory Department of Cell growth and Differentiation Center for iPS Cell Research and Application (CiRA) Kyoto University Kyoto Japan
                [ 2 ] Thyas Co. Ltd Kyoto Japan
                [ 3 ] Division of Cancer Immunotherapy Exploratory Oncology Research and Clinical Trial Center National Cancer Center Kashiwa Japan
                [ 4 ] Department of Life Science Frontiers Center for iPS Cell Research and Application (CiRA) Kyoto University Kyoto Japan
                [ 5 ] Department of Gastroenterology and Hepatology Kyoto University Graduate School of Medicine Kyoto Japan
                [ 6 ] Department of Immunology Yamaguchi University Graduate School of Medicine Yamaguchi Japan
                Author notes
                [*] [* ] Correspondence

                Shin Kaneko, Shin Kaneko Laboratory, Department of Cell growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara‐cho, Shogoin, Sakyo‐ku, Kyoto 606‐8507, Japan.

                Email: kaneko.shin@ 123456cira.kyoto-u.ac.jp

                Author information
                https://orcid.org/0000-0003-3918-2385
                https://orcid.org/0000-0003-2291-4586
                Article
                CAS14374
                10.1111/cas.14374
                7226201
                32133731
                ef697e8e-1399-4b98-bdd2-c7ffe31eb5e2
                © 2020 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 28 November 2019
                : 07 February 2020
                : 08 February 2020
                Page count
                Figures: 5, Tables: 3, Pages: 13, Words: 7463
                Funding
                Funded by: the Ministry of Education, Culture, Sports, Science and Technology Japan
                Award ID: 15H04655
                Award ID: 25293226
                Award ID: 26293357
                Funded by: Japan Agency for Medical Research and Development , open-funder-registry 10.13039/100009619;
                Funded by: National Cancer Center Research Fund
                Categories
                Original Article
                Original Articles
                Basic and Clinical Immunology
                Custom metadata
                2.0
                May 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.1 mode:remove_FC converted:15.05.2020

                Oncology & Radiotherapy
                chimeric antigen receptor,gpc3,ilc,immunotherapy,ipsc,nk
                Oncology & Radiotherapy
                chimeric antigen receptor, gpc3, ilc, immunotherapy, ipsc, nk

                Comments

                Comment on this article