69
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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.

          Introductory paragraph

          The adoptive transfer of anti-CD19 chimeric antigen receptor (CAR)-engineered T cells has shown impressive clinical responses in patients with refractory B-cell malignancies 17 . However, the therapeutic effects of CAR-T cells targeting other malignancies have not yet resulted in significant clinical benefit 811 . Although inefficient tumor trafficking and various immunosuppressive mechanisms can impede CAR-T cell effector responses, the signals delivered by the current CAR constructs may still be insufficient to fully activate antitumor T cell functions. Optimal T cell activation and proliferation requires multiple signals, including T cell receptor (TCR) engagement (signal 1), costimulation (signal 2), and cytokine engagement (signal 3) 12 . However, CAR gene constructs currently being tested in the clinic contain a CD3z (TCR signaling) domain and a costimulatory domain(s) but not a domain transmitting signal 3 1318 . Here, we have developed a novel CAR construct capable of inducing cytokine signaling upon antigen stimulation. This new generation CD19 CAR encodes a truncated cytoplasmic domain of IL-2Rβ and a STAT3-binding YXXQ motif together with CD3z and CD28 domains (28-ΔIL2RB-z (YXXQ)). The 28-ΔIL2RB-z (YXXQ) CAR-T cells showed antigen-dependent JAK-STAT3/5 pathway activation, which promoted their proliferation and prevented terminal differentiation in vitro. The 28-ΔIL2RB-z (YXXQ) CAR-T cells demonstrated superior in vivo persistence and antitumor effects in both liquid and solid tumor models compared with CAR-T cells with a CD28 or 4-1BB domain alone. Taken together, these results suggest that our new generation CAR has the potential to demonstrate superior antitumor effects with minimal toxicities in the clinic. Clinical translation of this novel CAR is warranted.

          Related collections

          Most cited references17

          • Record: found
          • Abstract: found
          • Article: not found

          Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition.

          Following immune attack, solid tumors upregulate coinhibitory ligands that bind to inhibitory receptors on T cells. This adaptive resistance compromises the efficacy of chimeric antigen receptor (CAR) T cell therapies, which redirect T cells to solid tumors. Here, we investigated whether programmed death-1-mediated (PD-1-mediated) T cell exhaustion affects mesothelin-targeted CAR T cells and explored cell-intrinsic strategies to overcome inhibition of CAR T cells. Using an orthotopic mouse model of pleural mesothelioma, we determined that relatively high doses of both CD28- and 4-1BB-based second-generation CAR T cells achieved tumor eradication. CAR-mediated CD28 and 4-1BB costimulation resulted in similar levels of T cell persistence in animals treated with low T cell doses; however, PD-1 upregulation within the tumor microenvironment inhibited T cell function. At lower doses, 4-1BB CAR T cells retained their cytotoxic and cytokine secretion functions longer than CD28 CAR T cells. The prolonged function of 4-1BB CAR T cells correlated with improved survival. PD-1/PD-1 ligand [PD-L1] pathway interference, through PD-1 antibody checkpoint blockade, cell-intrinsic PD-1 shRNA blockade, or a PD-1 dominant negative receptor, restored the effector function of CD28 CAR T cells. These findings provide mechanistic insights into human CAR T cell exhaustion in solid tumors and suggest that PD-1/PD-L1 blockade may be an effective strategy for improving the potency of CAR T cell therapies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The Principles of Engineering Immune Cells to Treat Cancer

            Chimeric antigen receptor (CAR) T cells have proven that engineered immune cells can serve as a powerful new class of cancer therapeutics. Clinical experience has helped to define the major challenges that must be met to make engineered T cells a reliable, safe, and effective platform that can be deployed against a broad range of tumors. The emergence of synthetic biology approaches for cellular engineering is providing us with a broadly expanded set of tools for programming immune cells. We discuss how these tools could be used to design the next generation of smart T cell precision therapeutics.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer.

              Alterations in EGFR, KRAS, and ALK are oncogenic drivers in lung cancer, but how oncogenic signaling influences immunity in the tumor microenvironment is just beginning to be understood. Immunosuppression likely contributes to lung cancer, because drugs that inhibit immune checkpoints like PD-1 and PD-L1 have clinical benefit. Here, we show that activation of the AKT-mTOR pathway tightly regulates PD-L1 expression in vitro and in vivo. Both oncogenic and IFNγ-mediated induction of PD-L1 was dependent on mTOR. In human lung adenocarcinomas and squamous cell carcinomas, membranous expression of PD-L1 was significantly associated with mTOR activation. These data suggest that oncogenic activation of the AKT-mTOR pathway promotes immune escape by driving expression of PD-L1, which was confirmed in syngeneic and genetically engineered mouse models of lung cancer where an mTOR inhibitor combined with a PD-1 antibody decreased tumor growth, increased tumor-infiltrating T cells, and decreased regulatory T cells.
                Bookmark

                Author and article information

                Journal
                9502015
                8791
                Nat Med
                Nat. Med.
                Nature medicine
                1078-8956
                1546-170X
                22 December 2017
                05 February 2018
                March 2018
                05 August 2018
                : 24
                : 3
                : 352-359
                Affiliations
                [1 ]Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2M9, Canada
                [2 ]Takara Bio, Inc., Kusatsu, Shiga 525-0058, Japan
                [3 ]Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
                [4 ]Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
                [5 ]Department of Hematology/Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 2M9, Canada
                Author notes
                Correspondence and requests for materials should be addressed to N.H. ( naoto.hirano@ 123456utoronto.ca ). Naoto Hirano, MD, PhD, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada, Phone: (416) 946-2190, Fax: (416) 946-6529, naoto.hirano@ 123456utoronto.ca
                Article
                NIHMS930181
                10.1038/nm.4478
                5839992
                29400710
                8460cccb-2b9f-4cbb-9b1f-f98074402778

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Medicine
                Medicine

                Comments

                Comment on this article