4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Targeting of Nrf2 improves antitumoral responses by human NK cells, TIL and CAR T cells during oxidative stress

      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

          Background

          Adoptive cell therapy using cytotoxic lymphocytes is an efficient immunotherapy against solid and hematological cancers. However, elevated levels of reactive oxygen species (ROS) in the hostile tumor microenvironment can impair NK cell and T cell function. Auranofin, a gold (I)-containing phosphine compound, is a strong activator of the transcription factor Nrf2. Nrf2 controls a wide range of downstream targets important for the cells to obtain increased resistance to ROS. In this study, we present a strategy using auranofin to render human cytotoxic lymphocytes resistant toward oxidative stress.

          Methods

          Melanoma patient-derived tumor infiltrating lymphocytes (TIL) and healthy donor-derived NK cells and CD19-directed CAR T cells were pretreated with a low dose of auranofin. Their resistance toward oxidative stress was assessed by measuring antitumoral responses (killing-assay, degranulation/CD107a, cytokine production) and intracellular ROS levels (flow cytometry) in conditions of oxidative stress. To confirm that the effects were Nrf2 dependent, the transcription level of Nrf2-driven target genes was analyzed by qPCR.

          Results

          Pretreatment of human TIL and NK cells ex vivo with a low-dose auranofin significantly lowered their accumulation of intracellular ROS and preserved their antitumoral activity despite high H 2O 2 levels or monocyte-derived ROS. Furthermore, auranofin pretreatment of CD19 CAR-T cells or TIL increased their elimination of CD19 +tumor cells or autologous tumor spheroids, respectively, especially during ROS exposure. Analysis of Nrf2-driven target genes revealed that the increased resistance against ROS was Nrf2 dependent.

          Conclusion

          These novel findings suggest that Nrf2 activation in human cytotoxic lymphocytes could be used to enhance the efficacy of adoptive cell therapy.

          Related collections

          Most cited references52

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

          Reactive oxygen species (ROS) as pleiotropic physiological signalling agents

          'Reactive oxygen species' (ROS) is an umbrella term for an array of derivatives of molecular oxygen that occur as a normal attribute of aerobic life. Elevated formation of the different ROS leads to molecular damage, denoted as 'oxidative distress'. Here we focus on ROS at physiological levels and their central role in redox signalling via different post-translational modifications, denoted as 'oxidative eustress'. Two species, hydrogen peroxide (H2O2) and the superoxide anion radical (O2·-), are key redox signalling agents generated under the control of growth factors and cytokines by more than 40 enzymes, prominently including NADPH oxidases and the mitochondrial electron transport chain. At the low physiological levels in the nanomolar range, H2O2 is the major agent signalling through specific protein targets, which engage in metabolic regulation and stress responses to support cellular adaptation to a changing environment and stress. In addition, several other reactive species are involved in redox signalling, for instance nitric oxide, hydrogen sulfide and oxidized lipids. Recent methodological advances permit the assessment of molecular interactions of specific ROS molecules with specific targets in redox signalling pathways. Accordingly, major advances have occurred in understanding the role of these oxidants in physiology and disease, including the nervous, cardiovascular and immune systems, skeletal muscle and metabolic regulation as well as ageing and cancer. In the past, unspecific elimination of ROS by use of low molecular mass antioxidant compounds was not successful in counteracting disease initiation and progression in clinical trials. However, controlling specific ROS-mediated signalling pathways by selective targeting offers a perspective for a future of more refined redox medicine. This includes enzymatic defence systems such as those controlled by the stress-response transcription factors NRF2 and nuclear factor-κB, the role of trace elements such as selenium, the use of redox drugs and the modulation of environmental factors collectively known as the exposome (for example, nutrition, lifestyle and irradiation).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements

            CRISPR-Cas9 is poised to become the gene editing tool of choice in clinical contexts. Thus far, exploration of Cas9-induced genetic alterations has been limited to the immediate vicinity of the target site and distal off-target sequences, leading to the conclusion that CRISPR-Cas9 was reasonably specific. Here we report significant on-target mutagenesis, such as large deletions and more complex genomic rearrangements at the targeted sites in mouse embryonic stem cells, mouse hematopoietic progenitors and a human differentiated cell line. Using long-read sequencing and long-range PCR genotyping, we show that DNA breaks introduced by single-guide RNA/Cas9 frequently resolved into deletions extending over many kilobases. Furthermore, lesions distal to the cut site and crossover events were identified. The observed genomic damage in mitotically active cells caused by CRISPR-Cas9 editing may have pathogenic consequences.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found

              Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases

              The transcription factor NF-E2 p45-related factor 2 (NRF2; encoded by NFE2L2) and its principal negative regulator, the E3 ligase adaptor Kelch-like ECH-associated protein 1 (KEAP1), are critical in the maintenance of redox, metabolic and protein homeostasis, as well as the regulation of inflammation. Thus, NRF2 activation provides cytoprotection against numerous pathologies including chronic diseases of the lung and liver; autoimmune, neurodegenerative and metabolic disorders; and cancer initiation. One NRF2 activator has received clinical approval and several electrophilic modifiers of the cysteine-based sensor KEAP1 and inhibitors of its interaction with NRF2 are now in clinical development. However, challenges regarding target specificity, pharmacodynamic properties, efficacy and safety remain.
                Bookmark

                Author and article information

                Journal
                J Immunother Cancer
                J Immunother Cancer
                jitc
                jitc
                Journal for Immunotherapy of Cancer
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                2051-1426
                2022
                22 June 2022
                : 10
                : 6
                : e004458
                Affiliations
                [1 ]departmentDepartment of Oncology-Pathology , Karolinska Institutet , Stockholm, Sweden
                [2 ]departmentDepartment of Clinical Immunology and Transfusion Medicine , Karolinska University Hospital , Stockholm, Sweden
                [3 ]departmentGloria and Seymour Epstein Chair in Cell Therapy and Transplantation , Princess Margaret Hospital Cancer Centre , Toronto, Ontario, Canada
                [4 ]departmentTheme Cancer, Patient area Head and Neck, Lung and Skin , Karolinska University Hospital , Stockholm, Sweden
                [5 ]departmentDepartment of Medical Biochemistry and Biophysics , Karolinska Institutet , Stockholm, Sweden
                [6 ]departmentDepartment of Selenoprotein Research and National Tumor Biology Laboratory , National Institute of Oncology , Budapest, Hungary
                [7 ]departmentClinical Neuroscience , Karolinska Institutet , Stockholm, Sweden
                Author notes
                [Correspondence to ] Dr Stina Linnea Wickström; stina.wickstrom@ 123456ki.se
                Author information
                http://orcid.org/0000-0002-2743-5886
                http://orcid.org/0000-0003-0440-6924
                http://orcid.org/0000-0002-9709-2970
                http://orcid.org/0000-0002-4807-6114
                http://orcid.org/0000-0003-0663-5763
                http://orcid.org/0000-0003-0349-4918
                Article
                jitc-2021-004458
                10.1136/jitc-2021-004458
                9226989
                35738800
                14c8e17a-6804-4012-a661-fdc52a286b6b
                © Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.

                This is an open access article distributed in accordance with the Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits others to copy, redistribute, remix, transform and build upon this work for any purpose, provided the original work is properly cited, a link to the licence is given, and indication of whether changes were made. See https://creativecommons.org/licenses/by/4.0/.

                History
                : 09 May 2022
                Funding
                Funded by: Swedish Medical Research Council;
                Award ID: 2019-01212
                Award ID: LS 2018-1157
                Funded by: http://dx.doi.org/10.13039/501100004063, Knut and Alice Wallenberg Foundation;
                Award ID: KAW2015.0063
                Funded by: FundRef http://dx.doi.org/10.13039/501100002794, Cancerfonden;
                Award ID: 190002FE
                Award ID: 190104Pj01H
                Award ID: 190108Us01H
                Award ID: CAN 2018/451
                Funded by: FundRef http://dx.doi.org/10.13039/501100004047, Karolinska Institutet;
                Award ID: 2018 (2-5586/2017)
                Award ID: 2020-01354
                Funded by: Cancer Society in Stockholm;
                Award ID: 181183
                Award ID: 181201
                Award ID: 194123
                Award ID: 201232
                Funded by: The Hungarian Thematic Excellence Programme (TKP2021-EGA-44);
                Funded by: The Hungarian National Research, Development and Innovation Office (ED_18-1-2019-0025);
                Funded by: The Hungarian National Tumor Biology Laboratory;
                Funded by: The Swedish Research Council/vetenskapsrådet (2021-02214),;
                Funded by: http://dx.doi.org/10.13039/501100004063, Knut and Alice Wallenberg Foundation;
                Award ID: KAW 2019.0059
                Funded by: http://dx.doi.org/10.13039/501100002794, Cancerfonden;
                Award ID: 21 1463 Pj
                Categories
                Clinical/Translational Cancer Immunotherapy
                1506
                2435
                Original research
                Custom metadata
                unlocked

                immunotherapy, adoptive,lymphocytes, tumor-infiltrating,cytotoxicity, immunologic,t-lymphocytes,killer cells, natural

                Comments

                Comment on this article