7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Bempegaldesleukin selectively depletes intratumoral Tregs and potentiates T cell-mediated cancer therapy

      research-article
      1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 2 , 3 , 3 , 1 , 1 , 4 , 5 , 4 , 4 , 6 , 7 , 1 , 2 , 2 , 2 , 2 , 1 , 1 , 2 , 8 , 9 ,
      Nature Communications
      Nature Publishing Group UK
      Cancer therapy, Tumour immunology, Cytokines, Immunotherapy, Lymphocytes

      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

          High dose interleukin-2 (IL-2) is active against metastatic melanoma and renal cell carcinoma, but treatment-associated toxicity and expansion of suppressive regulatory T cells (Tregs) limit its use in patients with cancer. Bempegaldesleukin (NKTR-214) is an engineered IL-2 cytokine prodrug that provides sustained activation of the IL-2 pathway with a bias to the IL-2 receptor CD122 (IL-2Rβ). Here we assess the therapeutic impact and mechanism of action of NKTR-214 in combination with anti-PD-1 and anti-CTLA-4 checkpoint blockade therapy or peptide-based vaccination in mice. NKTR-214 shows superior anti-tumor activity over native IL-2 and systemically expands anti-tumor CD8 + T cells while inducing Treg depletion in tumor tissue but not in the periphery. Similar trends of intratumoral Treg dynamics are observed in a small cohort of patients treated with NKTR-214. Mechanistically, intratumoral Treg depletion is mediated by CD8 + Teff-associated cytokines IFN-γ and TNF-α. These findings demonstrate that NKTR-214 synergizes with T cell-mediated anti-cancer therapies.

          Abstract

          Interleukin-2 can induce an anti-tumour response, but is associated with toxicity. Here, the authors demonstrate that an engineered interleukin-2 promotes intratumoral T regulatory cell depletion while enhancing effective anti-tumour CD8 + T cell responses that result in potent tumor suppression.

          Related collections

          Most cited references37

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

          gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma.

          Stimulating an immune response against cancer with the use of vaccines remains a challenge. We hypothesized that combining a melanoma vaccine with interleukin-2, an immune activating agent, could improve outcomes. In a previous phase 2 study, patients with metastatic melanoma receiving high-dose interleukin-2 plus the gp100:209-217(210M) peptide vaccine had a higher rate of response than the rate that is expected among patients who are treated with interleukin-2 alone. We conducted a randomized, phase 3 trial involving 185 patients at 21 centers. Eligibility criteria included stage IV or locally advanced stage III cutaneous melanoma, expression of HLA*A0201, an absence of brain metastases, and suitability for high-dose interleukin-2 therapy. Patients were randomly assigned to receive interleukin-2 alone (720,000 IU per kilogram of body weight per dose) or gp100:209-217(210M) plus incomplete Freund's adjuvant (Montanide ISA-51) once per cycle, followed by interleukin-2. The primary end point was clinical response. Secondary end points included toxic effects and progression-free survival. The treatment groups were well balanced with respect to baseline characteristics and received a similar amount of interleukin-2 per cycle. The toxic effects were consistent with those expected with interleukin-2 therapy. The vaccine-interleukin-2 group, as compared with the interleukin-2-only group, had a significant improvement in centrally verified overall clinical response (16% vs. 6%, P=0.03), as well as longer progression-free survival (2.2 months; 95% confidence interval [CI], 1.7 to 3.9 vs. 1.6 months; 95% CI, 1.5 to 1.8; P=0.008). The median overall survival was also longer in the vaccine-interleukin-2 group than in the interleukin-2-only group (17.8 months; 95% CI, 11.9 to 25.8 vs. 11.1 months; 95% CI, 8.7 to 16.3; P=0.06). In patients with advanced melanoma, the response rate was higher and progression-free survival longer with vaccine and interleukin-2 than with interleukin-2 alone. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00019682.).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation.

            Adoptive cell transfer therapies (ACTs) with cytotoxic T cells that target melanocytic antigens can achieve remissions in patients with metastatic melanomas, but tumours frequently relapse. Hypotheses explaining the acquired resistance to ACTs include the selection of antigen-deficient tumour cell variants and the induction of T-cell tolerance. However, the lack of appropriate experimental melanoma models has so far impeded clear insights into the underlying mechanisms. Here we establish an effective ACT protocol in a genetically engineered mouse melanoma model that recapitulates tumour regression, remission and relapse as seen in patients. We report the unexpected observation that melanomas acquire ACT resistance through an inflammation-induced reversible loss of melanocytic antigens. In serial transplantation experiments, melanoma cells switch between a differentiated and a dedifferentiated phenotype in response to T-cell-driven inflammatory stimuli. We identified the proinflammatory cytokine tumour necrosis factor (TNF)-α as a crucial factor that directly caused reversible dedifferentiation of mouse and human melanoma cells. Tumour cells exposed to TNF-α were poorly recognized by T cells specific for melanocytic antigens, whereas recognition by T cells specific for non-melanocytic antigens was unaffected or even increased. Our results demonstrate that the phenotypic plasticity of melanoma cells in an inflammatory microenvironment contributes to tumour relapse after initially successful T-cell immunotherapy. On the basis of our work, we propose that future ACT protocols should simultaneously target melanocytic and non-melanocytic antigens to ensure broad recognition of both differentiated and dedifferentiated melanoma cells, and include strategies to sustain T-cell effector functions by blocking immune-inhibitory mechanisms in the tumour microenvironment.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tumor Regression and Autoimmunity after Reversal of a Functionally Tolerant State of Self-reactive CD8+ T Cells

              Many tumor-associated antigens are derived from nonmutated “self” proteins. T cells infiltrating tumor deposits recognize self-antigens presented by tumor cells and can be expanded in vivo with vaccination. These T cells exist in a functionally tolerant state, as they rarely result in tumor eradication. We found that tumor growth and lethality were unchanged in mice even after adoptive transfer of large numbers of T cells specific for an MHC class I–restricted epitope of the self/tumor antigen gp100. We sought to develop new strategies that would reverse the functionally tolerant state of self/tumor antigen-reactive T cells and enable the destruction of large (with products of perpendicular diameters of >50 mm2), subcutaneous, unmanipulated, poorly immunogenic B16 tumors that were established for up to 14 d before the start of treatment. We have defined three elements that are all strictly necessary to induce tumor regression in this model: (a) adoptive transfer of tumor-specific T cells; (b) T cell stimulation through antigen-specific vaccination with an altered peptide ligand, rather than the native self-peptide; and (c) coadministration of a T cell growth and activation factor. Cells, vaccination, or cyto-kine given alone or any two in combination were insufficient to induce tumor destruction. Autoimmune vitiligo was observed in mice cured of their disease. These findings illustrate that adoptive transfer of T cells and IL-2 can augment the function of a cancer vaccine. Furthermore, these data represent the first demonstration of complete cures of large, established, poorly immunogenic, unmanipulated solid tumors using T cells specific for a true self/tumor antigen and form the basis for a new approach to the treatment of patients with cancer.
                Bookmark

                Author and article information

                Contributors
                woverwijk@nektar.com
                Journal
                Nat Commun
                Nat Commun
                Nature Communications
                Nature Publishing Group UK (London )
                2041-1723
                31 January 2020
                31 January 2020
                2020
                : 11
                : 661
                Affiliations
                [1 ]ISNI 0000 0001 2291 4776, GRID grid.240145.6, Department of Melanoma Medical Oncology, , University of Texas MD Anderson Cancer Center, ; Houston, TX USA
                [2 ]ISNI 0000 0004 0410 3955, GRID grid.476522.0, Nektar Therapeutics, ; 455 Mission Bay Blvd South, San Francisco, CA USA
                [3 ]ISNI 0000 0001 2291 4776, GRID grid.240145.6, Department of Infectious Diseases, Infection Control and Employee Health, , The University of Texas MD Anderson Cancer Center, ; Houston, TX 77054 USA
                [4 ]ISNI 0000 0001 2160 926X, GRID grid.39382.33, Department of Molecular and Cellular Biology, , Baylor College of Medicine, ; Houston, TX USA
                [5 ]ISNI 0000 0001 2160 926X, GRID grid.39382.33, Dan L. Duncan Cancer Center, ; Houston, TX USA
                [6 ]GRID grid.433818.5, Yale Comprehensive Cancer Center, ; New Haven, CT USA
                [7 ]ISNI 0000000419368710, GRID grid.47100.32, Yale University Cancer Center, Yale University, ; New Haven, CT USA
                [8 ]ISNI 0000 0001 2291 4776, GRID grid.240145.6, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, ; Houston, TX USA
                [9 ]ISNI 0000 0001 2291 4776, GRID grid.240145.6, Department of Immunology, , University of Texas MD Anderson Cancer Center, ; Houston, TX USA
                Author information
                http://orcid.org/0000-0003-1028-9920
                http://orcid.org/0000-0002-1317-9287
                http://orcid.org/0000-0002-4183-4939
                Article
                14471
                10.1038/s41467-020-14471-1
                6994577
                32005826
                1a54556d-21fc-4f72-8f2a-02c17c1d1d70
                © The Author(s) 2020

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 15 July 2019
                : 10 January 2020
                Categories
                Article
                Custom metadata
                © The Author(s) 2020

                Uncategorized
                cancer therapy,tumour immunology,cytokines,immunotherapy,lymphocytes
                Uncategorized
                cancer therapy, tumour immunology, cytokines, immunotherapy, lymphocytes

                Comments

                Comment on this article