18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Modulation of intratumoural myeloid cells, the hallmark of the anti-tumour efficacy induced by a triple combination: tumour-associated peptide, TLR-3 ligand and α-PD-1

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          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

          Anti-programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) monoclonal antibodies (mAbs) show remarkable clinical anti-tumour efficacy. However, rational combinations are needed to extend the clinical benefit to primary resistant tumours. The design of such combinations requires the identification of the kinetics of critical immune cell populations in the tumour microenvironment.

          Methods

          In this study, we compared the kinetics of immune cells in the tumour microenvironment upon treatment with immunotherapy combinations with different anti-tumour efficacies in the non-inflamed tumour model TC-1/A9. Tumour-bearing C57BL/6J mice were treated with all possible combinations of a human papillomavirus (HPV) E7 long peptide, polyinosinic–polycytidylic acid (PIC) and anti-PD-1 mAb. Tumour growth and kinetics of the relevant immune cell populations were assessed over time. The involvement of key immune cells was confirmed by depletion with mAbs and immunophenotyping with multiparametric flow cytometry.

          Results

          The maximum anti-tumour efficacy was achieved after intratumoural administration of HPV E7 long peptide and PIC combined with the systemic administration of anti-PD-1 mAb. The intratumoural immune cell kinetics of this combination was characterised by a biphasic immune response. An initial upsurge of proinflammatory myeloid cells led to a further rise in effector CD8 + T lymphocytes at day 8. Depletion of either myeloid cells or CD8 + T lymphocytes diminished the anti-tumour efficacy of the combination.

          Conclusions

          The anti-tumour efficacy of a successful immunotherapy combination in a non-inflamed tumour model relies on an early inflammatory process that remodels the myeloid cell compartment.

          Related collections

          Most cited references48

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

          Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.

          Blockade of programmed death 1 (PD-1), an inhibitory receptor expressed by T cells, can overcome immune resistance. We assessed the antitumor activity and safety of BMS-936558, an antibody that specifically blocks PD-1. We enrolled patients with advanced melanoma, non-small-cell lung cancer, castration-resistant prostate cancer, or renal-cell or colorectal cancer to receive anti-PD-1 antibody at a dose of 0.1 to 10.0 mg per kilogram of body weight every 2 weeks. Response was assessed after each 8-week treatment cycle. Patients received up to 12 cycles until disease progression or a complete response occurred. A total of 296 patients received treatment through February 24, 2012. Grade 3 or 4 drug-related adverse events occurred in 14% of patients; there were three deaths from pulmonary toxicity. No maximum tolerated dose was defined. Adverse events consistent with immune-related causes were observed. Among 236 patients in whom response could be evaluated, objective responses (complete or partial responses) were observed in those with non-small-cell lung cancer, melanoma, or renal-cell cancer. Cumulative response rates (all doses) were 18% among patients with non-small-cell lung cancer (14 of 76 patients), 28% among patients with melanoma (26 of 94 patients), and 27% among patients with renal-cell cancer (9 of 33 patients). Responses were durable; 20 of 31 responses lasted 1 year or more in patients with 1 year or more of follow-up. To assess the role of intratumoral PD-1 ligand (PD-L1) expression in the modulation of the PD-1-PD-L1 pathway, immunohistochemical analysis was performed on pretreatment tumor specimens obtained from 42 patients. Of 17 patients with PD-L1-negative tumors, none had an objective response; 9 of 25 patients (36%) with PD-L1-positive tumors had an objective response (P=0.006). Anti-PD-1 antibody produced objective responses in approximately one in four to one in five patients with non-small-cell lung cancer, melanoma, or renal-cell cancer; the adverse-event profile does not appear to preclude its use. Preliminary data suggest a relationship between PD-L1 expression on tumor cells and objective response. (Funded by Bristol-Myers Squibb and others; ClinicalTrials.gov number, NCT00730639.).
            • Record: found
            • Abstract: found
            • Article: not found

            Oncology meets immunology: the cancer-immunity cycle.

            The genetic and cellular alterations that define cancer provide the immune system with the means to generate T cell responses that recognize and eradicate cancer cells. However, elimination of cancer by T cells is only one step in the Cancer-Immunity Cycle, which manages the delicate balance between the recognition of nonself and the prevention of autoimmunity. Identification of cancer cell T cell inhibitory signals, including PD-L1, has prompted the development of a new class of cancer immunotherapy that specifically hinders immune effector inhibition, reinvigorating and potentially expanding preexisting anticancer immune responses. The presence of suppressive factors in the tumor microenvironment may explain the limited activity observed with previous immune-based therapies and why these therapies may be more effective in combination with agents that target other steps of the cycle. Emerging clinical data suggest that cancer immunotherapy is likely to become a key part of the clinical management of cancer. Copyright © 2013 Elsevier Inc. All rights reserved.
              • Record: found
              • Abstract: found
              • Article: not found

              Elements of cancer immunity and the cancer–immune set point

              Immunotherapy is proving to be an effective therapeutic approach in a variety of cancers. But despite the clinical success of antibodies against the immune regulators CTLA4 and PD-L1/PD-1, only a subset of people exhibit durable responses, suggesting that a broader view of cancer immunity is

                Author and article information

                Contributors
                pberraondol@unav.es
                mgarrido@unav.es
                Journal
                Br J Cancer
                Br J Cancer
                British Journal of Cancer
                Nature Publishing Group UK (London )
                0007-0920
                1532-1827
                3 February 2021
                30 March 2021
                : 124
                : 7
                : 1275-1285
                Affiliations
                [1 ]GRID grid.5924.a, ISNI 0000000419370271, Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, , University of Navarra, ; Pamplona, Spain
                [2 ]Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
                [3 ]GRID grid.5924.a, ISNI 0000000419370271, Program of Immunology and Immunotherapy, , CIMA Universidad de Navarra, ; Pamplona, Spain
                [4 ]GRID grid.417993.1, ISNI 0000 0001 2260 0793, Merck & Co., Inc, ; Kenilworth, NJ USA
                [5 ]GRID grid.413448.e, ISNI 0000 0000 9314 1427, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ; Madrid, Spain
                Author information
                http://orcid.org/0000-0002-5969-9181
                http://orcid.org/0000-0003-3700-8658
                http://orcid.org/0000-0003-3559-4710
                Article
                1239
                10.1038/s41416-020-01239-z
                8007692
                33531689
                fb04e79a-0cd4-4983-a8ee-e5a4a4c51201
                © The Author(s), under exclusive licence to Cancer Research UK 2021

                Note This work is published under the standard license to publish agreement. After 12 months the work will become freely available and the license terms will switch to a Creative Commons Attribution 4.0 International (CC BY 4.0).

                History
                : 20 May 2020
                : 5 November 2020
                : 10 December 2020
                Funding
                Funded by: Instituto de Salud Carlos III (PI19/01128) cofinanced by Fondos Feder and Joint Translational Call for Proposals 2015 (JTC 2015) TRANSCAN-2 (code: TRS-2016-00000371) Merck & Co., Inc (USA)
                Funded by: Merck & Co., Inc (USA)
                Funded by: Merck & Co., Inc (USA)
                Funded by: Instituto de Salud Carlos III (PI19/01128) cofinanced by Fondos Feder and Joint Translational Call for Proposals 2015 (JTC 2015) TRANSCAN-2 (code: TRS-2016-00000371) Merck & Co., Inc (USA)
                Funded by: P.B. was supported by Spanish Society Against Cancer investigator Award (INVES19041ALVA)
                Funded by: Instituto de Salud Carlos III (PI19/01128) cofinanced by Fondos Feder and Joint Translational Call for Proposals 2015 (JTC 2015) TRANSCAN-2 (code: TRS-2016-00000371) Merck & Co., Inc (USA)
                Categories
                Article
                Custom metadata
                © Cancer Research UK 2021

                Oncology & Radiotherapy
                oncology,cancer
                Oncology & Radiotherapy
                oncology, cancer

                Comments

                Comment on this article

                Related Documents Log