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

      Recent progress in targeting the sialylated glycan-SIGLEC axis in cancer immunotherapy

      review-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

          Malignant tumors are complex structures composed of cancer cells and tumor microenvironmental cells. In this complex structure, cells cross-talk and interact, thus jointly promoting cancer development and metastasis. Recently, immunoregulatory molecule-based cancer immunotherapy has greatly improved treatment efficacy for solid cancers, thus enabling some patients to achieve persistent responses or cure. However, owing to the development of drug-resistance and the low response rate, immunotherapy against the available targets PD-1/PD-L1 or CTLA-4 has limited benefits. Although combination therapies have been proposed to enhance the response rate, severe adverse effects are observed. Thus, alternative immune checkpoints must be identified. The SIGLECs are a family of immunoregulatory receptors (known as glyco-immune checkpoints) discovered in recent years. This review systematically describes the molecular characteristics of the SIGLECs, and discusses recent progress in areas including synthetic ligands, monoclonal antibody inhibitors, and Chimeric antigen receptor T (CAR-T) cells, with a focus on available strategies for blocking the sialylated glycan-SIGLEC axis. Targeting glyco-immune checkpoints can expand the scope of immune checkpoints and provide multiple options for new drug development.

          Related collections

          Most cited references132

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

          CD24 signalling through macrophage Siglec-10 is a new target for cancer immunotherapy

          Summary: Ovarian cancer and triple-negative breast cancer (TNBC) are among the most lethal diseases affecting women, with few targeted therapies and high rates of metastasis. Here we show that CD24 can be the dominant innate immune checkpoint in ovarian cancer and breast cancer, and is a new, promising target for cancer immunotherapy. Cancer cells are capable of evading clearance by macrophages through the overexpression of anti-phagocytic surface proteins, called “don’t eat me” signals, including CD47 1 , programmed cell death ligand 1 (PD-L1) 2 , and the beta-2 microglobulin subunit of the major histocompatibility class I complex (B2M) 3 . Monoclonal antibodies which antagonize the interaction of “don’t eat me” signals with their macrophage-expressed receptors have demonstrated therapeutic potential in several cancers 4–5 . However, variability in the magnitude and durability of the response to these agents has suggested the presence of additional, as yet unknown, “don’t eat me” signals. Here we demonstrate a novel role for tumor-expressed CD24 in promoting immune evasion through its interaction with the inhibitory receptor, Sialic Acid Binding Ig Like Lectin 10 (Siglec-10), expressed by tumor-associated macrophages (TAMs). We observe that many tumors overexpress CD24 and that TAMs express high levels of Siglec-10. Both genetic ablation of CD24 or Siglec-10, and monoclonal antibody blockade of the CD24–Siglec-10 interaction, robustly augment the phagocytosis of all CD24-expressing human tumors tested. Genetic ablation as well as therapeutic blockade of CD24 resulted in a macrophage-dependent reduction of tumor growth and extension of survival, in vivo. These data highlight CD24 as a highly-expressed, anti-phagocytic signal in several cancers and demonstrate the therapeutic potential for CD24-blockade as cancer immunotherapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Siglecs and their roles in the immune system.

            Cell surfaces in the immune system are richly equipped with a complex mixture of glycans, which can be recognized by diverse glycan-binding proteins. The Siglecs are a family of sialic-acid-binding immunoglobulin-like lectins that are thought to promote cell-cell interactions and regulate the functions of cells in the innate and adaptive immune systems through glycan recognition. In this Review, we describe recent studies on signalling mechanisms and discuss the potential role of Siglecs in triggering endocytosis and in pathogen recognition. Finally, we discuss the postulated functions of the recently discovered CD33-related Siglecs and consider the factors that seem to be driving their rapid evolution.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Macrophages as tools and targets in cancer therapy

              Tumour-associated macrophages are an essential component of the tumour microenvironment and have a role in the orchestration of angiogenesis, extracellular matrix remodelling, cancer cell proliferation, metastasis and immunosuppression, as well as in resistance to chemotherapeutic agents and checkpoint blockade immunotherapy. Conversely, when appropriately activated, macrophages can mediate phagocytosis of cancer cells and cytotoxic tumour killing, and engage in effective bidirectional interactions with components of the innate and adaptive immune system. Therefore, they have emerged as therapeutic targets in cancer therapy. Macrophage-targeting strategies include inhibitors of cytokines and chemokines involved in the recruitment and polarization of tumour-promoting myeloid cells as well as activators of their antitumorigenic and immunostimulating functions. Early clinical trials suggest that targeting negative regulators (checkpoints) of myeloid cell function indeed has antitumor potential. Finally, given the continuous recruitment of myelomonocytic cells into tumour tissues, macrophages are candidates for cell therapy with the development of chimeric antigen receptor effector cells. Macrophage-centred therapeutic strategies have the potential to complement, and synergize with, currently available tools in the oncology armamentarium.
                Bookmark

                Author and article information

                Journal
                Cancer Biol Med
                Cancer Biol Med
                CBM
                Cancer Biology & Medicine
                Compuscript (Ireland )
                2095-3941
                15 May 2023
                03 May 2023
                : 20
                : 5
                : 369-384
                Affiliations
                [1 ]Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
                [2 ]Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
                Author notes
                Correspondence to: Yingyan Yu and Wenjie Peng, E-mail: yingyan3y@ 123456sjtu.edu.cn and wenjiep@ 123456sjtu.edu.cn
                Author information
                https://orcid.org/0000-0002-3585-3252
                https://orcid.org/0000-0001-5093-7115
                Article
                j.issn.2095-3941.2023.0046
                10.20892/j.issn.2095-3941.2023.0046
                10246440
                37133224
                3f4c553b-0295-4e48-88f3-83955df4ce9a
                Copyright: © 2023, Cancer Biology & Medicine

                This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY) 4.0, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

                History
                : 09 February 2023
                : 27 March 2023
                Page count
                Figures: 3, Tables: 2, References: 132, Pages: 16
                Funding
                Funded by: Shanghai Science and Technology Committee
                Award ID: 20DZ2201900 to Y.Y.
                Funded by: Shanghai Science and Technology Committee
                Award ID: 23ZR1432500 to W.P.
                Funded by: National Natural Science Foundation of China
                Award ID: 82072602 to Y.Y.
                Funded by: National Natural Science Foundation of China
                Award ID: 91853121
                Funded by: National Natural Science Foundation of China
                Award ID: 21977066
                Funded by: National Natural Science Foundation of China
                Award ID: 22177069 to W.P.
                Funded by: Innovation Foundation of Translational Medicine of Shanghai Jiao Tong University School of Medicine
                Award ID: TM202001 to Y.Y.
                Funded by: Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai
                Award ID: CCTS-2022202 to Y.Y.
                Funded by: Shanghai Pilot Program for Basic Research-Shanghai Jiao Tong University
                Award ID: 21TQ1400210 to W.P.
                Funded by: Medical-Engineering Interdisciplinary Research Foundation of Shanghai Jiao Tong University
                Award ID: YG2022ZD001 to W.P.
                This work was supported by the Shanghai Science and Technology Committee (Grant Nos. 20DZ2201900 to Y.Y. and 23ZR1432500 to W.P.), National Natural Science Foundation of China (Grant Nos. 82072602 to Y.Y.; 91853121, 21977066, and 22177069 to W.P.), Innovation Foundation of Translational Medicine of Shanghai Jiao Tong University School of Medicine (Grant No. TM202001 to Y.Y.), Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai (Grant No. CCTS-2022202 to Y.Y.), Shanghai Pilot Program for Basic Research-Shanghai Jiao Tong University (Grant No. 21TQ1400210 to W.P.), and Medical-Engineering Interdisciplinary Research Foundation of Shanghai Jiao Tong University (Grant No. YG2022ZD001 to W.P.).
                Categories
                Review

                siglec,sialylated glycan,glyco-immune checkpoint,high affinity siglec-ligands,anti-siglec antibodies

                Comments

                Comment on this article