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

      CD5: from antiquated T cell marker to immunotherapy’s new hope

      brief-report

      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

          In a recent study published in Science, He et al. investigated the role of CD5 on dendritic cells (DCs) in priming effector T cells. 1 By proving the importance of CD5 in engaging effective antitumor immune responses, they highlighted the untapped potential of CD5 for targeted immunotherapy. Checkpoint-inhibitor immunotherapy can safely be considered as a revolutionary development in cancer treatment. However, serious side-effects are frequent, prediction of therapy success is still challenging and nonresponding patients are omnipresent. Improved understanding of the interaction of antigen-presenting with effector cells, especially in the tumor microenvironment (TME), might lay the foundation for advanced therapeutic approaches. DCs, the most important antigen-presenting cells, are further divided in CD5+ and CD5- subsets with differential capacity for T cell activation. Thus, He et al. scrutinized the functional role of CD5 in immunologic interactions and its prognostic value for patient survival. One major finding was the induction of T cell proliferation by CD1c+ CD5+ DCs isolated from both skin and lymph nodes (see Fig. 1a). T cell proliferation was additionally accompanied by increased production of the activation markers IFNγ and TNFα as well as the effector molecules granzyme B and perforin. Moreover, T cell proliferation could further be improved by increased expression levels of CD5 on DCs. Beside the dependence of stimulating DC capacity on CD5, the authors also showed strengthened immune response against classical viral recall antigens (influenza, Epstein Barr and cytomegalovirus) when these were presented on CD5+ DCs. The stimulation of T cells with antigen-loaded CD5- or CD14+ DCs was significantly less effective. Fig. 1 The role of CD5 in tumor immunology and therapeutic chances. a CD5+ cDC2 play an important role in tumor immunology. (upper left) Under influence of IL-6, hematopoietic progenitor cells differentiate into CD5+ cDC2. (upper right) CD5+ cDC2 induce strong proliferation of CD5high T cells, while proliferation induced by CD5- cDC2 is less effective and limited to CD5low T cells. (lower left) Successful T-cell-induced tumor cell elimination depends on CD5 expression on both cDC2 and T cells. Beside interaction of CD5+ cDC2 with CD5 on T cells, yet unidentified receptors might be involved, too. (lower right) Anti-PD-1 therapy is most effective when CD5+ cDC2 are present in the TME and can induce activation and proliferation of CD5+ T cells. b CD5’s role in tumor immunology imply therapeutic chances. (upper left) The use of cross-linking antibodies could enhance antitumor response. (upper right) CD5 could be used for the selection of DCs instead of CD14 to improve T cell activating capacity. (lower left) Biopsy screening for CD5+ cDC2 might guide therapeutic decisions for immunotherapy. (lower right) In the autoimmune disease situation, blocking CD5 could regulate immune activity. cDC2 conventional dendritic cells 2, IL-6 interleukin 6, MHC major histocompatibility complex, PD-1 programmed cell death protein 1, TCR T cell receptor. Created with BioRender.com Further, in vivo experiments showed the functional dependence on CD5+ DCs for strong antitumor responses. When CD5+ DC were depleted, a majority of mice was unable to initiate a forceful immune reaction involving CD4+ and CD8+ effector T cells, while mice with unaltered DCs could and did successfully reject their tumors. Aside from CD5’s role on DCs, the authors also elucidated CD5 expression on T cells and showed that it very closely mirrored the CD5 expression of DCs. It had been clear, that the antiquated T cell marker CD5 is important for T cell receptor signaling fine tuning and for protection from activation-induced cell death. 2 Historically, CD5 has been considered as a purely negative regulator but more recent data imply, that via signaling through NF-κB, it helps maintaining higher levels of intracellular IκBα, which enables CD5+ T cells to better persist as effector/memory cells. 3 Together with the data of the Klechevsky laboratory on CD5-mediated inhibition of apoptosis in DCs, this hints towards a general and assumedly context-dependent mechanism. Remarkable differences regarding CD5-proficient and CD5-deficient immune cells were observed in experimental immunotherapy. The authors validated the necessity of CD5 on both DCs and T cells for noteworthy tumor regression induced by immune checkpoint inhibitor treatment (anti-PD-1 or anti-CTLA-4). Further, He et al. validated older observations of reduced CD5 expression in the TME compared to healthy tissue, hinting at a tumor-induced downregulation. 4 The parallel to specific modulation of known immune checkpoint molecules like PD-1 in the TME is striking. While immune checkpoints like PD-1 are expressed on T cells, they can also be found on CD5+ DCs. Taking the demonstrated utmost importance of CD5 into account, its induction by immune checkpoint inhibitors could restore tumor-suppressed DC activity via inducing CD5 expression and the proliferation of CD5+ DCs. The subsequent activation of tumor-reactive T cells could tip the scale back in the direction of equilibrium or even tumor elimination. This hypothesis is further strengthened by the increase of IL-6 in the TME of anti-PD1-treated mice and the observation of IL-6-dependent progenitor cell differentiation into CD5+ DCs. The study of He and coworkers adds further important pieces to the puzzle of tumor-immune cell interaction in the TME of affected patients. Moreover, it raises a number of important questions like the role of CD5 on further immune cell types like B or NK cells and their subsets as well as their potential involvement in the CD5-dependent immune regulation. B cells with their strong antigen-presenting capacity are regularly present in the TME, but contrary to DCs and T cells, CD5+ B cells seem to be suppressive, 5 thereby highlighting the duality in function of CD5, which must be clarified. However, CD5+ DCs seem to be pivotal for immunotherapy success. Retrospective analyses of tumor patient cohorts treated with checkpoint inhibitors should be performed to clarify CD5’s therapeutic impact. If significantly correlating with response, CD5 could indeed be the desired biomarker to amend or even overcome the insufficient predictive significance of immune infiltration, PD-L1 expression or tumor mutational burden (see Fig. 1b). For cellular therapy approaches, the results of He and coworkers have even stronger implications. Currently, DCs are mainly selected based on CD14 expression. If CD5 expression is better suited than CD14 to define T cell-activating DCs, CD5 determination or even selection should be taken into consideration for future DC vaccine preparation. As the authors also showed the positive effects of crosslinking and agonistic CD5 antibodies for boosting T cell responses, further research should focus on that, too. However, since CD5 might be a crucial factor for initiating strong immune reactions, this opens up the possibility of targeting this protein in overreacting immune cells present in autoimmunity (see Fig. 1b). Thus, CD5-targeted therapy approaches must be developed in a cell-specific and context-dependent manner to avoid undesirable interferences in the CD5+ cells’ multifunctional network. With a positive prognostic value of the CD5+ DC signature observed for melanoma, lung squamous cell carcinoma, sarcoma, breast cancer, cervical squamous cell carcinoma and endocervical adenocarcinoma patient cohorts, CD5 could be of ubiquitous importance in a broad spectrum of tumors. But these data from The Cancer Genome Atlas (TCGA) need to be validated in independent clinical sample collections to round out this promising picture. It seems reasonable to assume that there will, for example, be clear differences between hypermutated and non-hypermutated subgroups of a given cancer type. Ultimately, unraveling the presumable even more complex function of CD5 in cell fate decisions under pathological conditions, will clarify its duality in function and trigger therapeutic developments. Considering all aspects of CD5’s involvement in antitumoral immune response, the study of He and colleagues might well mark the emergence of a new phase particularly in immunotherapy.

          Related collections

          Most cited references5

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

          In situ sensory adaptation of tumor-infiltrating T lymphocytes to peptide-MHC levels elicits strong antitumor reactivity.

          We have isolated from tumor-infiltrating lymphocytes (TIL) and PBL of a lung carcinoma patient several tumor-specific T cell clones displaying similar peptide-MHC tetramer staining and expressing a unique TCR. Although these clones elicited identical functional avidity and similar cytolytic potential, only T cell clones derived from TIL efficiently lysed autologous tumor cells. Interestingly, all of these clones expressed the same T cell surface markers except for the TCR inhibitory molecule CD5, which was expressed at much lower levels in TIL than in PBL. Video-imaging recordings demonstrated that, although both T cell clones could form stable conjugates with tumor cells, the Ca(2+) response occurred in TIL clones only. Significantly, analysis of a panel of circulating clones indicated that antitumor cytolytic activity was inversely proportional to CD5 expression levels. Importantly, CD5 levels in TIL appeared to parallel the signaling intensity of the TCR/peptide-MHC interaction. Thus, in situ regulation of CD5 expression may be a strategy used by CTL to adapt their sensitivity to intratumoral peptide-MHC levels.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            CD5 dynamically calibrates basal NF-κB signaling in T cells during thymic development and peripheral activation

            CD5 is a surface receptor that negatively regulates T cell activation, but T cells expressing more CD5 have been shown to survive better in the effector/memory phase after pathogenic insult. Our data suggest a mechanistic framework for this duality, by identifying the NF-κB pathway as a target of CD5 signaling. CD5 controls the amounts of lymphocyte IκBα independently of the canonical binding partner SHP-1 and in addition to T cell receptor signals. The increased basal levels of NF-κB transcription factors in CD5 hi cells also conferred an NF-κB–dependent survival advantage. Immature T cells undergo a process of positive selection in the thymus when their new T cell receptor (TCR) engages and signals in response to self-peptides. As the T cell matures, a slew of negative regulatory molecules, including the inhibitory surface glycoprotein CD5, are up-regulated in proportion to the strength of the self-peptide signal. Together these regulators dampen TCR-proximal signaling and help avoid any subsequent peripheral activation of T cells by self-peptides. Paradoxically, antigen-specific T cells initially expressing more CD5 (CD5 hi ) have been found to better persist as effector/memory cells after a peripheral challenge. The molecular mechanisms underlying such a duality in CD5 function is not clear. We found that CD5 alters the basal activity of the NF-κB signaling in resting peripheral T cells. When CD5 was conditionally ablated, T cells were unable to maintain higher expression of the cytoplasmic NF-κB inhibitor IκBα. Consistent with this, resting CD5 hi T cells expressed more of the NF-κB p65 protein than CD5 lo cells, without significant increases in transcript levels, in the absence of TCR signals. This posttranslationally stabilized cellular NF-κB depot potentially confers a survival advantage to CD5 hi T cells over CD5 lo ones. Taken together, these data suggest a two-step model whereby the strength of self-peptide–induced TCR signal lead to the up-regulation of CD5, which subsequently maintains a proportional reserve of NF-κB in peripheral T cells poised for responding to agonistic antigen-driven T cell activation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Human CD5 protects circulating tumor antigen-specific CTL from tumor-mediated activation-induced cell death.

              We previously characterized several tumor-specific T cell clones from PBL and tumor-infiltrating lymphocytes of a lung cancer patient with identical TCR rearrangements and similar lytic potential, but with different antitumor response. A role of the TCR inhibitory molecule CD5 to impair reactivity of peripheral T cells against the tumor was found to be involved in this process. In this report, we demonstrate that CD5 also controls the susceptibility of specific T cells to activation-induced cell death (AICD) triggered by the tumor. Using a panel of tumor-infiltrating lymphocytes and PBL-derived clones expressing different levels of CD5, our results indicate that T lymphocyte AICD in response to the cognate tumor is inversely proportional to the surface expression level of CD5. They also suggest a direct involvement of CD5 in this process, as revealed by an increase in tumor-mediated T lymphocyte AICD following neutralization of the molecule with specific mAb. Mechanistically, our data indicate that down-regulation of FasL expression and subsequent inhibition of caspase-8 activation are involved in CD5-induced T cell survival. These results provide evidence for a role of CD5 in the fate of peripheral tumor-specific T cells and further suggest its contribution to regulate the extension of CTL response against tumor.
                Bookmark

                Author and article information

                Contributors
                Michael.linnebacher@med.uni-rostock.de
                Journal
                Signal Transduct Target Ther
                Signal Transduct Target Ther
                Signal Transduction and Targeted Therapy
                Nature Publishing Group UK (London )
                2095-9907
                2059-3635
                25 May 2023
                25 May 2023
                2023
                : 8
                : 216
                Affiliations
                GRID grid.413108.f, ISNI 0000 0000 9737 0454, Molecular Oncology and Immunotherapy, Clinic of General Surgery, , Rostock University Medical Center, ; 18057 Rostock, Germany
                Author information
                http://orcid.org/0000-0002-8112-4392
                http://orcid.org/0000-0001-8054-1402
                Article
                1494
                10.1038/s41392-023-01494-5
                10212910
                37230972
                aa874e04-8c18-4b22-a8a1-f27307fc0b1a
                © The Author(s) 2023

                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
                : 11 April 2023
                : 28 April 2023
                : 7 May 2023
                Categories
                Research Highlight
                Custom metadata
                © West China Hospital, Sichuan University 2023

                tumour immunology,preclinical research,immunotherapy

                Comments

                Comment on this article