30
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Glutaminyl cyclase is an enzymatic modifier of the CD47- SIRPα axis and target for cancer immunotherapy

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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

          Cancer cells are able to evade immune surveillance through the expression of inhibitory ligands that bind their cognate receptors on immune effector cells. Expression of Programmed Death-Ligand 1 (PD-L1) in tumor micro-environments is a major immune checkpoint for tumor-specific T cell responses, by binding to Programmed Cell Death protein-1 (PD-1) on activated and dysfunctional T cells 1 . The activity of myeloid cells, such as macrophages and neutrophils, is likewise regulated by a balance between stimulatory and inhibitory signals. In particular, cell surface expression of the CD47 protein has been shown to form a “don’t eat me” signal on tumor cells, by binding to SIRPα expressed on myeloid cells 2–5 . Using a haploid genetic screen, we here identify glutaminyl-peptide cyclotransferase-like (QPCTL) as a major component of the CD47-SIRPα checkpoint. Biochemical analysis demonstrates that QPCTL is critical for pyroglutamate formation on CD47 at the SIRPα binding site shortly after biosynthesis. Both genetic and pharmacological interference with QPCTL activity enhances antibody-dependent cellular phagocytosis and cellular cytotoxicity of tumor cells. Furthermore, interference with QPCTL expression leads to a major increase in neutrophil-mediated tumor cell killing in vivo. These data identify QPCTL as a novel target to interfere with the CD47 pathway, and thereby augment antibody therapy of cancer.

          Related collections

          Most cited references33

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

          The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer.

          Immune checkpoint inhibitors, including those targeting CTLA-4/B7 and the PD-1/PD-L1 inhibitory pathways, are now available for clinical use in cancer patients, with other interesting checkpoint inhibitors being currently in development. Most of these have the purpose to promote adaptive T cell-mediated immunity against cancer. Here, we review another checkpoint acting to potentiate the activity of innate immune cells towards cancer. This innate immune checkpoint is composed of what has become known as the 'don't-eat me' signal CD47, which is a protein broadly expressed on normal cells and often overexpressed on cancer cells, and its counter-receptor, the myeloid inhibitory immunoreceptor SIRPα. Blocking CD47-SIRPα interactions has been shown to promote the destruction of cancer cells by phagocytes, including macrophages and neutrophils. Furthermore, there is growing evidence that targeting of the CD47-SIRPα axis may also promote antigen-presenting cell function and thereby stimulate adaptive T cell-mediated anti-cancer immunity. The development of CD47-SIRPα checkpoint inhibitors and the potential side effects that these may have are discussed. Collectively, this identifies the CD47-SIRPα axis as a promising innate immune checkpoint in cancer, and with data of the first clinical studies with CD47-SIRPα checkpoint inhibitors expected within the coming years, this is an exciting and rapidly developing field.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Alternative 3'UTRs act as scaffolds to regulate membrane protein localization

            About half of human genes use alternative cleavage and polyadenylation (ApA) to generate mRNA transcripts that differ in the length of their 3' untranslated regions (3'UTRs) while producing the same protein 1–3 . Here we show in human cell lines that alternative 3' UTRs differentially regulate the localization of membrane proteins. The long 3'UTR of CD47 enables efficient cell surface expression of CD47 protein, whereas the short 3'UTR primarily localizes CD47 protein to the endoplasmic reticulum. CD47 protein localization occurs post-translationally and independently of RNA localization. In our model of 3' UTR-dependent protein localization, the long 3' UTR of CD47 acts as a scaffold to recruit a protein complex containing the RNA-binding protein HuR (also known as ELAVL1) and SET 4 to the site of translation. This facilitates interaction of SET with the newly translated cytoplasmic domains of CD47 and results in subsequent translocation of CD47 to the plasma membrane via activated RAC1 5 . We also show that CD47 protein has different functions depending on whether it was generated by the short or long 3'UTR isoforms. Thus, ApA contributes to the functional diversity of the proteome without changing the amino acid sequence. 3' UTR-dependent protein localization has the potential to be a widespread trafficking mechanism for membrane proteins because HuR binds to thousands of mRNAs 6–9 , and we show that the long 3' UTRs of CD44, ITGA1 and TNFRSF13C, which are bound by HuR, increase surface protein expression compared to their corresponding short 3' UTRs. We propose that during translation the scaffold function of 3' UTRs facilitates binding of proteins to nascent proteins to direct their transport or function—and that this role of 3' UTRs can be regulated by ApA.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Durable antitumor responses to CD47 blockade require adaptive immune stimulation.

              Therapeutic antitumor antibodies treat cancer by mobilizing both innate and adaptive immunity. CD47 is an antiphagocytic ligand exploited by tumor cells to blunt antibody effector functions by transmitting an inhibitory signal through its receptor signal regulatory protein alpha (SIRPα). Interference with the CD47-SIRPα interaction synergizes with tumor-specific monoclonal antibodies to eliminate human tumor xenografts by enhancing macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), but synergy between CD47 blockade and ADCP has yet to be demonstrated in immunocompetent hosts. Here, we show that CD47 blockade alone or in combination with a tumor-specific antibody fails to generate antitumor immunity against syngeneic B16F10 tumors in mice. Durable tumor immunity required programmed death-ligand 1 (PD-L1) blockade in combination with an antitumor antibody, with incorporation of CD47 antagonism substantially improving response rates. Our results highlight an underappreciated contribution of the adaptive immune system to anti-CD47 adjuvant therapy and suggest that targeting both innate and adaptive immune checkpoints can potentiate the vaccinal effect of antitumor antibody therapy.
                Bookmark

                Author and article information

                Journal
                9502015
                Nat Med
                Nat. Med.
                Nature medicine
                1078-8956
                1546-170X
                18 October 2019
                04 March 2019
                April 2019
                17 February 2020
                : 25
                : 4
                : 612-619
                Affiliations
                [1 ]Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
                [2 ]Laboratory for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
                [3 ]Division of Biochemistry, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
                [4 ]Department of Blood Cell Research, Sanquin Research, Landsteiner Laboratory, and Department of Molecular Cell Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
                [5 ]Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
                [6 ]Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
                [7 ]Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
                [8 ]Institute for Chemical Immunology
                [9 ]Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
                Author notes
                [* ]Correspondence to t.schumacher@ 123456nki.nl
                [†, #]

                These authors contributed equally

                Article
                EMS81308
                10.1038/s41591-019-0356-z
                7025889
                30833751
                c8808c1a-5455-4ed1-ab8b-fde8ecd9153a

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Medicine
                Medicine

                Comments

                Comment on this article