4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      ATR-mediated CD47 and PD-L1 up-regulation restricts radiotherapy-induced immune priming and abscopal responses in colorectal cancer

      1 , 2 , 3 , 4 , 5 , 1 , 2 , 1 , 2 , 1 , 2 , 2 , 6 , 2 , 7 , 8 , 9 , 8 , 1 , 2 , 1 , 2 , 1 , 3 , 2 , 6 , 1 , 2 , 10 , 2 , 11 , 12 , 1 , 2 , 13 , 1 , 2 , 1 , 1 , 1 , 1 , 2 , 1 , 2 , 1 , 2 , 1 , 1 , 2 , 3 , 14 , 2 , 7 , 15 , 16 , 2 , 15 , 1 , 2 , 1 , 2
      Science Immunology
      American Association for the Advancement of Science (AAAS)

      Read this article at

      ScienceOpenPublisherPubMed
      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

          Radiotherapy (RT) of colorectal cancer (CRC) can prime adaptive immunity against tumor-associated antigen (TAA)–expressing CRC cells systemically. However, abscopal tumor remissions are extremely rare, and the postirradiation immune escape mechanisms in CRC remain elusive. Here, we found that irradiated CRC cells used ATR-mediated DNA repair signaling pathway to up-regulate both CD47 and PD-L1, which through engagement of SIRPα and PD-1, respectively, prevented phagocytosis by antigen-presenting cells and thereby limited TAA cross-presentation and innate immune activation. This postirradiation CD47 and PD-L1 up-regulation was observed across various human solid tumor cells. Concordantly, rectal cancer patients with poor responses to neoadjuvant RT exhibited significantly elevated postirradiation CD47 levels. The combination of RT, anti-SIRPα, and anti–PD-1 reversed adaptive immune resistance and drove efficient TAA cross-presentation, resulting in robust TAA-specific CD8 T cell priming, functional activation of T effectors, and increased T cell clonality and clonal diversity. We observed significantly higher complete response rates to RT/anti-SIRPα/anti–PD-1 in both irradiated and abscopal tumors and prolonged survival in three distinct murine CRC models, including a cecal orthotopic model. The efficacy of triple combination therapy was STING dependent as knockout animals lost most benefit of adding anti-SIRPα and anti–PD-1 to RT. Despite activation across the myeloid stroma, the enhanced dendritic cell function accounts for most improvements in CD8 T cell priming. These data suggest ATR-mediated CD47 and PD-L1 up-regulation as a key mechanism restraining radiation-induced immune priming. RT combined with SIRPα and PD-1 blockade promotes robust antitumor immune priming, leading to systemic tumor regressions.

          Abstract

          ATR signaling up-regulates CD47 and PD-L1 in irradiated CRC cells, preventing phagocytosis and tumor antigen cross-presentation by APCs.

          Engulfing to improve radiation

          Despite the wide use of radiation therapy (RT) in colorectal cancer, many patients experience progression at non-irradiated sites of disease. Thus, it is crucial to improve the abscopal effects of RT. Here, Hsieh et al. used mouse modeling and human cancer cell lines to show that RT increased the expression of CD47 and PD-L1 in a DNA repair signaling–dependent manner. Targeting CD47 and PD-L1 with anti-SIRPα and anti-PD-1, respectively, in combination with RT led to clearance of primary tumors and robust abscopal effects. This triple combination depended on host STING expression, leading to improved tumor cell phagocytosis and subsequent cross-priming of tumor antigen–specific T cells. Together, these data suggest that targeting immune checkpoints and phagocytosis during RT may help patients with colorectal cancer.

          Related collections

          Most cited references85

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

          Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

          This article provides an update on the global cancer burden using the GLOBOCAN 2020 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer. Worldwide, an estimated 19.3 million new cancer cases (18.1 million excluding nonmelanoma skin cancer) and almost 10.0 million cancer deaths (9.9 million excluding nonmelanoma skin cancer) occurred in 2020. Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), followed by lung (11.4%), colorectal (10.0 %), prostate (7.3%), and stomach (5.6%) cancers. Lung cancer remained the leading cause of cancer death, with an estimated 1.8 million deaths (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%) cancers. Overall incidence was from 2-fold to 3-fold higher in transitioned versus transitioning countries for both sexes, whereas mortality varied <2-fold for men and little for women. Death rates for female breast and cervical cancers, however, were considerably higher in transitioning versus transitioned countries (15.0 vs 12.8 per 100,000 and 12.4 vs 5.2 per 100,000, respectively). The global cancer burden is expected to be 28.4 million cases in 2040, a 47% rise from 2020, with a larger increase in transitioning (64% to 95%) versus transitioned (32% to 56%) countries due to demographic changes, although this may be further exacerbated by increasing risk factors associated with globalization and a growing economy. Efforts to build a sustainable infrastructure for the dissemination of cancer prevention measures and provision of cancer care in transitioning countries is critical for global cancer control.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown

            High-throughput sequencing of messenger RNA (RNA-seq) has become the standard method for measuring and comparing the levels of gene expression in a wide variety of species and conditions. RNA-seq experiments generate very large, complex data sets that demand fast, accurate, and flexible software to reduce the raw read data to comprehensible results. HISAT, StringTie, and Ballgown are free, open-source software tools for comprehensive analysis of RNA-seq experiments. Together, they allow scientists to align reads to a genome, assemble transcripts including novel splice variants, compute the abundance of these transcripts in each sample, and compare experiments to identify differentially expressed genes and transcripts. This protocol describes all the steps necessary to process a large set of raw sequencing reads and create lists of gene transcripts, expression levels, and differentially expressed genes and transcripts. The protocol’s execution time depends on the computing resources, but typically takes under 45 minutes of computer time. Pertea et al. describe a protocol to analyze RNA-seq data using HISAT, StringTie, and Ballgown (the “new Tuxedo” package). The protocol can be used for assembly of transcripts, quantification of gene expression levels and differential expression analysis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway.

              The presence of DNA in the cytoplasm of mammalian cells is a danger signal that triggers host immune responses such as the production of type I interferons. Cytosolic DNA induces interferons through the production of cyclic guanosine monophosphate-adenosine monophosphate (cyclic GMP-AMP, or cGAMP), which binds to and activates the adaptor protein STING. Through biochemical fractionation and quantitative mass spectrometry, we identified a cGAMP synthase (cGAS), which belongs to the nucleotidyltransferase family. Overexpression of cGAS activated the transcription factor IRF3 and induced interferon-β in a STING-dependent manner. Knockdown of cGAS inhibited IRF3 activation and interferon-β induction by DNA transfection or DNA virus infection. cGAS bound to DNA in the cytoplasm and catalyzed cGAMP synthesis. These results indicate that cGAS is a cytosolic DNA sensor that induces interferons by producing the second messenger cGAMP.
                Bookmark

                Author and article information

                Contributors
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                Journal
                Science Immunology
                Sci. Immunol.
                American Association for the Advancement of Science (AAAS)
                2470-9468
                June 10 2022
                June 10 2022
                : 7
                : 72
                Affiliations
                [1 ]Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [2 ]University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
                [3 ]Department of Radiation Oncology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan.
                [4 ]Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL, USA.
                [5 ]Department of Pathology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan.
                [6 ]Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [7 ]Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [8 ]Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [9 ]Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan.
                [10 ]Parker Institute for Cancer Immunotherapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [11 ]Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [12 ]Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [13 ]Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, TX, USA.
                [14 ]Department of Radiation Oncology, Loyola University Stritch School of Medicine, Chicago, IL, USA.
                [15 ]Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                [16 ]Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
                Article
                10.1126/sciimmunol.abl9330
                35687697
                c7e1585c-a5ae-4ae8-b34e-6ab6359b9ea8
                © 2022
                History

                Comments

                Comment on this article