Immunological Mechanisms Responsible for Radiation-Induced Abscopal Effect

María E Rodríguez-Ruiz, Claire Vanpouille-Box, Ignacio Melero, Silvia Chiara Formenti, Sandra Demaria, María E Rodríguez-Ruiz, Claire Vanpouille-Box, Ignacio Melero, Silvia Chiara Formenti, Sandra Demaria

Abstract

Radiotherapy has been used for more than a hundred years as a local tumor treatment. The occurrence of systemic antitumor effects manifesting as regression of tumors outside of the irradiated field (abscopal effect) was occasionally observed but deemed too rare and unpredictable to be a therapeutic goal. This has changed with the advent of immunotherapy. Remarkable systemic effects have been observed in patients receiving radiotherapy to control tumors that were progressing during immune checkpoint blockade, stimulating interest in using radiation to overcome primary and acquired cancer resistance to immunotherapy. Here, we review the immunological mechanisms that are responsible for the ability of focal radiation to promote antitumor T cell responses that mediate tumor rejection and, in some cases, result in systemic effects.

Keywords: DNA damage; abscopal effect; antitumor immunity; immunotherapy; ionizing radiation; tumor microenvironment.

Copyright © 2018 Elsevier Ltd. All rights reserved.

Figures

Figure 1:. Radiation-induced secretion of IFN-I is…
Figure 1:. Radiation-induced secretion of IFN-I is critical for abscopal responses.
Ionizing radiation leads to the release of tumor associated antigens (TAA) and damaged DNA that can stimulate the production of type-I interferon via the cGAS/STING pathway in tumor cells. The subsequent secretion of IFN-I and interferon-stimulated genes (including CXCL10) promote the recruitment and activation of BATF3-DCs. Once in the tumor, BATF3-DCs take up the TAA and tumor-derived DNA to further stimulate the production of IFN-I. Activated BATF3-DCs then migrate to the tumor draining lymph node where they can prime CD8+ T cells to initiate cytotoxic T-cell responses. Once activated, cytotoxic T lymphocytes (CTLs) migrate to the irradiated tumor and eliminate residual cancer cells, and to distant metastatic sites leading to systemic tumor regression (abscopal effect).
Figure I:
Figure I:
A- Direct effect. Following X-ray, an electron is ejected and directly damages DNA. B- Indirect effect. The incident photon ejects an electron, which creates a free radical. The diffusing free radical then causes damage to DNA.
Figure II:
Figure II:
Major type of DBS breaks. Simple DSBs involve two broken DNA ends (i.e. two-ended DSB) in close proximity. Complex DSBs consist in two broken DNA ends in proximity to additional DNA damage (i.e., cross-links, single stranded breaks, etc...) or a DSB within a replication fork (one-ended DSB).

References

    1. Formenti SC and Demaria S (2009) Systemic effects of local radiotherapy. Lancet Oncol 10 (7), 718–26.
    1. Abuodeh Y et al. (2016) Systematic review of case reports on the abscopal effect. Curr Probl Cancer 40 (1), 25–37.
    1. Sharma P and Allison JP (2015) Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161 (2), 205–14.
    1. Demaria S et al. (2005) Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 11 (2 Pt 1), 728–34.
    1. Deng L et al. (2014) Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 124 (2), 687–95.
    1. Postow MA et al. (2012) Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 366 (10), 925–31.
    1. Grimaldi AM et al. (2014) Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 3, e28780.
    1. Kang J et al. (2016) Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer 4, 51.
    1. Willers H and Held KD (2006) Introduction to clinical radiation biology. Hematol Oncol Clin North Am 20 (1), 1–24.
    1. Jeggo PA et al. (2016) DNA repair, genome stability and cancer: a historical perspective. Nat Rev Cancer 16 (1), 35–42.
    1. Harding SM et al. (2017) Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548 (7668), 466–470.
    1. Mackenzie KJ et al. (2017) cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548 (7668), 461–465.
    1. Bartsch K et al. (2017) Absence of RNase H2 triggers generation of immunogenic micronuclei removed by autophagy. Hum Mol Genet 26 (20), 3960–3972.
    1. Cai X et al. (2014) The cGAS-cGAMP-STING pathway of cytosolic DNA sensing and signaling. Mol Cell 54 (2), 289–96.
    1. Vanpouille-Box C et al. (2017) DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 8, 15618.
    1. Deng L et al. (2014) STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 41 (5), 843–52.
    1. Galluzzi L et al. (2017) Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol 17 (2), 97–111.
    1. Apetoh L et al. (2007) Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 13 (9), 1050–9.
    1. Obeid M et al. (2007) Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ 14 (10), 1848–50.
    1. Shrivastav M et al. (2008) Regulation of DNA double-strand break repair pathway choice. Cell Res 18 (1), 134–47.
    1. Meng Y et al. (2012) Radiation-inducible immunotherapy for cancer: senescent tumor cells as a cancer vaccine. Mol Ther 20 (5), 1046–55.
    1. Dou Z et al. (2017) Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550 (7676), 402–406.
    1. Ranoa DR et al. (2016) Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs. Oncotarget 7 (18), 26496–515.
    1. Sharma P and Allison JP (2015) The future of immune checkpoint therapy. Science 348 (6230), 56–61.
    1. Sharma P et al. (2017) Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 168 (4), 707–723.
    1. Barsoum IB et al. (2014) A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 74 (3), 665–74.
    1. Dewhirst MW et al. (2008) Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer 8 (6), 425–37.
    1. Noman MZ et al. (2014) PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 211 (5), 781–90.
    1. Voron T et al. (2014) Control of the immune response by pro-angiogenic factors. Front Oncol 4, 70.
    1. Barsoum IB et al. (2011) Hypoxia induces escape from innate immunity in cancer cells via increased expression of ADAM10: role of nitric oxide. Cancer Res 71 (24), 7433–41.
    1. Palazon A et al. (2017) An HIF-1alpha/VEGF-A Axis in Cytotoxic T Cells Regulates Tumor Progression. Cancer Cell 32 (5), 669–683e5.
    1. Moeller BJ et al. (2004) Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell 5 (5), 429–41.
    1. Li F et al. (2007) Regulation of HIF-lalpha stability through S-nitrosylation. Mol Cell 26 (1), 63–74.
    1. Hellevik T and Martinez-Zubiaurre I (2014) Radiotherapy and the tumor stroma: the importance of dose and fractionation. Front Oncol 4, 1.
    1. Tommelein J et al. (2018) Radiotherapy-Activated Cancer-Associated Fibroblasts Promote Tumor Progression through Paracrine IGF1R Activation. Cancer Res 78 (3), 659–670.
    1. Spadaro O et al. (2017) IGF1 Shapes Macrophage Activation in Response to Immunometabolic Challenge. Cell Rep 19 (2), 225–234.
    1. Barcellos-Hoff MH and Cucinotta FA (2014) New tricks for an old fox: impact of TGFbeta on the DNA damage response and genomic stability. Sci Signal 7 (341), re5.
    1. Jobling MF et al. (2006) Isoform-specific activation of latent transforming growth factor beta (LTGF-beta) by reactive oxygen species. Radiat Res 166 (6), 839–48.
    1. Vanpouille-Box C et al. (2015) TGFbeta Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res 75 (11), 2232–42.
    1. Xu J et al. (2013) CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res 73 (9), 2782–94.
    1. Kioi M et al. (2010) Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 120 (3), 694–705.
    1. Kalbasi A et al. (2017) Tumor-Derived CCL2 Mediates Resistance to Radiotherapy in Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 23 (1), 137–148.
    1. Sanford DE et al. (2013) Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 19 (13), 3404–15.
    1. Dovedi SJ et al. (2014) Acquired Resistance to Fractionated Radiotherapy Can Be Overcome by Concurrent PD-L1 Blockade. Cancer Research 74 (19), 5458–5468.
    1. Matsumura S et al. (2008) Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol 181 (5), 3099–107.
    1. Burnette BC et al. (2011) The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res 71 (7), 2488–96.
    1. Lugade AA et al. (2005) Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol 174 (12), 7516–23.
    1. Klug F et al. (2013) Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 24 (5), 589–602.
    1. Gasser S et al. (2005) The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436 (7054), 1186–90.
    1. Lam AR et al. (2014) RAE1 ligands for the NKG2D receptor are regulated by STING-dependent DNA sensor pathways in lymphoma. Cancer Res 74 (8), 2193–2203.
    1. Ruocco MG et al. (2012) Suppressing T cell motility induced by anti-CTLA-4 monotherapy improves antitumor effects. J Clin Invest 122 (10), 3718–30.
    1. Chakraborty M et al. (2003) Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 170 (12), 6338–47.
    1. Reits EA et al. (2006) Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 203 (5), 1259–71.
    1. Rudqvist NP et al. (2018) Radiotherapy and CTLA-4 Blockade Shape the TCR Repertoire of Tumor-Infiltrating T Cells. Cancer Immunol Res 6 (2), 139–150.
    1. Twyman-Saint Victor C et al. (2015) Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520 (7547), 373–7.
    1. Zhang B et al. (2007) Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J Exp Med 204 (1), 49–55.
    1. Brody JD et al. (2010) In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol 28 (28), 4324–32.
    1. Formenti SC and Demaria S (2012) Radiation therapy to convert the tumor into an in situ vaccine. Int J Radiat Oncol Biol Phys 84 (4), 879–80.
    1. Lee Y et al. (2009) Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 114 (3), 589–95.
    1. Finn OJ (2017) The dawn of vaccines for cancer prevention. Nat Rev Immunol.
    1. Golden EB et al. (2015) Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 16 (7), 795–803.
    1. Golden EB et al. (2013) An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res 1 (6), 365–72.
    1. Zatloukal P et al. (2009) Randomized phase II clinical trial comparing tremelimumab (CP-675, 206) with best supportive care (BSC) following first-line platinum-based therapy in patients (pts) with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 27 (15S), 8071.
    1. Lynch TJ et al. (2012) Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol 30 (17), 2046–54.
    1. Demaria S et al. (2016) Radiotherapy: Changing the Game in Immunotherapy. Trends Cancer 2 (6), 286–294.
    1. Dewan MZ et al. (2009) Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 15 (17), 5379–88.
    1. Filatenkov A et al. (2015) Ablative Tumor Radiation Can Change the Tumor Immune Cell Microenvironment to Induce Durable Complete Remissions. Clin Cancer Res 21 (16), 3727–39.
    1. Sanchez-Paulete AR et al. (2017) Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy. Ann Oncol 28 (suppl_12), xii44–xii55.
    1. Stetson DB et al. (2008) Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell 134 (4), 587–98.
    1. Vanpouille-Box C et al. (2018) Toward Precision Radiotherapy for Use with Immune Checkpoint Blockers. Clin Cancer Res 24 (2), 259–265.
    1. Menis J et al. (2016) The European Organization for Research and Treatment of Cancer perspective on designing clinical trials with immune therapeutics. Ann Transl Med 4 (14), 267.
    1. Shaverdian N et al. (2017) Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol 18 (7), 895–903.
    1. Koller KM et al. (2017) Improved survival and complete response rates in patients with advanced melanoma treated with concurrent ipilimumab and radiotherapy versus ipilimumab alone. Cancer Biol Ther 18 (1), 36–42.
    1. Tang C et al. (2017) Ipilimumab with Stereotactic Ablative Radiation Therapy: Phase I Results and Immunologic Correlates from Peripheral T Cells. Clin Cancer Res 23 (6), 1388–1396.
    1. Hiniker SM et al. (2016) A Prospective Clinical Trial Combining Radiation Therapy With Systemic Immunotherapy in Metastatic Melanoma. Int J Radiat Oncol Biol Phys 96 (3), 578–88.
    1. Kwon ED et al. (2014) Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184–043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 15 (7), 700–12.
    1. Golden EB and Formenti SC (2015) Radiation therapy and immunotherapy: growing pains. Int J Radiat Oncol Biol Phys 91 (2), 252–4.
    1. Antonia SJ et al. (2017) Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 377 (20), 1919–1929.
    1. Golden EB and Formenti SC (2014) Is tumor (R)ejection by the immune system the “5th R” of radiobiology? Oncoimmunology 3 (1), e28133.
    1. Ceccaldi R et al. (2016) Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol 26 (1), 52–64.
    1. Goodarzi AA et al. (2010) The influence of heterochromatin on DNA double strand break repair: Getting the strong, silent type to relax. DNA Repair (Amst) 9 (12), 1273–82.
    1. Shibata A et al. (2014) DNA double-strand break repair pathway choice is directed by distinct MRE11 nuclease activities. Mol Cell 53 (1), 7–18.
    1. Arnoult N et al. (2017) Regulation of DNA repair pathway choice in S and G2 phases by the NHEJ inhibitor CYREN. Nature 549 (7673), 548–552.

Source: PubMed

3
Sottoscrivi