Novel Approaches to Improve the Efficacy of Immuno-Radiotherapy

Maxim Shevtsov, Hiro Sato, Gabriele Multhoff, Atsushi Shibata, Maxim Shevtsov, Hiro Sato, Gabriele Multhoff, Atsushi Shibata

Abstract

Radiotherapy (RT) has been applied for decades as a treatment modality in the management of various types of cancer. Ionizing radiation induces tumor cell death, which in turn can either elicit protective anti-tumor immune responses or immunosuppression in the tumor micromilieu that contributes to local tumor recurrence. Immunosuppression is frequently accompanied by the attraction of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), N2 neutrophils, and by the release of immunosuppressive cytokines (TGF-β, IL-10) and chemokines. Immune checkpoint pathways, particularly of the PD-1/PD-L1 axis, have been determined as key regulators of cancer immune escape. While IFN-dependent upregulation of PD-L1 has been extensively investigated, up-to-date studies indicated the importance of DNA damage signaling in the regulation of PD-L1 expression following RT. DNA damage dependent PD-L1 expression is upregulated by ATM/ATR/Chk1 kinase activities and cGAS/STING-dependent pathway, proving the role of DNA damage signaling in PD-L1 induced expression. Checkpoint blockade immunotherapies (i.e., application of anti-PD-1 and anti-PD-L1 antibodies) combined with RT were shown to significantly improve the objective response rates in therapy of various primary and metastatic malignancies. Further improvements in the therapeutic potential of RT are based on combinations of RT with other immunotherapeutic approaches including vaccines, cytokines and cytokine inducers, and an adoptive immune cell transfer (DCs, NK cells, T cells). In the current review we provide immunological rationale for a combination of RT with various immunotherapies as well as analysis of the emerging preclinical evidences for these therapies.

Keywords: PD-1; PD-L1; immune checkpoint inhibitors; immunosuppression; radiotherapy.

Figures

Figure 1
Figure 1
Radiation-induced immunosuppressive effects in the tumor micromilieu. RT induces recruitment, proliferation and polarization of immunosuppressive cell subtypes including myeloid-derived suppressor cells (MDSCs), M2 tumor-associated macrophages, N2 neutrophils, and regulatory T cells (CD4+CD25+Foxp3+). RT induces increased levels of suppressive factors including nitric oxide synthase (NOS) and reactive nitrogen intermediates (RNI), reactive oxygen species (ROS), cytokines (IL-4, IL-10, TGF-β), matrix metalloproteinases (MMPs), arginase I, collagenase, lipoxygenase (LOX) which in turn leads to the suppression of the T cell activity.
Figure 2
Figure 2
Regulation of PD-L1 expression in response to DNA damage in cancer cells. As per the DNA damage response pathway, DNA damage induced by IR or chemotherapeutic regents activates ATM/ATR/Chk1 signals, followed by the STAT-IRF pathway. In this pathway, STAT1/3-IRF1 appears to play an important role in PD-L1 upregulation after DNA damage. Alternatively, PD-L1 expression is regulated by the neoantigen pathway in the context of DNA damage and repair in cancer cells. Levels of mutation burden are associated with MSI. Mutation burdens and MSI are augmented by the defect of mismatch repair, chromatin remodeling, or abnormal DNA replication. Neoantigens presented by MHC class I, which is recognized by T cell receptors, activate T cells, followed by the release of IFNγ. IFNγ stimulates the STAT-IRF pathway via the IFNγ receptor (IFNGR) and upregulates PD-L1 expression in cancer cells. Another novel pathway, the cGAS/STING pathway, may also be involved in the activation of the IFN-STAT/IRF-PD-L1 pathway. Cytosolic DNA fragments induced by DNA damage activate the cGAS/STING pathway. Activation of the cGAS/STING pathway induces IFN type I (IFNα and IFNβ), which is incorporated into cancer cells via the IFNα/β receptor (IFNAR). IFNα/β-dependent signaling also activates the STAT-IRF pathway, resulting in PD-L1 upregulation.
Figure 3
Figure 3
Repair pathways and signaling in response to DSB induction by IR. After DSB induction, the Ku70/80 heterodimer complex (Ku) rapidly binds to all DSB ends. Ku bound to DSB ends plays the following roles: 1) Ku70/80 complex promotes NHEJ and 2) Ku70/80 complex protects DNA ends from unscheduled digestion by DNA nucleases. In the NHEJ pathway, DSBs are rapidly rejoined by DNA-PKcs and XLF/XRCC4/LIG4 following Ku binding to DSB ends (81). On the other hand, DSB ends are digested in the 5′ to 3′ direction by EXO1 to direct repair pathway toward HR. The resected ssDNA is coated with RPA. BRCA2 promotes the protein switch from RPA to RAD51, facilitating strand invasion into the template strand for recombination-mediated repair. In terms of DNA damage signaling, ATM, which serves as a sensor of DSBs, is the major DNA damage response (DDR) kinase and is activated at unresected DSB ends. At DSB ends during HR, resection promotes a switch from ATM to ATR activation, followed by Chk1 activation. In the context of DNA damage-dependent PD-L1 expression, the activation of Chk1 is a critical step leading to STAT/IRF-mediated PD-L1 upregulation.

References

    1. Ahn GO, Tseng D, Liao CH, Dorie MJ, Czechowicz A, Brown JM. Inhibition of Mac-1 (CD11b/CD18) enhances tumor response to radiation by reducing myeloid cell recruitment. Proc Natl Acad Sci USA. (2010) 107:8363–8. 10.1073/pnas.0911378107
    1. Chiang CS, Fu SY, Wang SC, Yu CF, Chen FH, Lin CM, et al. . Irradiation promotes an m2 macrophage phenotype in tumor hypoxia. Front Oncol. (2012) 2:89. 10.3389/fonc.2012.00089
    1. Crittenden MR, Cottam B, Savage T, Nguyen C, Newell P, Gough MJ. Expression of NF-kappaB p50 in tumor stroma limits the control of tumors by radiation therapy. PLoS ONE. (2012) 7:e39295. 10.1371/journal.pone.0039295
    1. Ahn GO, Brown JM. Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrow-derived myelomonocytic cells. Cancer Cell. (2008) 13:193–205. 10.1016/j.ccr.2007.11.032
    1. Lewis JS, Landers RJ, Underwood JC, Harris AL, Lewis CE. Expression of vascular endothelial growth factor by macrophages is up-regulated in poorly vascularized areas of breast carcinomas. J Pathol. (2000) 192:150–8. 10.1002/1096-9896(2000)9999:9999<::AID-PATH687>;2-G
    1. Hiratsuka S, Watanabe A, Aburatani H, Maru Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol. (2006) 8:1369–75. 10.1038/ncb1507
    1. Shojaei F, Wu X, Zhong C, Yu L, Liang XH, Yao J, et al. . Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature. (2007) 450:825–31. 10.1038/nature06348
    1. Green CE, Liu T, Montel V, Hsiao G, Lester RD, Subramaniam S, et al. . Chemoattractant signaling between tumor cells and macrophages regulates cancer cell migration, metastasis and neovascularization. PLoS ONE. (2009) 4:e6713. 10.1371/journal.pone.0006713
    1. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al. . Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. (2004) 64:5839–49. 10.1158/0008-5472.CAN-04-0465
    1. Noh H, Hu J, Wang X, Xia X, Satelli A, Li S. Immune checkpoint regulator PD-L1 expression on tumor cells by contacting CD11b positive bone marrow derived stromal cells. Cell Commun Signal. (2015) 13:14. 10.1186/s12964-015-0093-y
    1. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. (1998) 101:890–8. 10.1172/JCI1112
    1. Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest. (2002) 109:41–50. 10.1172/JCI0211638
    1. Kachikwu EL, Iwamoto KS, Liao YP, Demarco JJ, Agazaryan N, Economou JS, et al. . Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys. (2011) 81:1128–35. 10.1016/j.ijrobp.2010.09.034
    1. Shevach EM. CD4+ CD25+ suppressor T cells: more questions than answers. Nat Rev Immunol. (2002) 2:389–400. 10.1038/nri821
    1. Chen ML, Pittet MJ, Gorelik L, Flavell RA, Weissleder R, Von Boehmer H, et al. . Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc Natl Acad Sci USA. (2005) 102:419–24. 10.1073/pnas.0408197102
    1. Takeshima T, Pop LM, Laine A, Iyengar P, Vitetta ES, Hannan R. Key role for neutrophils in radiation-induced antitumor immune responses: potentiation with G-CSF. Proc Natl Acad Sci USA. (2016) 113:11300–5. 10.1073/pnas.1613187113
    1. Nam JS, Terabe M, Mamura M, Kang MJ, Chae H, Stuelten C, et al. . An anti-transforming growth factor beta antibody suppresses metastasis via cooperative effects on multiple cell compartments. Cancer Res. (2008) 68:3835–43. 10.1158/0008-5472.CAN-08-0215
    1. Zilio S, Serafini P. Neutrophils and Granulocytic MDSC: the janus god of cancer immunotherapy. Vaccines. (2016) 4:31. 10.3390/vaccines4030031
    1. Elvington M, Scheiber M, Yang X, Lyons K, Jacqmin D, Wadsworth C, et al. . Complement-dependent modulation of antitumor immunity following radiation therapy. Cell Rep. (2014) 8:818–30. 10.1016/j.celrep.2014.06.051
    1. Deng L, Liang H, Burnette B, Weicheslbaum RR, Fu YX. Radiation and anti-PD-L1 antibody combinatorial therapy induces T cell-mediated depletion of myeloid-derived suppressor cells and tumor regression. Oncoimmunology. (2014) 3:e28499. 10.4161/onci.28499
    1. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, Mckenna C, Jones S, Cheadle EJ, et al. . Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. (2014) 74:5458–68. 10.1158/0008-5472.CAN-14-1258
    1. Derer A, Spiljar M, Baumler M, Hecht M, Fietkau R, Frey B, et al. . Chemoradiation increases PD-L1 expression in certain melanoma and glioblastoma cells. Front Immunol. (2016) 7:610. 10.3389/fimmu.2016.00610
    1. Parikh F, Duluc D, Imai N, Clark A, Misiukiewicz K, Bonomi M, et al. . Chemoradiotherapy-induced upregulation of PD-1 antagonizes immunity to HPV-related oropharyngeal cancer. Cancer Res. (2014) 74:7205–16. 10.1158/0008-5472.CAN-14-1913
    1. Bouquet F, Pal A, Pilones KA, Demaria S, Hann B, Akhurst RJ, et al. . TGFbeta1 inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin Cancer Res. (2011) 17:6754–65. 10.1158/1078-0432.CCR-11-0544
    1. Vaupel P, Multhoff G. Accomplices of the hypoxic tumor microenvironment compromising antitumor immunity: adenosine, lactate, acidosis, vascular endothelial growth factor, potassium ions, and phosphatidylserine. Front Immunol. (2017) 8:1887. 10.3389/fimmu.2017.01887
    1. Wennerberg E, Lhuillier C, Vanpouille-Box C, Pilones KA, Garcia-Martinez E, Rudqvist NP, et al. . Barriers to radiation-induced in situ tumor vaccination. Front Immunol. (2017) 8:229. 10.3389/fimmu.2017.00229
    1. Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. (2005) 5:263–74. 10.1038/nrc1586
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. (2007) 25:267–96. 10.1146/annurev.immunol.25.022106.141609
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. (2011) 144:646–74. 10.1016/j.cell.2011.02.013
    1. Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol. (2013) 14:1212–8. 10.1038/ni.2762
    1. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. . Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. (2015) 373:1627–39. 10.1056/NEJMoa1507643
    1. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. . Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. (2015) 373:123–35. 10.1056/NEJMoa1504627
    1. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. . Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. (2015) 372:320–30. 10.1056/NEJMoa1412082
    1. Ferris RL, Blumenschein GJr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. . Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. (2016) 375:1856–67. 10.1056/NEJMoa1602252
    1. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, Von Pawel J, et al. . Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. (2017) 389:255–65. 10.1016/S0140-6736(16)32517-X
    1. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci. (2017) 24:26. 10.1186/s12929-017-0329-9
    1. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. (2018) 62:29–39. 10.1016/j.intimp.2018.06.001
    1. Kataoka K, Ogawa S. Genetic biomarkers for PD-1/PD-L1 blockade therapy. Oncoscience. (2016) 3:311–2. 10.18632/oncoscience.328
    1. Zhang P, Su DM, Liang M, Fu J. Chemopreventive agents induce programmed death-1-ligand 1 (PD-L1) surface expression in breast cancer cells and promote PD-L1-mediated T cell apoptosis. Mol Immunol. (2008) 45:1470–6. 10.1016/j.molimm.2007.08.013
    1. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al. . Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. (2014) 124:687–95. 10.1172/JCI67313
    1. Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, et al. . Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci USA. (2004) 101:17174–9. 10.1073/pnas.0406351101
    1. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. . Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA. (2007) 104:3360–5. 10.1073/pnas.0611533104
    1. Chen MF, Chen PT, Chen WC, Lu MS, Lin PY, Lee KD. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget. (2016) 7:7913–24. 10.18632/oncotarget.6861
    1. Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson V, Dalle S, et al. . Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N Engl J Med. (2018) 378:1789–801. 10.1056/NEJMoa1802357
    1. Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. . Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. (2018) 24:1449–58. 10.1038/s41591-018-0101-z
    1. Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov. (2015) 5:915–9. 10.1158/-15-0563
    1. Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. . Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. (2017) 7:188–201. 10.1158/-16-1223
    1. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. . Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. (2017) 19:1189–201. 10.1016/j.celrep.2017.04.031
    1. Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. . The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. (2015) 5:43–51. 10.1158/-14-0863
    1. Eroglu Z, Zaretsky JM, Hu-Lieskovan S, Kim DW, Algazi A, Johnson DB, et al. . High response rate to PD-1 blockade in desmoplastic melanomas. Nature. (2018) 553:347–50. 10.1038/nature25187
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. (2018) 359:1350–5. 10.1126/science.aar4060
    1. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. . PD-1 Blockade in tumors with mismatch-repair deficiency. N Engl J Med. (2015) 372:2509–20. 10.1056/NEJMoa1500596
    1. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. . Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. (2017) 357:409–13. 10.1126/science.aan6733
    1. Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL, Watkins JC, Rodig S, et al. . Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol. (2015) 1:1319–23. 10.1001/jamaoncol.2015.2151
    1. Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. . ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. (2018) 24:556–62. 10.1038/s41591-018-0012-z
    1. Strickland KC, Howitt BE, Shukla SA, Rodig S, Ritterhouse LL, Liu JF, et al. . Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget. (2016) 7:13587–98. 10.18632/oncotarget.7277
    1. Rothkamm K, Lobrich M. Evidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray doses. Proc Natl Acad Sci USA. (2003) 100:5057–62. 10.1073/pnas.0830918100
    1. Barazzuol L, Rickett N, Ju L, Jeggo PA. Low levels of endogenous or X-ray-induced DNA double-strand breaks activate apoptosis in adult neural stem cells. J Cell Sci. (2015) 128:3597–606. 10.1242/jcs.171223
    1. Sato H, Niimi A, Yasuhara T, Permata TBM, Hagiwara Y, Isono M, et al. . DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat Commun. (2017) 8:1751. 10.1038/s41467-017-01883-9
    1. Vendetti FP, Karukonda P, Clump DA, Teo T, Lalonde R, Nugent K, et al. . ATR kinase inhibitor AZD6738 potentiates CD8+ T cell-dependent antitumor activity following radiation. J Clin Invest. (2018) 128:3926–40. 10.1172/JCI96519
    1. Shibata A, Conrad S, Birraux J, Geuting V, Barton O, Ismail A, et al. . Factors determining DNA double-strand break repair pathway choice in G2 phase. EMBO J. (2011) 30:1079–92. 10.1038/emboj.2011.27
    1. Shibata A, Jeggo P, Lobrich M. The pendulum of the Ku-Ku clock. DNA Repair. (2018) 71:164–71. 10.1016/j.dnarep.2018.08.020
    1. Shibata A, Moiani D, Arvai AS, Perry J, Harding SM, Genois MM, et al. . DNA double-strand break repair pathway choice is directed by distinct MRE11 nuclease activities. Mol Cell. (2014) 53:7–18. 10.1016/j.molcel.2013.11.003
    1. Isono M, Niimi A, Oike T, Hagiwara Y, Sato H, Sekine R, et al. . BRCA1 Directs the repair pathway to homologous recombination by promoting 53BP1 dephosphorylation. Cell Rep. (2017) 18:520–32. 10.1016/j.celrep.2016.12.042
    1. Shibata A, Barton O, Noon AT, Dahm K, Deckbar D, Goodarzi AA, et al. . Role of ATM and the damage response mediator proteins 53BP1 and MDC1 in the maintenance of G(2)/M checkpoint arrest. Mol Cell Biol. (2010) 30:3371–83. 10.1128/MCB.01644-09
    1. Jazayeri A, Falck J, Lukas C, Bartek J, Smith GC, Lukas J, et al. . ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nat Cell Biol. (2006) 8:37–45. 10.1038/ncb1337
    1. Smits VA, Reaper PM, Jackson SP. Rapid PIKK-dependent release of Chk1 from chromatin promotes the DNA-damage checkpoint response. Curr Biol. (2006) 16:150–9. 10.1016/j.cub.2005.11.066
    1. Goto H, Kasahara K, Inagaki M. Novel insights into Chk1 regulation by phosphorylation. Cell Struct Funct. (2015) 40:43–50. 10.1247/csf.14017
    1. Hagiwara Y, Sato H, Permata TBM, Niimi A, Yamauchi M, Oike T, et al. . Analysis of programmed death-ligand 1 expression in primary normal human dermal fibroblasts after DNA damage. Hum Immunol. (2018) 79:627–31. 10.1016/j.humimm.2018.05.008
    1. Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature. (2005) 436:1186–90. 10.1038/nature03884
    1. Nakajima NI, Niimi A, Isono M, Oike T, Sato H, Nakano T, et al. . Inhibition of the HDAC/Suv39/G9a pathway restores the expression of DNA damage-dependent major histocompatibility complex class I-related chain A and B in cancer cells. Oncol Rep. (2017) 38:693–702. 10.3892/or.2017.5773
    1. Ablasser A, Goldeck M, Cavlar T, Deimling T, Witte G, Rohl I, et al. . cGAS produces a 2'-5'-linked cyclic dinucleotide second messenger that activates STING. Nature. (2013) 498:380–4. 10.1038/nature12306
    1. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. (2013) 339:786–91. 10.1126/science.1232458
    1. Wu J, Sun L, Chen X, Du F, Shi H, Chen C, et al. . Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science. (2013) 339:826–30. 10.1126/science.1229963
    1. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, et al. . cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci USA. (2017) 114:1637–42. 10.1073/pnas.1621363114
    1. Parkes EE, Walker SM, Taggart LE, Mccabe N, Knight LA, Wilkinson R, et al. . Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. J Natl Cancer Inst. (2017) 109:199. 10.1093/jnci/djw199
    1. Shibata A, Jeggo PA. DNA double-strand break repair in a cellular context. Clin Oncol. (2014) 26:243–9. 10.1016/j.clon.2014.02.004
    1. Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature. (2017) 548:466–70. 10.1038/nature23470
    1. Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, et al. . cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. (2017) 548:461–5. 10.1038/nature23449
    1. Dunphy G, Flannery SM, Almine JF, Connolly DJ, Paulus C, Jonsson KL, et al. . Non-canonical activation of the DNA sensing adaptor STING by ATM and IFI16 mediates NF-kappaB signaling after nuclear DNA damage. Mol Cell. (2018) 71:745–60 e745. 10.1016/j.molcel.2018.07.034
    1. Shibata A, Jeggo P. A historical reflection on our understanding of radiation-induced DNA double strand break repair in somatic mammalian cells; interfacing the past with the present. Int J Radiat Biol. (2019) 2019:1–36. 10.1080/09553002.2018.1564083
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. . Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. (2007) 13:1050–9. 10.1038/nm1622
    1. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. (2010) 11:373–84. 10.1038/ni.1863
    1. Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, et al. . Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. (2007) 110:296–304. 10.1182/blood-2006-10-051482
    1. Wang K, Wang J, Wei F, Zhao N, Yang F, Ren X. Expression of TLR4 in non-small cell lung cancer is associated with PD-L1 and poor prognosis in patients receiving pulmonectomy. Front Immunol. (2017) 8:456. 10.3389/fimmu.2017.00456
    1. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, et al. . Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. (2013) 86:343–9. 10.1016/j.ijrobp.2012.12.025
    1. Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, et al. . Stereotactic radiation therapy augments antigen-specific PD-1-mediated antitumor immune responses via cross-presentation of tumor antigen. Cancer Immunol Res. (2015) 3:345–55. 10.1158/2326-6066.CIR-14-0196
    1. Park SS, Dong H, Liu X, Harrington SM, Krco CJ, Grams MP, et al. . PD-1 restrains radiotherapy-induced abscopal effect. Cancer Immunol Res. (2015) 3:610–9. 10.1158/2326-6066.CIR-14-0138
    1. Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. . Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. (2015) 520:373–7. 10.1038/nature14292
    1. Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. . Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. (2017) 377:1919–29. 10.1056/NEJMoa1709937
    1. Qin Q, Nan X, Miller T, Fisher R, Teh B, Pandita S, et al. . Complete local and abscopal responses from a combination of radiation and nivolumab in refractory hodgkin's lymphoma. Radiat Res. (2018) 190:322–9. 10.1667/RR15048.1
    1. Roger A, Finet A, Boru B, Beauchet A, Mazeron JJ, Otzmeguine Y, et al. . Efficacy of combined hypo-fractionated radiotherapy and anti-PD-1 monotherapy in difficult-to-treat advanced melanoma patients. Oncoimmunology. (2018) 7:e1442166. 10.1080/2162402X.2018.1442166
    1. Shaverdian N, Lisberg AE, Bornazyan K, Veruttipong D, Goldman JW, Formenti SC, et al. . 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. (2017) 18:895–903. 10.1016/S1470-2045(17)30380-7
    1. Ribeiro Gomes J, Schmerling RA, Haddad CK, Racy DJ, Ferrigno R, Gil E, et al. . Analysis of the abscopal effect with anti-PD1 therapy in patients with metastatic solid tumors. J Immunother. (2016) 39:367–72. 10.1097/CJI.0000000000000141
    1. Demaria S, Formenti SC. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol. (2012) 2:153. 10.3389/fonc.2012.00153
    1. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. (2005) 174:7516–23. 10.4049/jimmunol.174.12.7516
    1. Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. . Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. (2009) 114:589–95. 10.1182/blood-2009-02-206870
    1. Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. (2009) 15:5379–88. 10.1158/1078-0432.CCR-09-0265
    1. Schaue D, Ratikan JA, Iwamoto KS, Mcbride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys. (2012) 83:1306–10. 10.1016/j.ijrobp.2011.09.049
    1. Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. . DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. (2017) 8:15618. 10.1038/ncomms15618
    1. Chajon E, Castelli J, Marsiglia H, De Crevoisier R. The synergistic effect of radiotherapy and immunotherapy: A promising but not simple partnership. Crit Rev Oncol Hematol. (2017) 111:124–32. 10.1016/j.critrevonc.2017.01.017
    1. Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology. (2014) 3:e28518. 10.4161/onci.28518
    1. Perez CA, Fu A, Onishko H, Hallahan DE, Geng L. Radiation induces an antitumour immune response to mouse melanoma. Int J Radiat Biol. (2009) 85:1126–36. 10.3109/09553000903242099
    1. Garg AD, Elsen S, Krysko DV, Vandenabeele P, De Witte P, Agostinis P. Resistance to anticancer vaccination effect is controlled by a cancer cell-autonomous phenotype that disrupts immunogenic phagocytic removal. Oncotarget. (2015) 6:26841–60. 10.18632/oncotarget.4754
    1. Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. (2014) 5:403–16. 10.18632/oncotarget.1719
    1. Andersson U, Erlandsson-Harris H, Yang H, Tracey KJ. HMGB1 as a DNA-binding cytokine. J Leukoc Biol. (2002) 72:1084–91. 10.1189/jlb.72.6.1084
    1. Rovere-Querini P, Capobianco A, Scaffidi P, Valentinis B, Catalanotti F, Giazzon M, et al. . HMGB1 is an endogenous immune adjuvant released by necrotic cells. EMBO Rep. (2004) 5:825–30. 10.1038/sj.embor.7400205
    1. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. . Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. (2007) 13:54–61. 10.1038/nm1523
    1. Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, et al. . Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. (2012) 189:558–66. 10.4049/jimmunol.1200563
    1. Parker JJ, Jones JC, Strober S, Knox SJ. Characterization of direct radiation-induced immune function and molecular signaling changes in an antigen presenting cell line. Clin Immunol. (2013) 148:44–55. 10.1016/j.clim.2013.03.008
    1. Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RR, et al. . The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. (2011) 71:2488–96. 10.1158/0008-5472.CAN-10-2820
    1. Gerber SA, Sedlacek AL, Cron KR, Murphy SP, Frelinger JG, Lord EM. IFN-gamma mediates the antitumor effects of radiation therapy in a murine colon tumor. Am J Pathol. (2013) 182:2345–54. 10.1016/j.ajpath.2013.02.041
    1. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. (2004) 21:137–48. 10.1016/j.immuni.2004.07.017
    1. Ganss R, Ryschich E, Klar E, Arnold B, Hammerling GJ. Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res. (2002) 62:1462–70.
    1. Hallahan D, Kuchibhotla J, Wyble C. Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer Res. (1996) 56:5150–5.
    1. Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, et al. . Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. (2008) 181:3099–107. 10.4049/jimmunol.181.5.3099
    1. Ariyoshi K, Takabatake T, Shinagawa M, Kadono K, Daino K, Imaoka T, et al. . Age dependence of hematopoietic progenitor survival and chemokine family gene induction after gamma irradiation in bone marrow tissue in C3H/He mice. Radiat Res. (2014) 181:302–13. 10.1667/RR13466
    1. Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, et al. . Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. (2006) 203:1259–71. 10.1084/jem.20052494
    1. Sato H, Suzuki Y, Ide M, Katoh T, Noda SE, Ando K, et al. . HLA class I expression and its alteration by preoperative hyperthermo-chemoradiotherapy in patients with rectal cancer. PLoS ONE. (2014) 9:e108122. 10.1371/journal.pone.0108122
    1. Stangl S, Gross C, Pockley AG, Asea AA, Multhoff G. Influence of Hsp70 and HLA-E on the killing of leukemic blasts by cytokine/Hsp70 peptide-activated human natural killer (NK) cells. Cell Stress Chaperones. (2008) 13:221–30. 10.1007/s12192-007-0008-y
    1. Multhoff G, Pockley AG, Schmid TE, Schilling D. The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett. (2015) 368:179–84. 10.1016/j.canlet.2015.02.013
    1. Schilling D, Kuhnel A, Konrad S, Tetzlaff F, Bayer C, Yaglom J, et al. . Sensitizing tumor cells to radiation by targeting the heat shock response. Cancer Lett. (2015) 360:294–301. 10.1016/j.canlet.2015.02.033
    1. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. (2004) 64:4328–37. 10.1158/0008-5472.CAN-04-0073
    1. Gulley JL, Arlen PM, Bastian A, Morin S, Marte J, Beetham P, et al. . Combining a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancer. Clin Cancer Res. (2005) 11:3353–62. 10.1158/1078-0432.CCR-04-2062
    1. Chi KH, Liu SJ, Li CP, Kuo HP, Wang YS, Chao Y, et al. . Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory hepatoma. J Immunother. (2005) 28:129–35. 10.1097/01.cji.0000154248.74383.5e
    1. Finkelstein SE, Iclozan C, Bui MM, Cotter MJ, Ramakrishnan R, Ahmed J, et al. . Combination of external beam radiotherapy (EBRT) with intratumoral injection of dendritic cells as neo-adjuvant treatment of high-risk soft tissue sarcoma patients. Int J Radiat Oncol Biol Phys. (2012) 82:924–32. 10.1016/j.ijrobp.2010.12.068
    1. Finkelstein SE, Rodriguez F, Dunn M, Farmello MJ, Smilee R, Janssen W, et al. . Serial assessment of lymphocytes and apoptosis in the prostate during coordinated intraprostatic dendritic cell injection and radiotherapy. Immunotherapy. (2012) 4:373–82. 10.2217/imt.12.24
    1. Algarra I, Collado A, Garrido F. Protein bound polysaccharide PSK abrogates more efficiently experimental metastases derived from H-2 negative than from H-2 positive fibrosarcoma tumor clones. J Exp Clin Cancer Res. (1997) 16:373–80.
    1. Oka H, Shiraishi Y, Sasaki H, Yoshinaga K, Emori Y, Takei M. Antimetastatic effect of an immunomodulatory arabinomannan extracted from Mycobacterium tuberculosis strain Aoyama B, Z-100, through the production of interleukin-12. Biol Pharm Bull. (2003) 26:1336–41. 10.1248/bpb.26.1336
    1. Kanazawa M, Mori Y, Yoshihara K, Iwadate M, Suzuki S, Endoh Y, et al. . Effect of PSK on the maturation of dendritic cells derived from human peripheral blood monocytes. Immunol Lett. (2004) 91:229–38. 10.1016/j.imlet.2003.12.007
    1. Sadahiro S, Suzuki T, Maeda Y, Tanaka A, Kamijo A, Murayama C, et al. . Effects of preoperative immunochemoradiotherapy and chemoradiotherapy on immune responses in patients with rectal adenocarcinoma. Anticancer Res. (2010) 30:993–9.
    1. Stebbing J, Dalgleish A, Gifford-Moore A, Martin A, Gleeson C, Wilson G, et al. . An intra-patient placebo-controlled phase I trial to evaluate the safety and tolerability of intradermal IMM-101 in melanoma. Ann Oncol. (2012) 23:1314–9. 10.1093/annonc/mdr363
    1. Sugiyama T, Fujiwara K, Ohashi Y, Yokota H, Hatae M, Ohno T, et al. . Phase III placebo-controlled double-blind randomized trial of radiotherapy for stage IIB-IVA cervical cancer with or without immunomodulator Z-100: a JGOG study. Ann Oncol. (2014) 25:1011–7. 10.1093/annonc/mdu057
    1. Levy HB, Lvovsky E, Riley F, Harrington D, Anderson A, Moe J, et al. . Immune modulating effects of poly ICLC. Ann N Y Acad Sci. (1980) 350:33–41. 10.1111/j.1749-6632.1980.tb20604.x
    1. Butowski N, Lamborn KR, Lee BL, Prados MD, Cloughesy T, Deangelis LM, et al. . A North American brain tumor consortium phase II study of poly-ICLC for adult patients with recurrent anaplastic gliomas. J Neurooncol. (2009) 91:183–9. 10.1007/s11060-008-9705-3
    1. Stanley MA. Imiquimod and the imidazoquinolones: mechanism of action and therapeutic potential. Clin Exp Dermatol. (2002) 27:571–7. 10.1046/j.1365-2230.2002.01151.x
    1. Brody JD, Ai WZ, Czerwinski DK, Torchia JA, Levy M, Advani RH, et al. . In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol. (2010) 28:4324–32. 10.1200/JCO.2010.28.9793
    1. Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. . Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. (2004) 58:862–70. 10.1016/j.ijrobp.2003.09.012
    1. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. (1994) 271:907–13. 10.1001/jama.1994.03510360033032
    1. Hallahan DE, Vokes EE, Rubin SJ, O'brien S, Samuels B, Vijaykumar S, et al. . Phase I dose-escalation study of tumor necrosis factor-alpha and concomitant radiation therapy. Cancer J Sci Am. (1995) 1:204–9.
    1. Figlin RA, Thompson JA, Bukowski RM, Vogelzang NJ, Novick AC, Lange P, et al. . Multicenter, randomized, phase III trial of CD8(+) tumor-infiltrating lymphocytes in combination with recombinant interleukin-2 in metastatic renal cell carcinoma. J Clin Oncol. (1999) 17:2521–9. 10.1200/JCO.1999.17.8.2521
    1. Nukui Y, Picozzi VJ, Traverso LW. Interferon-based adjuvant chemoradiation therapy improves survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am J Surg. (2000) 179:367–71. 10.1016/S0002-9610(00)00369-X
    1. Picozzi VJ, Abrams RA, Decker PA, Traverso W, O'reilly EM, Greeno E, et al. . Multicenter phase II trial of adjuvant therapy for resected pancreatic cancer using cisplatin, 5-fluorouracil, and interferon-alfa-2b-based chemoradiation: ACOSOG Trial Z05031. Ann Oncol. (2011) 22:348–54. 10.1093/annonc/mdq384
    1. Schmidt J, Abel U, Debus J, Harig S, Hoffmann K, Herrmann T, et al. . Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma. J Clin Oncol. (2012) 30:4077–83. 10.1200/JCO.2011.38.2960
    1. Le DT, Jaffee EM. Regulatory T-cell modulation using cyclophosphamide in vaccine approaches: a current perspective. Cancer Res. (2012) 72:3439–44. 10.1158/0008-5472.CAN-11-3912
    1. Hassel JC, Jiang H, Bender C, Winkler J, Sevko A, Shevchenko I, et al. . Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe). Oncoimmunology. (2017) 6:e1326440. 10.1080/2162402X.2017.1326440
    1. Califano JA, Khan Z, Noonan KA, Rudraraju L, Zhang Z, Wang H, et al. . Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. (2015) 21:30–8. 10.1158/1078-0432.CCR-14-1716

Source: PubMed

3
購読する