STING: a master regulator in the cancer-immunity cycle

Yuanyuan Zhu, Xiang An, Xiao Zhang, Yu Qiao, Tongsen Zheng, Xiaobo Li, Yuanyuan Zhu, Xiang An, Xiao Zhang, Yu Qiao, Tongsen Zheng, Xiaobo Li

Abstract

The aberrant appearance of DNA in the cytoplasm triggers the activation of cGAS-cGAMP-STING signaling and induces the production of type I interferons, which play critical roles in activating both innate and adaptive immune responses. Recently, numerous studies have shown that the activation of STING and the stimulation of type I IFN production are critical for the anticancer immune response. However, emerging evidence suggests that STING also regulates anticancer immunity in a type I IFN-independent manner. For instance, STING has been shown to induce cell death and facilitate the release of cancer cell antigens. Moreover, STING activation has been demonstrated to enhance cancer antigen presentation, contribute to the priming and activation of T cells, facilitate the trafficking and infiltration of T cells into tumors and promote the recognition and killing of cancer cells by T cells. In this review, we focus on STING and the cancer immune response, with particular attention to the roles of STING activation in the cancer-immunity cycle. Additionally, the negative effects of STING activation on the cancer immune response and non-immune roles of STING in cancer have also been discussed.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
DNA-driven cGAS-cGAMP-STING signaling mediates innate immune response. The left cell exhibits the main components of cGAS-cGAMP-STING signaling pathway and IFN signaling pathway, and the right cell shows that IFN could activate neighbor cells in a paracrine manner and cGAMP could be transferred to neighbor cells through GAP junction
Fig. 2
Fig. 2
Activation of STING positively regulates each step of cancer-immunity cycle
Fig. 3
Fig. 3
The positive and negative roles of STING activation in antitumor immune response. On the one hand, STING facilitates antitumor immune response through promoting the infiltration of effector cells and eradication of tumor cells. On the other hand, constant STING activation may hamper immune response by inducing the infiltration of immune suppressive cells, such as Treg and MDSC, and upregulating the expression of PD-L1 on tumor cells and PD-1 on T cells. Moreover, STING activation is associated with the enhanced activity of IDO, an enzyme catalyzing the transformation of tryptophan into kynurenine. Diminished tryptophan restricts the proliferation of T cells whereas elevated kynurenine promotes differentiation of Tregs but hampers antigen presenting ability of DCs. Additionally, aberrant STING activation also directly inhibits T cell proliferation and even promotes apoptosis of lymphocytes

References

    1. Kienle GS. Fever in Cancer treatment: Coley’s therapy and epidemiologic observations. Glob Adv Health Med. 2012;1:92–100. doi: 10.7453/gahmj.2012.1.1.016.
    1. McCarthy EF. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop J. 2006;26:154–158.
    1. Medzhitov R, Janeway C., Jr Innate immunity. N Engl J Med. 2000;343:338–344. doi: 10.1056/NEJM200008033430506.
    1. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022.
    1. Wang X, Smith C, Yin H. Targeting toll-like receptors with small molecule agents. Chem Soc Rev. 2013;42:4859–4866. doi: 10.1039/c3cs60039d.
    1. Motta V, Soares F, Sun T, Philpott DJ. NOD-like receptors: versatile cytosolic sentinels. Physiol Rev. 2015;95:149–178. doi: 10.1152/physrev.00009.2014.
    1. Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol. 2015;33:257–290. doi: 10.1146/annurev-immunol-032414-112240.
    1. O'Neill LA. DNA makes RNA makes innate immunity. Cell. 2009;138:428–430. doi: 10.1016/j.cell.2009.07.021.
    1. Ablasser A, Hertrich C, Wassermann R, Hornung V. Nucleic acid driven sterile inflammation. Clin Immunol. 2013;147:207–215. doi: 10.1016/j.clim.2013.01.003.
    1. Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674. doi: 10.1038/nature07317.
    1. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461:788–792. doi: 10.1038/nature08476.
    1. Sun W, Li Y, Chen L, Chen H, You F, Zhou X, Zhou Y, Zhai Z, Chen D, Jiang Z. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proc Natl Acad Sci U S A. 2009;106:8653–8658. doi: 10.1073/pnas.0900850106.
    1. Zhong B, Yang Y, Li S, Wang YY, Li Y, Diao F, Lei C, He X, Zhang L, Tien P, Shu HB. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity. 2008;29:538–550. doi: 10.1016/j.immuni.2008.09.003.
    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;339:786–791. doi: 10.1126/science.1232458.
    1. Wu J, Sun L, Chen X, Du F, Shi H, Chen C, Chen ZJ. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science. 2013;339:826–830. doi: 10.1126/science.1229963.
    1. Jin L, Waterman PM, Jonscher KR, Short CM, Reisdorph NA, Cambier JC. MPYS, a novel membrane tetraspanner, is associated with major histocompatibility complex class II and mediates transduction of apoptotic signals. Mol Cell Biol. 2008;28:5014–5026. doi: 10.1128/MCB.00640-08.
    1. Tao J, Zhou X, Jiang Z. cGAS-cGAMP-STING: the three musketeers of cytosolic DNA sensing and signaling. IUBMB Life. 2016;68:858–870. doi: 10.1002/iub.1566.
    1. Ablasser A, Schmid-Burgk JL, Hemmerling I, Horvath GL, Schmidt T, Latz E, Hornung V. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature. 2013;503:530–534. doi: 10.1038/nature12640.
    1. Bridgeman A, Maelfait J, Davenne T, Partridge T, Peng Y, Mayer A, Dong T, Kaever V, Borrow P, Rehwinkel J. Viruses transfer the antiviral second messenger cGAMP between cells. Science. 2015;349:1228–1232. doi: 10.1126/science.aab3632.
    1. Gentili M, Kowal J, Tkach M, Satoh T, Lahaye X, Conrad C, et al. Transmission of innate immune signaling by packaging of cGAMP in viral particles. Science. 2015;349:1232–1236. doi: 10.1126/science.aab3628.
    1. Corrales L, Matson V, Flood B, Spranger S, Gajewski TF. Innate immune signaling and regulation in cancer immunotherapy. Cell Res. 2017;27:96–108. doi: 10.1038/cr.2016.149.
    1. Galluzzi L, Vanpouille-Box C, Bakhoum SF, Demaria S. SnapShot: CGAS-STING Signaling. Cell. 2018;173:276. doi: 10.1016/j.cell.2018.03.015.
    1. Piehler J, Thomas C, Garcia KC, Schreiber G. Structural and dynamic determinants of type I interferon receptor assembly and their functional interpretation. Immunol Rev. 2012;250:317–334. doi: 10.1111/imr.12001.
    1. Dunn GP, Bruce AT, Sheehan KC, Shankaran V, Uppaluri R, Bui JD, Diamond MS, Koebel CM, Arthur C, White JM, Schreiber RD. A critical function for type I interferons in cancer immunoediting. Nat Immunol. 2005;6:722–729. doi: 10.1038/ni1213.
    1. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, Gajewski TF. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J Exp Med. 2011;208:2005–2016. doi: 10.1084/jem.20101159.
    1. Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208:1989–2003. doi: 10.1084/jem.20101158.
    1. Gresser I, Bandu MT, Brouty-Boye D. Interferon and cell division. IX. Interferon-resistant L1210 cells: characteristics and origin. J Natl Cancer Inst. 1974;52:553–559. doi: 10.1093/jnci/52.2.553.
    1. Gresser I, Bourali C. Antitumor effects of interferon preparations in mice. J Natl Cancer Inst. 1970;45:365–376.
    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;19:1189–1201. doi: 10.1016/j.celrep.2017.04.031.
    1. Morimoto Y, Kishida T, Kotani SI, Takayama K, Mazda O. Interferon-beta signal may up-regulate PD-L1 expression through IRF9-dependent and independent pathways in lung cancer cells. Biochem Biophys Res Commun. 2018;507:330–336. doi: 10.1016/j.bbrc.2018.11.035.
    1. Weichselbaum RR, Ishwaran H, Yoon T, Nuyten DS, Baker SW, Khodarev N, et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc Natl Acad Sci U S A. 2008;105:18490–18495. doi: 10.1073/pnas.0809242105.
    1. Erdal E, Haider S, Rehwinkel J, Harris AL, McHugh PJ. A prosurvival DNA damage-induced cytoplasmic interferon response is mediated by end resection factors and is limited by Trex1. Genes Dev. 2017;31:353–369. doi: 10.1101/gad.289769.116.
    1. Walsh Scott R., Bastin Donald, Chen Lan, Nguyen Andrew, Storbeck Christopher J., Lefebvre Charles, Stojdl David, Bramson Jonathan L., Bell John C., Wan Yonghong. Type I IFN blockade uncouples immunotherapy-induced antitumor immunity and autoimmune toxicity. Journal of Clinical Investigation. 2018;129(2):518–530. doi: 10.1172/JCI121004.
    1. Woo SR, Corrales L, Gajewski TF. The STING pathway and the T cell-inflamed tumor microenvironment. Trends Immunol. 2015;36:250–256. doi: 10.1016/j.it.2015.02.003.
    1. Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MY, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–842. doi: 10.1016/j.immuni.2014.10.017.
    1. Demaria O, De Gassart A, Coso S, Gestermann N, Di Domizio J, Flatz L, et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci U S A. 2015;112:15408–15413. doi: 10.1073/pnas.1512832112.
    1. Ohkuri T, Ghosh A, Kosaka A, Zhu J, Ikeura M, David M, Watkins SC, Sarkar SN, Okada H. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol Res. 2014;2:1199–1208. doi: 10.1158/2326-6066.CIR-14-0099.
    1. Ching LM, Cao Z, Kieda C, Zwain S, Jameson MB, Baguley BC. Induction of endothelial cell apoptosis by the antivascular agent 5,6-Dimethylxanthenone-4-acetic acid. Br J Cancer. 2002;86:1937–1942. doi: 10.1038/sj.bjc.6600368.
    1. Jassar AS, Suzuki E, Kapoor V, Sun J, Silverberg MB, Cheung L, Burdick MD, Strieter RM, Ching LM, Kaiser LR, Albelda SM. Activation of tumor-associated macrophages by the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid induces an effective CD8+ T-cell-mediated antitumor immune response in murine models of lung cancer and mesothelioma. Cancer Res. 2005;65:11752–11761. doi: 10.1158/0008-5472.CAN-05-1658.
    1. Roberts ZJ, Ching LM, Vogel SN. IFN-beta-dependent inhibition of tumor growth by the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) J Interf Cytokine Res. 2008;28:133–139. doi: 10.1089/jir.2007.0992.
    1. Prantner D, Perkins DJ, Lai W, Williams MS, Sharma S, Fitzgerald KA, Vogel SN. 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) activates stimulator of interferon gene (STING)-dependent innate immune pathways and is regulated by mitochondrial membrane potential. J Biol Chem. 2012;287:39776–39788. doi: 10.1074/jbc.M112.382986.
    1. Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J Immunol. 2013;190:5216–5225. doi: 10.4049/jimmunol.1300097.
    1. Lara PN, Jr, Douillard JY, Nakagawa K, von Pawel J, McKeage MJ, Albert I, et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J Clin Oncol. 2011;29:2965–2971. doi: 10.1200/JCO.2011.35.0660.
    1. Curran E, Chen X, Corrales L, Kline DE, Dubensky TW, Jr, Duttagupta P, Kortylewski M, Kline J. STING pathway activation stimulates potent immunity against acute myeloid leukemia. Cell Rep. 2016;15:2357–2366. doi: 10.1016/j.celrep.2016.05.023.
    1. Kitai Y, Kawasaki T, Sueyoshi T, Kobiyama K, Ishii KJ, Zou J, Akira S, Matsuda T, Kawai T. DNA-containing Exosomes derived from Cancer cells treated with Topotecan activate a STING-dependent pathway and reinforce antitumor immunity. J Immunol. 2017;198:1649–1659. doi: 10.4049/jimmunol.1601694.
    1. Pantelidou C, Sonzogni O, De Oliveria TM, Mehta AK, Kothari A, Wang D, et al. PARP inhibitor efficacy depends on CD8(+) T-cell recruitment via Intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast Cancer. Cancer Discov. 2019;9:722–737. doi: 10.1158/-18-1218.
    1. Chandra D, Quispe-Tintaya W, Jahangir A, Asafu-Adjei D, Ramos I, Sintim HO, Zhou J, Hayakawa Y, Karaolis DK, Gravekamp C. STING ligand c-di-GMP improves cancer vaccination against metastatic breast cancer. Cancer Immunol Res. 2014;2:901–910. doi: 10.1158/2326-6066.CIR-13-0123.
    1. Gaston J, Cheradame L, Yvonnet V, Deas O, Poupon MF, Judde JG, Cairo S, Goffin V. Intracellular STING inactivation sensitizes breast cancer cells to genotoxic agents. Oncotarget. 2016;7:77205–77224. doi: 10.18632/oncotarget.12858.
    1. Chen J, Markelc B, Kaeppler J, Ogundipe VML, Cao Y, McKenna WG, Muschel RJ. STING-dependent interferon-lambda1 induction in HT29 cells, a human colorectal Cancer cell line, after gamma-radiation. Int J Radiat Oncol Biol Phys. 2018;101:97–106. doi: 10.1016/j.ijrobp.2018.01.091.
    1. Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41:843–852. doi: 10.1016/j.immuni.2014.10.019.
    1. Liang H, Deng L, Hou Y, Meng X, Huang X, Rao E, et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat Commun. 2017;8:1736. doi: 10.1038/s41467-017-01566-5.
    1. Baird JR, Bell RB, Troesch V, Friedman D, Bambina S, Kramer G, et al. Evaluation of explant responses to STING ligands: personalized Immunosurgical therapy for head and neck squamous cell carcinoma. Cancer Res. 2018;78:6308–6319. doi: 10.1158/0008-5472.CAN-18-1652.
    1. Lu S, Concha-Benavente F, Shayan G, Srivastava RM, Gibson SP, Wang L, Gooding WE, Ferris RL. STING activation enhances cetuximab-mediated NK cell activation and DC maturation and correlates with HPV(+) status in head and neck cancer. Oral Oncol. 2018;78:186–193. doi: 10.1016/j.oraloncology.2018.01.019.
    1. Gadkaree SK, Fu J, Sen R, Korrer MJ, Allen C, Kim YJ. Induction of tumor regression by intratumoral STING agonists combined with anti-programmed death-L1 blocking antibody in a preclinical squamous cell carcinoma model. Head Neck. 2017;39:1086–1094. doi: 10.1002/hed.24704.
    1. Moore E, Clavijo PE, Davis R, Cash H, Van Waes C, Kim Y, Allen C. Established T cell-inflamed tumors rejected after adaptive resistance was reversed by combination STING activation and PD-1 pathway blockade. Cancer Immunol Res. 2016;4:1061–1071. doi: 10.1158/2326-6066.CIR-16-0104.
    1. Chabanon RM, Muirhead G, Krastev DB, Adam J, Morel D, Garrido M, et al. PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non-small cell lung cancer. J Clin Invest. 2019;129:1211–1228. doi: 10.1172/JCI123319.
    1. Sen T, Rodriguez BL, Chen L, Corte CMD, Morikawa N, Fujimoto J, et al. Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung Cancer. Cancer Discov. 2019;9:646–661. doi: 10.1158/-18-1020.
    1. Downey CM, Aghaei M, Schwendener RA, Jirik FR. DMXAA causes tumor site-specific vascular disruption in murine non-small cell lung cancer, and like the endogenous non-canonical cyclic dinucleotide STING agonist, 2′3’-cGAMP, induces M2 macrophage repolarization. PLoS One. 2014;9:e99988. doi: 10.1371/journal.pone.0099988.
    1. Yang H, Lee WS, Kong SJ, Kim CG, Kim JH, Chang SK, Kim S, Kim G, Chon HJ, Kim C. STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade. J Clin Invest. 2019;130:4350–4364. doi: 10.1172/JCI125413.
    1. Tang CH, Zundell JA, Ranatunga S, Lin C, Nefedova Y, Del Valle JR, Hu CC. Agonist-mediated activation of STING induces apoptosis in malignant B cells. Cancer Res. 2016;76:2137–2152. doi: 10.1158/0008-5472.CAN-15-1885.
    1. Marcus A, Mao AJ, Lensink-Vasan M, Wang L, Vance RE, Raulet DH. Tumor-Derived cGAMP Triggers a STING-Mediated Interferon Response in Non-tumor Cells to Activate the NK Cell Response. Immunity. 2018;49:754–63.e4. doi: 10.1016/j.immuni.2018.09.016.
    1. Zhang CX, Ye SB, Ni JJ, Cai TT, Liu YN, Huang DJ, et al. STING signaling remodels the tumor microenvironment by antagonizing myeloid-derived suppressor cell expansion. Cell Death Differ. 2019;26(11):2314–2328. doi: 10.1038/s41418-019-0302-0.
    1. Ghaffari A, Peterson N, Khalaj K, Vitkin N, Robinson A, Francis JA, Koti M. STING agonist therapy in combination with PD-1 immune checkpoint blockade enhances response to carboplatin chemotherapy in high-grade serous ovarian cancer. Br J Cancer. 2018;119:440–449. doi: 10.1038/s41416-018-0188-5.
    1. Jing W, McAllister D, Vonderhaar EP, Palen K, Riese MJ, Gershan J, Johnson BD, Dwinell MB. STING agonist inflames the pancreatic cancer immune microenvironment and reduces tumor burden in mouse models. J Immunother Cancer. 2019;7:115. doi: 10.1186/s40425-019-0573-5.
    1. Ho SS, Zhang WY, Tan NY, Khatoo M, Suter MA, Tripathi S, Cheung FS, Lim WK, Tan PH, Ngeow J, Gasser S. The DNA structure-specific endonuclease MUS81 mediates DNA sensor STING-dependent host rejection of prostate Cancer cells. Immunity. 2016;44:1177–1189. doi: 10.1016/j.immuni.2016.04.010.
    1. Ager CR, Reilley MJ, Nicholas C, Bartkowiak T, Jaiswal AR, Curran MA. Intratumoral STING activation with T-cell checkpoint modulation generates systemic antitumor immunity. Cancer Immunol Res. 2017;5:676–684. doi: 10.1158/2326-6066.CIR-17-0049.
    1. Liang D, Xiao-Feng H, Guan-Jun D, Er-Ling H, Sheng C, Ting-Ting W, Qin-Gang H, Yan-Hong N, Ya-Yi H. Activated STING enhances Tregs infiltration in the HPV-related carcinogenesis of tongue squamous cells via the c-Jun/CCL22 signal. Biochim Biophys Acta. 1852;2015:2494–2503.
    1. Koshy Sandeep T., Cheung Alexander S., Gu Luo, Graveline Amanda R., Mooney David J. Liposomal Delivery Enhances Immune Activation by STING Agonists for Cancer Immunotherapy. Advanced Biosystems. 2017;1(1-2):1600013. doi: 10.1002/adbi.201600013.
    1. Konate Karidia, Dussot Marion, Aldrian Gudrun, Vaissière Anaïs, Viguier Véronique, Neira Isabel Ferreiro, Couillaud Franck, Vivès Eric, Boisguerin Prisca, Deshayes Sébastien. Peptide-Based Nanoparticles to Rapidly and Efficiently “Wrap ’n Roll” siRNA into Cells. Bioconjugate Chemistry. 2018;30(3):592–603. doi: 10.1021/acs.bioconjchem.8b00776.
    1. Berger G, Lawler SE. Novel non-nucleotidic STING agonists for cancer immunotherapy. Future Med Chem. 2018;10:2767–2769. doi: 10.4155/fmc-2018-0367.
    1. Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang SY, et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564:439–443. doi: 10.1038/s41586-018-0705-y.
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10. doi: 10.1016/j.immuni.2013.07.012.
    1. Tsioulias GJ, Triadafilopoulos G, Goldin E, Papavassiliou ED, Rizos S, Bassioukas P, Rigas B. Expression of HLA class I antigens in sporadic adenomas and histologically Normal mucosa of the Colon. Cancer Res. 1993;53:2374–2378.
    1. Sze A, Belgnaoui SM, Olagnier D, Lin R, Hiscott J, van Grevenynghe J. Host restriction factor SAMHD1 limits human T cell leukemia virus type 1 infection of monocytes via STING-mediated apoptosis. Cell Host Microbe. 2013;14:422–434. doi: 10.1016/j.chom.2013.09.009.
    1. Setterblad N, Blancheteau V, Delaguillaumie A, Michel F, Becart S, Lombardi G, Acuto O, Charron D, Mooney N. Cognate MHC-TCR interaction leads to apoptosis of antigen-presenting cells. J Leukoc Biol. 2004;75:1036–1044. doi: 10.1189/jlb.0703356.
    1. Nagy ZA, Hubner B, Lohning C, Rauchenberger R, Reiffert S, Thomassen-Wolf E, et al. Fully human, HLA-DR-specific monoclonal antibodies efficiently induce programmed death of malignant lymphoid cells. Nat Med. 2002;8:801–807. doi: 10.1038/nm736.
    1. Pisapia L, Barba P, Cortese A, Cicatiello V, Morelli F, Del Pozzo G. EBP1 protein modulates the expression of human MHC class II molecules in non-hematopoietic cancer cells. Int J Oncol. 2015;47:481–489. doi: 10.3892/ijo.2015.3051.
    1. Park IA, Hwang SH, Song IH, Heo SH, Kim YA, Bang WS, Park HS, Lee M, Gong G, Lee HJ. Expression of the MHC class II in triple-negative breast cancer is associated with tumor-infiltrating lymphocytes and interferon signaling. PLoS One. 2017;12:e0182786. doi: 10.1371/journal.pone.0182786.
    1. He Y, Rozeboom L, Rivard CJ, Ellison K, Dziadziuszko R, Yu H, Zhou C, Hirsch FR. MHC class II expression in lung cancer. Lung Cancer. 2017;112:75–80. doi: 10.1016/j.lungcan.2017.07.030.
    1. Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–1577. doi: 10.1126/science.1208347.
    1. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008;8:279–289. doi: 10.1038/nri2215.
    1. Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell. 2010;140:798–804. doi: 10.1016/j.cell.2010.02.015.
    1. Luft T, Pang KC, Thomas E, Hertzog P, Hart DN, Trapani J, Cebon J. Type I IFNs enhance the terminal differentiation of dendritic cells. J Immunol. 1998;161:1947–1953.
    1. Paquette RL, Hsu NC, Kiertscher SM, Park AN, Tran L, Roth MD, Glaspy JA. Interferon-alpha and granulocyte-macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells. J Leukoc Biol. 1998;64:358–367. doi: 10.1002/jlb.64.3.358.
    1. Radvanyi LG, Banerjee A, Weir M, Messner H. Low levels of interferon-alpha induce CD86 (B7.2) expression and accelerates dendritic cell maturation from human peripheral blood mononuclear cells. Scand J Immunol. 1999;50:499–509. doi: 10.1046/j.1365-3083.1999.00625.x.
    1. Skrnjug I, Guzman CA, Rueckert C. Cyclic GMP-AMP displays mucosal adjuvant activity in mice. PLoS One. 2014;9:e110150. doi: 10.1371/journal.pone.0110150.
    1. Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, Chen ZJ. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci U S A. 2017;114:1637–1642. doi: 10.1073/pnas.1621363114.
    1. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–770. doi: 10.1038/nri3921.
    1. Bui JD, Schreiber RD. Cancer immunosurveillance, immunoediting and inflammation: independent or interdependent processes? Curr Opin Immunol. 2007;19:203–208. doi: 10.1016/j.coi.2007.02.001.
    1. Fu J, Kanne DB, Leong M, Glickman LH, McWhirter SM, Lemmens E, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7:283ra52. doi: 10.1126/scitranslmed.aaa4306.
    1. Cerboni S, Jeremiah N, Gentili M, Gehrmann U, Conrad C, Stolzenberg MC, et al. Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes. J Exp Med. 2017;214:1769–1785. doi: 10.1084/jem.20161674.
    1. Larkin B, Ilyukha V, Sorokin M, Buzdin A, Vannier E, Poltorak A. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J Immunol. 2017;199:397–402. doi: 10.4049/jimmunol.1601999.
    1. Zlotnik A, Yoshie O. The chemokine superfamily revisited. Immunity. 2012;36:705–716. doi: 10.1016/j.immuni.2012.05.008.
    1. Viola A, Sarukhan A, Bronte V, Molon B. The pros and cons of chemokines in tumor immunology. Trends Immunol. 2012;33:496–504. doi: 10.1016/j.it.2012.05.007.
    1. Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69:3077–3085. doi: 10.1158/0008-5472.CAN-08-2281.
    1. Padovan E, Spagnoli GC, Ferrantini M, Heberer M. IFN-alpha2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8+ effector T cells. J Leukoc Biol. 2002;71:669–676.
    1. Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015;11:1018–1030. doi: 10.1016/j.celrep.2015.04.031.
    1. Ohkuri T, Kosaka A, Ishibashi K, Kumai T, Hirata Y, Ohara K, et al. Intratumoral administration of cGAMP transiently accumulates potent macrophages for anti-tumor immunity at a mouse tumor site. Cancer Immunol Immunother. 2017;66:705–716. doi: 10.1007/s00262-017-1975-1.
    1. Motz GT, Coukos G. Deciphering and reversing tumor immune suppression. Immunity. 2013;39:61–73. doi: 10.1016/j.immuni.2013.07.005.
    1. Shimizu K, Iyoda T, Okada M, Yamasaki S, Fujii SI. Immune suppression and reversal of the suppressive tumor microenvironment. Int Immunol. 2018;30:445–454. doi: 10.1093/intimm/dxy042.
    1. Manning EA, Ullman JG, Leatherman JM, Asquith JM, Hansen TR, Armstrong TD, Hicklin DJ, Jaffee EM, Emens LA. A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism. Clin Cancer Res. 2007;13:3951–3959. doi: 10.1158/1078-0432.CCR-07-0374.
    1. Ebos JM, Kerbel RS. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol. 2011;8:210–221. doi: 10.1038/nrclinonc.2011.21.
    1. Ebos JM, Lee CR, Kerbel RS. Tumor and host-mediated pathways of resistance and disease progression in response to antiangiogenic therapy. Clin Cancer Res. 2009;15:5020–5025. doi: 10.1158/1078-0432.CCR-09-0095.
    1. Murphy KP, Travers P, Walport M, Janeway C. Janeway’s immunobiology. New York: Garland science; 2014.
    1. Propper DJ, Chao D, Braybrooke JP, Bahl P, Thavasu P, Balkwill F, et al. Low-dose IFN-gamma induces tumor MHC expression in metastatic malignant melanoma. Clin Cancer Res. 2003;9:84–92.
    1. Lirussi D, Ebensen T, Schulze K, Trittel S, Duran V, Liebich I, Kalinke U, Guzman CA. Type I IFN and not TNF, is Essential for Cyclic Di-nucleotide-elicited CTL by a Cytosolic Cross-presentation Pathway. EBioMedicine. 2017;22:100–111. doi: 10.1016/j.ebiom.2017.07.016.
    1. An X, Zhu Y, Zheng T, Wang G, Zhang M, Li J, et al. An analysis of the expression and association with immune cell infiltration of the cGAS/STING pathway in pan-Cancer. Mol Ther Nucleic Acids. 2018;14:80–89. doi: 10.1016/j.omtn.2018.11.003.
    1. Lemos H, Mohamed E, Huang L, Ou R, Pacholczyk G, Arbab AS, Munn D, Mellor AL. STING promotes the growth of tumors characterized by low antigenicity via IDO activation. Cancer Res. 2016;76:2076–2081. doi: 10.1158/0008-5472.CAN-15-1456.
    1. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, Brown C, Mellor AL. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281:1191–1193. doi: 10.1126/science.281.5380.1191.
    1. Wek RC, Jiang HY, Anthony TG. Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans. 2006;34:7–11. doi: 10.1042/BST0340007.
    1. Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, Mellor AL. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005;22:633–642. doi: 10.1016/j.immuni.2005.03.013.
    1. Sundrud MS, Koralov SB, Feuerer M, Calado DP, Kozhaya AE, Rhule-Smith A, et al. Halofuginone inhibits TH17 cell differentiation by activating the amino acid starvation response. Science. 2009;324:1334–1338. doi: 10.1126/science.1172638.
    1. Palm CA, Smukler SM, Sullivan CC, Mutuo PK, Nyadzi GI, Walsh MG. Identifying potential synergies and trade-offs for meeting food security and climate change objectives in sub-Saharan Africa. Proc Natl Acad Sci U S A. 2010;107:19661–19666. doi: 10.1073/pnas.0912248107.
    1. Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A. 2010;107:19961–19966. doi: 10.1073/pnas.1014465107.
    1. Munn DH, Mellor AL. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol. 2016;37:193–207. doi: 10.1016/j.it.2016.01.002.
    1. Huang L, Lemos HP, Li L, Li M, Chandler PR, Baban B, McGaha TL, Ravishankar B, Lee JR, Munn DH, Mellor AL. Engineering DNA nanoparticles as immunomodulatory reagents that activate regulatory T cells. J Immunol. 2012;188:4913–4920. doi: 10.4049/jimmunol.1103668.
    1. Huang L, Li L, Lemos H, Chandler PR, Pacholczyk G, Baban B, et al. Cutting edge: DNA sensing via the STING adaptor in myeloid dendritic cells induces potent tolerogenic responses. J Immunol. 2013;191:3509–3513. doi: 10.4049/jimmunol.1301419.
    1. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219–242. doi: 10.1111/j.1600-065X.2010.00923.x.
    1. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, Thompson CB, Bluestone JA. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1:405–413. doi: 10.1016/1074-7613(94)90071-X.
    1. Petrasek J, Iracheta-Vellve A, Csak T, Satishchandran A, Kodys K, Kurt-Jones EA, Fitzgerald KA, Szabo G. STING-IRF3 pathway links endoplasmic reticulum stress with hepatocyte apoptosis in early alcoholic liver disease. Proc Natl Acad Sci U S A. 2013;110:16544–16549. doi: 10.1073/pnas.1308331110.
    1. Moretti J, Roy S, Bozec D, Martinez J, Chapman JR, Ueberheide B, et al. STING Senses Microbial Viability to Orchestrate Stress-Mediated Autophagy of the Endoplasmic Reticulum. Cell. 2017;171:809–23.e13. doi: 10.1016/j.cell.2017.09.034.
    1. Liu Y, Gordesky-Gold B, Leney-Greene M, Weinbren NL, Tudor M, Cherry S. Inflammation-Induced, STING-Dependent Autophagy Restricts Zika Virus Infection in the Drosophila Brain. Cell Host Microbe. 2018;24:57–68.e3. doi: 10.1016/j.chom.2018.05.022.
    1. Liu Dong, Wu Hao, Wang Chenguang, Li Yanjun, Tian Huabin, Siraj Sami, Sehgal Sheikh Arslan, Wang Xiaohui, Wang Jun, Shang Yingli, Jiang Zhengfan, Liu Lei, Chen Quan. STING directly activates autophagy to tune the innate immune response. Cell Death & Differentiation. 2018;26(9):1735–1749. doi: 10.1038/s41418-018-0251-z.
    1. Gui X, Yang H, Li T, Tan X, Shi P, Li M, Du F, Chen ZJ. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature. 2019;567:262–266. doi: 10.1038/s41586-019-1006-9.
    1. Ranoa Diana Rose E., Widau Ryan C., Mallon Stephen, Parekh Akash D., Nicolae Claudia M., Huang Xiaona, Bolt Michael J., Arina Ainhoa, Parry Renate, Kron Stephen J., Moldovan George-Lucian, Khodarev Nikolai N., Weichselbaum Ralph R. STING Promotes Homeostasis via Regulation of Cell Proliferation and Chromosomal Stability. Cancer Research. 2018;79(7):1465–1479. doi: 10.1158/0008-5472.CAN-18-1972.
    1. Daei Farshchi Adli A, Jahanban-Esfahlan R, Seidi K, Samandari-Rad S, Zarghami N. An overview on Vadimezan (DMXAA): The vascular disrupting agent. Chem Biol Drug Des. 2018;91:996–1006. doi: 10.1111/cbdd.13166.
    1. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, et al. Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533:493–498. doi: 10.1038/nature18268.
    1. Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature. 2018;553:467–472. doi: 10.1038/nature25432.

Source: PubMed

3
Abonnere