Skip to main content

Role of the cGAS-STING pathway in radiotherapy for non-small cell lung cancer

Abstract

One of the most important therapeutic interventions for non-small cell lung cancer is radiotherapy. Ionizing radiation (IR) is classified by traditional radiobiology principles as a direct cytocidal therapeutic agent against cancer, although there is growing recognition of other antitumor immunological responses induced by this modality. The most effective therapeutic combinations to harness radiation-generated antitumor immunity and enhance treatment results for malignancies resistant to existing radiotherapy regimens could be determined by a more sophisticated understanding of the immunological pathways created by radiation. Innate immune signaling is triggered by the activation of cGAS-STING, and this promotes adaptive immune responses to help fight cancer. This identifies a molecular mechanism radiation can use to trigger antitumor immune responses by bridging the DNA-damaging ability of IR with the activation of CD8 + cytotoxic T cell-mediated killing of tumors. We also discuss radiotherapy-related parameters that affect cGAS-STING signaling, negative consequences of cGAS-STING activation, and intriguing treatment options being tested in conjunction with IR to support immune activation by activating STING-signaling. Improved therapeutic outcomes will result from a better understanding of how IR promotes cGAS-STING signaling in immune-based treatment regimens that maximize radiotherapy’s anticancer effectiveness.

Introduction

Lung cancer is one of the most common cancers and the leading cause of cancer-related deaths worldwide [1]. Among the common lung cancer types 85% of clinical cases are non-small cell lung cancer (NSCLC) [2, 3],which has a low overall cure rate and survival rate [4, 5]. Radiotherapy (RT), as a traditional treatment, is applied to more than 50% of patients with malignant tumors, and its principle of action is to directly cause fatal DNA damage in irradiated cells, or indirectly induce DNA damage by producing reactive oxygen species [6]. RT induces DNA damage, resulting in the formation of micronuclei containing chromatin and dsDNA in the cytosol, and the nuclear envelope of micronuclei is easily ruptured due to the lack of a stable nuclear lamina, exposing dsDNA to the cytoplasm and becoming the trigger point of the innate immune response, exerting anti-tumor effects [7]. Radiation therapy plays an important role in the treatment of non-small cell lung cancer, and inevitable lung tissue will be irradiated at a certain dose during the treatment, causing different degrees of radiation-induced lung injury, with an incidence of about 5–20%, and reducing the local control rate of the tumor to a certain extent, becoming an important limiting factor in the dose of radiotherapy [8, 9].

Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/interferon stimulating factors (STING) signaling pathway, as an important part of the innate immune system, plays an important role in maintaining the homeostasis of the body. Blocking the cGAS-STING pathway inhibited inflammatory response and reduced tissue damage, while activating the cGAS-STING pathway promoted antiviral and anti-tumor effects. Understanding the regulatory mechanism of cGAS-STING pathway links can use existing drugs or new drugs developed to intervene in the cGAS-STING pathway and provide new ideas and methods for the treatment of clinically relevant diseases. The purpose of this review is to discuss the immune activation mechanism of cGAS-STING in lung cancer radiotherapy and the mechanism of lung injury and to discuss new therapeutic methods.

Structure and signal transduction of the cGAS/STING pathway

The cGAS-STING signaling pathway is an important cytosolic DNA sensing pathway in vivo, which induces the expression of type I IFNs and affects the immune response of the body, and plays an important role in regulating pathogen infection, tumor immunity,and autoimmune diseases. cGAS has a highly conserved nucleotidyltransferase domain and can detect double-strands DNA (dsDNA) released into the cytosol. The Binding of cGAS and dsDNA is length-dependent and independent of DNA sequences [10]. dsDNA binds to cGAS as a dimer in a 2:2 manner, causing a conformational change in the active site of cGAS [11], which in turn promotes ATP and GTP synthesis of the second messenger cyclic guanylin acid-adenylate (cGAMP) [12]. Synthetic cGAMP binds to STING molecules localized as homodimers on the ER [13], resulting in conformational changes in STING to form STING tetramers and higher-order oligomers. This conformational change induces activation of STING, which shifts to a more closed conformation and translocates to the Golgi through the ER-Golgi intermediate compartment [14]. Structural studies have shown that two cysteine residues on STING protein are palmitoylated in the Golgi apparatus and recruit and activate TANK binding kinase-1 (TBK1) in the Golgi apparatus [15, 16]. Activation of TBK1 in turn transphosphorylates the C-terminal domain of STING, thereby recruiting interferon regulatory factor 3 (IRF3) [17]. Phosphorylated IRF3 dimerizes and translocates into the nucleus to activate transcription of genes encoding type I IFNs 1 7. STING can also simultaneously activate IκB kinases (IKK) to mediate the transcription of interleukin-6 (IL-6), tumor necrosis factor (TNF), and inflammatory cytokines such as type I IFN activated by nuclear factor kappa-B (NF-κB) [18]. It plays an immunoregulatory role and affects the body’s viral defense, inflammation, and cancer treatment.(Fig. 1).

Fig. 1
figure 1

The cGAS-STING DNA sensing signaling pathway

Antitumor immunity in the cGAS/STING pathway versus radiotherapy

DNA, as the blueprint of life, is regulated by DNA damage response or DNA damage repair system (DDR) to ensure the integrity of the genome. When DNA is damaged, the DDR pathway causes cell cycle arrest and repairs DNA, ensuring that cell cycle cycles occur orderly; however, when damage cannot be effectively repaired, cells enter senescence; if DNA is severely damaged, it directly leads to apoptosis [19]. Radiotherapy (RT) is used as a traditional treatment in more than 50% of patients with malignant tumors, and its principle of action is to directly cause fatal DNA damage in irradiated cells, or indirectly induce DNA damage by producing reactive oxygen species [6]; its types of induced DNA damage include base mutations, single-strand breaks (SSBs), and double-strand breaks (DSBs) [20]. Radiation induction regulates the tumor microenvironment and promotes the recruitment and infiltration of immune cells. Radiation therapy (RT) can cause DNA damage in tumor cells [21], leading to chromosomal instability (CIN). CIN is a characteristic feature of cancer and it will induce micronuclei formation [22, 23].Human mtDNA’s nucleotide sequence was discovered in 1981 to be a 16 569 bp double-stranded circular molecule that codes for 37 genes that can express a number of important proteins involved in oxidative phosphorylation and essential for cellular energy metabolism [24]. The permeability of the inner and outer mitochondrial membranes varies when cancer cells experience oxidative stress and mitochondrial malfunction, and mitochondrial DNA can exit the mitochondrial outer membrane into the cytosol by mitochondrial membrane permeability transition holes or BAX/BAK-dependent permeabilization of the outer mitochondrial membrane. Inflammation is brought on by mtDNA translocation into the cytoplasm, cGAS recognition and binding, and subsequent activation of downstream immune pathways [25]. In a model of cisplatin-induced acute kidney injury, cisplatin caused mtDNA leakage into the cytoplasm through the mitochondrial outer membrane BAX/BAK pore, activated the cGAS-STING signaling pathway, and resulted in inflammation and acute kidney injury. By inhibiting STING, however, the cisplatin-induced renal inflammation could be reduced [26]. dsDNA from micronuclei as well as DNA damage induction will be recognized by the cytoplasmic DNA sensor cGAS, thereby activating the cGAS-STING signaling pathway, shaping innate immunity in a type I IFN-dependent manner, promoting adaptive immune responses, and thus effectively anti-tumor [27, 28].

Cancer cells are irradiated and exposed to tumor-specific antigens, which make them visible for immune surveillance and promote the activation of cytotoxic T cells. In the absence of STING in the host, the amount of IFN-β induced in the tumor decreased after irradiation. Consistent with the immunogenicity of IFNs, the radiation effect was attenuated in STING deficient mice compared with controls, indicating that STING dependent cytosolic DNA sensing plays a critical role in the radiation effect in vivo. Generally, tumor cells escape immune response through the inactivation of the STING signaling pathway. When the cGAS-STING signaling pathway in tumor cells is activated, it induces expression of cytokines such as IL-6, TNF, and type I IFN, leading to tumor cell death or apoptosis, releases dsDNA, and other tumor-derived antigens, activates dendritic cells (DCs), and then initiates anti-tumor immunity. In the tumor microenvironment, tumor-derived DNA can be taken up by DCs through unknown mechanisms, further triggering stronger adaptive anti-tumor immune response [29]. Tumor cells enhanced cross-antigen presentation by DCs after irradiation, whereas DCs lacking STING was unable to cross-infect primary CD + 8 T cells [30]. Compared with irradiation alone, cGAMP combined with irradiation effectively reduced tumor burden in vivo, indicating that cGAMP treatment enhanced radiotherapy efficacy. DNA produced by tumor cells after radiotherapy promotes the anti-tumor effect of radiotherapy by triggering the cGAS/STING pathway to enhance T cell responses in mice to induce anticancer immunity in a STINE-dependent manner [30]. However, whether radiotherapy can induce cGAS-STING mediated anti-tumor effect is related to the applied radiation dose, too high dose will affect the survival of immune cells in the tumor microenvironment, but also up-regulate the negative regulators of the DNA sensing pathway, inhibit cGAS-STING pathway activation, and then promote immunosuppression, while lower dose can effectively activate the cGAS/STING pathway. Vanpouille-Box et al. found that Trex1 is a DNA exonuclease that acts as an upstream regulator of RT-driven anti-tumor immunity, and the DNA exonuclease Trex1 is produced in different cancer cells after irradiation at doses of 12 to 18 Gy, mainly by degrading cytosolic DNA, resulting in a decrease in dsDNA, a ligand required to activate cGAS/STING, and inhibiting anti-tumor immune effects [31].Repeated irradiation at doses that do not induce Trex1 promotes IFN-β production, thereby recruiting and activating Batf3-dependent DCs. This effect is essential for priming of CD + 8 T cells that mediate systemic tumor rejection. Radiotherapy leads to dsDNA accumulation in cancer cells when Trex1 is not activated type I IFNs are activated through the cGAS/STING pathway, downstream recruitment of DCs, and activation of CD + 8 T cells or anti-PD-1 antibodies to initiate tumor rejection. In tumors treated with irradiation doses exceeding the Trex1 induction threshold, clearance of dsDNA from cancer cell sol hinders IFN-β release in cancer cells, resulting in a reduced recruitment of DCs and insufficient activation of CD + 8 T cells leading to diminished local tumor regression [32]. Jason J. found unfavorable local tumor response was linked to overexpression of the DNase III (TREX1) exonuclease [33].RT acts as an inducer of antitumor immune responses, in part, depending on the type I IFN secretion caused by activation of the cGAS/STING signaling pathway and the interaction between immune cells. Exosomes are membrane microvesicles that range in size from 30 to 100 nm and are released by all types of cells. They offer a highly developed method of local and distant intercellular communication [34]. Julie M showed that irradiated mouse breast cancer cells (RT-TEX) create tumor-derived exosomes (TEX) that transmit dsDNA to DCs and cause them to upregulate costimulatory molecules and STING-dependent IFN-I activation [35].Therefore, exploring the molecular mechanisms affecting the activation of this pathway will provide new targets for enhancing RT-induced anti-tumor immunity.(Fig. 2).

Fig. 2
figure 2

The cGAS–cGAMP–STING pathway detects cytoplasmic DNA after Ionizing radiation and activate type I IFNs and other cytokines

CTL, cytotoxic T lymphocytes; CXCL9, chemokine (C-X-C motif) ligand 9; IFN, interferon; PD-1/PD-L1, anti-programmed death-1/programmed death-ligand 1; TA, tumor antigen;

cGAS/STING pathway and radiation-induced lung injury

Radiation therapy is one of the important treatment methods for thoracic malignant tumors. During the treatment, inevitable lung tissues will be irradiated with a certain dose [36], which causes different degrees of radiation-induced lung injury, with early manifestations of radiation pneumonitis and radiation-induced pulmonary fibrosis, thus making it difficult for the chest irradiation dose routinely used in clinical practice to reach the dose required for radical resection of tumors, and it is also difficult to further improve the local tumor control rate in patients with thoracic tumors, especially inoperable non-small cell lung cancer. Radiation-induced lung injury occurs in 13–37% of patients receiving radiotherapy for lung cancer [37], and with the substantial progress in the technology level and equipment of radiotherapy in recent years, the incidence of R ILI, although decreasing, is still higher than 15% [38, 39], and the mortality rate is as high as 4% [40]. It is an important dose-limiting factor in thoracic radiotherapy and affects the quality of life of patients.

High doses of ionizing radiation are well-known to cause DNA double-strand breaks [41]. DNA damage leads to oxidative stress, vascular damage, and inflammation. Pneumonia develops within hours or days of high-dose irradiation and is associated with increased capillary permeability and accumulation of inflammatory cells in the lung [9, 42]. The recruited inflammatory cells secrete profibrotic cytokines to activate resident fibroblasts, ultimately leading to excessive collagen production and deposition in the interstitial space of the lung. Some major signaling pathways have been identified that are associated with the amplification of inflammatory cytokine cascades, with NF-κB playing an important role in the regulation of gene expression of various inflammatory cytokines [43]. NF-κB is an important transcription factor widely present in eukaryotic cells, which can participate in the transcriptional regulation of a variety of genes and is closely related to important physiological and pathological processes such as immune response, inflammatory response, as well as cell proliferation, differentiation, and apoptosis [44]. NF -κB binds to its inhibitor protein IκB at rest and is inactive [45]. When cells are stimulated by external stimuli, such as oxidative stress, infection, or physicochemical stimuli, they can activate the transcriptional activity of NF-κB and promote the acidification and degradation of IκB, thereby releasing NF-κB into the nucleus and binding to the κB site of the target gene, rapidly inducing target gene transcription and up-regulating the expression of various biomacromolecules, including inflammatory cytokines, chemokines, and apoptosis-related factors. Among them, inflammatory factors such as TNF -α and I L-1β can also up-regulate NF-κB transcriptional activity, thus forming positive feedback and continuously expanding the stimulation signal [46].The continuous expression and accumulation of these inflammatory factors can cause damage to tissue cells. Studies have shown that nuclear factor-κB inflammation-related pathways and cytokines mainly mediate inflammatory responses such as tumor necrosis factor TNF -α, interleukin − 1, and interleukin − 6 play an important role in the initiation and promotion of radiation-induced lung injury. NF-κB is continuously activated throughout radiation-induced lung injury, which may be one of the causes of persistent chronic inflammation [47]. NF-κB is a major switch that regulates the body’s inflammatory response. When the body is irradiated, intracellular DNA damages and activates the cGAS/STING pathway, phosphorylates NF-κB to inhibit protein kinase, so that NF-κB inhibitory protein IκB is ubiquitinated and degraded, thus activating NF-κB into the nucleus to initiate the release of a variety of inflammatory factors. Activation of inflammatory factors in the lung stimulates the phenotypic transformation of lung epithelial cells into intercellular cells, producing large amounts of extracellular matrix proteins that accumulate and deposit in the pulmonary interstitium and promote the formation of pulmonary fibrosis [48]. Therefore, reducing lung fibrosis and lung inflammation effectively protects against RILI. Because NF-κB factor is a heterodimer composed of two peptides, p 50 and p65, and only the C -terminus of p 65 protein contains a transcriptional activation region that can act directly on targeted genes to activate transcription, changes in NF-κB activity can currently be evaluated by detecting the expression of p 65. Related studies have shown that 3,3 ′-diindolylmethane (DIM) can inhibit the NF-κB pathway, and in mouse models, DIM can effectively ameliorate RILI-induced pulmonary fibrosis and lung inflammation while reducing the number of inflammatory cells in BALF as shown by H E staining HE staining and Masson staining [48]. Savita et al. showed that Quercetin-3-Rutinoside provides radioprotection to the lung by regulating NF-κB/TGF-β1 signaling, scavenging free radicals, preventing perivascular invasion, and prolonging the inflammatory cascade, which may otherwise lead to chronic radiation fibrosis. Inhibition of the cGAS/STING/NF-κB pathway is therefore a potential target for the treatment of radiation-induced lung injury.

cGAS/STING pathway combined with radiotherapy immunotherapy

Immune checkpoint inhibitors mainly include PD-1/PD-L1 antibody and CTLA-4 antibody. Theoretically, ICI therapy can re-edit the tumor immune microenvironment to induce tumor regression, and the activity of PD-1/PD-L1 immune checkpoint inhibitors varies in different cancers, and many studies have demonstrated that they confer resistance in multiple cancer types. The effectiveness of immune checkpoint inhibitors depends on innate antitumor immunity, particularly the production of tumor antigens and tumor-specific cytotoxic T cells (CTLs). However, most cancer patients remain unresponsive to checkpoint inhibitor therapy, most of which are due to their inability to mount sufficient antitumor immunity.

Numerous studies have shown weak responses to multiple immune checkpoints in STING null mice, including PD-1, PD-L1, CTLA4, and CD47 [47]. STING activators are ideal sensitizers for enhancing anti-PD-1/PD-L1 therapy, and STING agonists can significantly enhance cytotoxic T cell infiltration in tumor cells [49]. Immune checkpoints negatively regulate T cells in the immune system, and immune checkpoint blockade therapy has shown good efficacy in tumors, while the STING signaling pathway is essential for the anti-tumor effect of immune checkpoint blockade therapy in mice. In wild-type mouse melanoma models, intramuscular injection of cGAMP away from the tumor site significantly enhanced the therapeutic effect of immune checkpoint blockade, and the combination of cGAMP and PD-L1 antibody effectively inhibited tumor growth in B16 melanoma mice45. When cGAS, STING is absent, responses to immunotherapies such as immunosuppressive molecule blockade are weak, and the combination of STING agonists and PD-1 blockers shows stronger antitumor efficacy, and when combined, tumor models that do not respond well to immune checkpoint blockade become sensitized [50]. Radiotherapy has gained broad public consensus as an adjuvant to activate immune responses. Jason R. Baird et al. demonstrated synergistic local and distant tumor control when radiation therapy was combined with a novel STING ligand in a mouse model of pancreatic cancer [51]. Radiation therapy can act synergistically with PD-1 inhibitors to enhance their respective antitumor activity, particularly in patients with advanced tumors [52].

NF-κB control plays a crucial role in immune-inflammatory responses, tumorigenesis, and development/radioresistance. Inhibition of the canonical NF-κB pathway has been shown to attenuate radiation efficacy, whereas non-canonical NF-κB deficiency promotes radiation-induced antitumor immunity [53]. Mechanistic studies have shown that non-canonical NF-κB signaling in dendritic cell DCs is associated with activation of the DNA-reception-mediated STING pathway [53]. It has been shown that modulation of type I interferons by application of the NF-κB2 competitive inhibitor S N52 facilitated the therapeutic effect of IR, but tumors receiving combination therapy were not completely eliminated. Sustained IFN-I signaling induces immunosuppressive mechanisms, including PD-L1 expression on DCs and other myeloid cells and PD-1 expression on T cells, which leads to CD8 + T cell depletion [54, 55]. Tumor-bearing mice were treated with anti-PD-L1 agents following SN52 and IR treatment. PD-L1 blockade enhanced the therapeutic effect of the SN52 + IR combination and led to tumor rejection. To investigate whether this combined treatment could generate long-lasting protective T cell immunity, tumor-free mice were rechallenged with high-dose MC38 tumor cells on the contralateral side. A few weeks later, no palpable tumors were detected in treated mice. These findings reveal that combining IR with manipulation of the STING-IFN pathway and immune checkpoint inhibition can better activate innate immunity and reduce immunosuppression, and may provide a novel approach for the treatment of tumors, that is, inhibition of non-canonical NF-kB plus antiPD-L1 antibody drugs [55].

Concsulion

Activation of the cGAS-STING pathway in lung cancer radiotherapy has shown great antitumor potential by promoting the secretion of type I IFNs, significantly increasing the infiltration of CD + 8 T cells and effectively stimulating anti-tumor immune responses in the body. When exosomes deliver dsDNA from radiation-damaged cancer cells to DCs, type I interferon (IFN-I) is activated via the cGAS/STING pathway.RT-TEX serve as transmitters of the molecular alterations that occurred in radioactively treated cancer cells. Adjuvants that activated DCs and might trigger protective antitumor T-cell responses were conveyed by RT-TEX together with tumor antigens.When the STING signaling pathway is used for anti-tumor therapy, therapeutic window and toxic side effects should be considered, and excessive activation of the pathway is associated with the occurrence of related injuries. Although the combination of radiation and immunotherapy has proven effective in preclinical studies, the future application of this combination remains challenging. Optimization of radiation dose and time and identification of potential biomarkers can further enhance the effectiveness of this unique combination, and a better understanding of how the cGAS/STING pathway mediates anti-tumor effects will contribute to the use of STING agonists in cancer therapy and design better treatment regimens.

Data Availability

Not applicable.

References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. https://doi.org/10.3322/caac.21660

    Article  PubMed  Google Scholar 

  2. Duma N, Santana-Davila R, Molina JR. Non-Small Cell Lung Cancer: Epidemiology, Screening, diagnosis, and treatment. Mayo Clin Proc. 2019;94(8):1623–40. https://doi.org/10.1016/j.mayocp.2019.01.013

    Article  CAS  PubMed  Google Scholar 

  3. Jazieh AR, Zeitouni M, Alghamdi M, et al. Management guidelines for stage III non-small cell lung cancer. Crit Rev Oncol Hematol. 2021;157:103144. https://doi.org/10.1016/j.critrevonc.2020.103144

    Article  PubMed  Google Scholar 

  4. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553(7689):446–54. https://doi.org/10.1038/nature25183

    Article  CAS  PubMed  Google Scholar 

  5. Arbour KC, Riely GJ. Systemic therapy for locally Advanced and Metastatic Non-Small Cell Lung Cancer: a review. JAMA. 2019;322(8):764–74. https://doi.org/10.1001/jama.2019.11058

    Article  CAS  PubMed  Google Scholar 

  6. Lu AG, Feng H, Wang PX, Han DP, Chen XH, Zheng MH. Emerging roles of the ribonucleotide reductase M2 in colorectal cancer and ultraviolet-induced DNA damage repair. World J Gastroenterol. 2012;18(34):4704–13. https://doi.org/10.3748/wjg.v18.i34.4704

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Huang N, Guo W, Ren K, et al. LncRNA AFAP1-AS1 supresses mir-139-5p and promotes cell proliferation and Chemotherapy Resistance of Non-small Cell Lung Cancer by competitively upregulating RRM2. Front Oncol. 2019;9:1103. https://doi.org/10.3389/fonc.2019.01103. Published 2019 Oct 22.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Yao Y, Zheng Z, Song Q. Mesenchymal stem cells: a double-edged sword in radiation-induced lung injury. Thorac Cancer. 2018;9(2):208–17. https://doi.org/10.1111/1759-7714.12573

    Article  PubMed  Google Scholar 

  9. Hanania AN, Mainwaring W, Ghebre YT, Hanania NA, Ludwig M. Radiation-Induced Lung Injury: Assessment and Management. Chest. 2019;156(1):150–62. https://doi.org/10.1016/j.chest.2019.03.033

    Article  PubMed  PubMed Central  Google Scholar 

  10. Luecke S, Holleufer A, Christensen MH, et al. cGAS is activated by DNA in a length-dependent manner. EMBO Rep. 2017;18(10):1707–15. https://doi.org/10.15252/embr.201744017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang X, Wu J, Du F, et al. The cytosolic DNA sensor cGAS forms an oligomeric complex with DNA and undergoes switch-like conformational changes in the activation loop. Cell Rep. 2014;6(3):421–30. https://doi.org/10.1016/j.celrep.2014.01.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ablasser A, Goldeck M, Cavlar T, et al. cGAS produces a 2’-5’-linked cyclic dinucleotide second messenger that activates STING. Nature. 2013;498(7454):380–4. https://doi.org/10.1038/nature12306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Burdette DL, Monroe KM, Sotelo-Troha K, et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478(7370):515–8. https://doi.org/10.1038/nature10429. Published 2011 Sep 25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Srikanth S, Woo JS, Wu B, et al. The Ca2+ sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum. Nat Immunol. 2019;20(2):152–62. https://doi.org/10.1038/s41590-018-0287-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mukai K, Konno H, Akiba T, et al. Activation of STING requires palmitoylation at the golgi. Nat Commun. 2016;7:11932. https://doi.org/10.1038/ncomms11932. Published 2016 Jun 21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang C, Shang G, Gui X, Zhang X, Bai XC, Chen ZJ. Structural basis of STING binding with and phosphorylation by TBK1. Nature. 2019;567(7748):394–8. https://doi.org/10.1038/s41586-019-1000-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Liu S, Cai X, Wu J, et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science. 2015;347(6227):aaa2630. https://doi.org/10.1126/science.aaa2630

    Article  CAS  PubMed  Google Scholar 

  18. Dunphy G, Flannery SM, Almine JF, et al. Non-canonical activation of the DNA sensing adaptor STING by ATM and IFI16 mediates NF-κB signaling after nuclear DNA damage. Mol Cell. 2018;71(5):745–760e5. https://doi.org/10.1016/j.molcel.2018.07.034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li T, Chen ZJ. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018;215(5):1287–99. https://doi.org/10.1084/jem.20180139

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Iwamoto K, Nakashiro K, Tanaka H, Tokuzen N, Hamakawa H. Ribonucleotide reductase M2 is a promising molecular target for the treatment of oral squamous cell carcinoma. Int J Oncol. 2015;46(5):1971–7. https://doi.org/10.3892/ijo.2015.2912

    Article  CAS  PubMed  Google Scholar 

  21. Toulany M. Targeting DNA, double-strand break repair pathways to improve Radiotherapy Response. Genes (Basel). 2019;10(1):25. https://doi.org/10.3390/genes10010025. Published 2019 Jan 4.

    Article  CAS  PubMed  Google Scholar 

  22. Xia T, Konno H, Ahn J, Barber GN. Deregulation of STING Signaling in Colorectal Carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Rep. 2016;14(2):282–97. https://doi.org/10.1016/j.celrep.2015.12.029

    Article  CAS  PubMed  Google Scholar 

  23. Liu Y, Dong Y, Kong L, Shi F, Zhu H, Yu J. Abscopal effect of radiotherapy combined with immune checkpoint inhibitors. J Hematol Oncol. 2018;11(1):104. https://doi.org/10.1186/s13045-018-0647-8. Published 2018 Aug 16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Anderson S, Bankier AT, Barrell BG, et al. Sequence and organization of the human mitochondrial genome. Nature. 1981;290(5806):457–65. https://doi.org/10.1038/290457a0

    Article  CAS  PubMed  Google Scholar 

  25. Riley JS, Quarato G, Cloix C, et al. Mitochondrial inner membrane permeabilisation enables mtDNA release during apoptosis. EMBO J. 2018;37(17):e99238. https://doi.org/10.15252/embj.201899238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Maekawa H, Inoue T, Ouchi H, et al. Mitochondrial damage causes inflammation via cGAS-STING signaling in Acute kidney Injury. Cell Rep. 2019;29(5):1261–1273e6. https://doi.org/10.1016/j.celrep.2019.09.050

    Article  CAS  PubMed  Google Scholar 

  27. Mackenzie KJ, Carroll P, Martin CA, et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. 2017;548(7668):461–5. https://doi.org/10.1038/nature23449

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell. 2013;154(1):47–60. https://doi.org/10.1016/j.cell.2013.06.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. https://doi.org/10.1038/ncomms15618. Published 2017 Jun 9.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Fu J, Kanne DB, Leong M, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7(283):283ra52. https://doi.org/10.1126/scitranslmed.aaa4306

    Article  PubMed  PubMed Central  Google Scholar 

  31. Li C, Zheng J, Chen S, et al. RRM2 promotes the progression of human glioblastoma. J Cell Physiol. 2018;233(10):6759–67. https://doi.org/10.1002/jcp.26529

    Article  CAS  PubMed  Google Scholar 

  32. Ahn J, Xia T, Konno H, Konno K, Ruiz P, Barber G. Inflammation-driven carcinogenesis is mediated through STING. Nat Commun. 2014;5:5166. https://doi.org/10.1038/ncomms6166. Published 2014 Oct 10.

    Article  CAS  PubMed  Google Scholar 

  33. Luke JJ, Onderdonk BE, Bhave SR, et al. Improved Survival Associated with local Tumor Response following Multisite Radiotherapy and Pembrolizumab: secondary analysis of a phase I Trial. Clin Cancer Res. 2020;26(24):6437–44. https://doi.org/10.1158/1078-0432.CCR-20-1790

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ludwig AK, Giebel B. Exosomes: small vesicles participating in intercellular communication. Int J Biochem Cell Biol. 2012;44(1):11–5. https://doi.org/10.1016/j.biocel.2011.10.005

    Article  CAS  PubMed  Google Scholar 

  35. Diamond JM, Vanpouille-Box C, Spada S, et al. Exosomes Shuttle TREX1-Sensitive IFN-Stimulatory dsDNA from irradiated Cancer cells to DCs. Cancer Immunol Res. 2018;6(8):910–20. https://doi.org/10.1158/2326-6066.CIR-17-0581

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. McBride WH, Schaue D. Radiation-induced tissue damage and response. J Pathol. 2020;250(5):647–55. https://doi.org/10.1002/path.5389

    Article  PubMed  PubMed Central  Google Scholar 

  37. Das SK, Zhou S, Zhang J, Yin FF, Dewhirst MW, Marks LB. Predicting lung radiotherapy-induced pneumonitis using a model combining parametric Lyman probit with nonparametric decision trees. Int J Radiat Oncol Biol Phys. 2007;68(4):1212–21. https://doi.org/10.1016/j.ijrobp.2007.03.064

    Article  PubMed  PubMed Central  Google Scholar 

  38. Jenkins P, Welsh A. Computed tomography appearance of early radiation injury to the lung: correlation with clinical and dosimetric factors. Int J Radiat Oncol Biol Phys. 2011;81(1):97–103. doi: 10.1016/j.ijrobp.2010.05.017. Epub 2011 May 3. PMID: 21543163.

  39. Jenkins P, Watts J. An improved model for predicting radiation pneumonitis incorporating clinical and dosimetric variables. Int J Radiat Oncol Biol Phys. 2011;80(4):1023–9. https://doi.org/10.1016/j.ijrobp.2010.03.058

    Article  PubMed  Google Scholar 

  40. Sekine I, Sumi M, Ito Y, et al. Retrospective analysis of steroid therapy for radiation-induced lung injury in lung cancer patients. Radiother Oncol. 2006;80(1):93–7. https://doi.org/10.1016/j.radonc.2006.06.007

    Article  CAS  PubMed  Google Scholar 

  41. Huang L, Snyder AR, Morgan WF. Radiation-induced genomic instability and its implications for radiation carcinogenesis. Oncogene. 2003;22(37):5848–54. https://doi.org/10.1038/sj.onc.1206697

    Article  CAS  PubMed  Google Scholar 

  42. Kim H, Park SH, Han SY, Lee YS, Cho J, Kim JM. LXA4-FPR2 signaling regulates radiation-induced pulmonary fibrosis via crosstalk with TGF-β/Smad signaling. Cell Death Dis. 2020;11(8):653. Published 2020 Aug 8. https://doi.org/10.1038/s41419-020-02846-7

  43. Jin H, Yoo Y, Kim Y, Kim Y, Cho J, Lee YS. Radiation-Induced Lung Fibrosis: Preclinical Animal Models and therapeutic strategies. Cancers (Basel). 2020;12(6):1561. https://doi.org/10.3390/cancers12061561. Published 2020 Jun 12.

    Article  CAS  PubMed  Google Scholar 

  44. Caamaño J, Hunter CA. NF-kappaB family of transcription factors: central regulators of innate and adaptive immune functions. Clin Microbiol Rev. 2002;15(3):414–29. https://doi.org/10.1128/CMR.15.3.414-429.2002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jacobs MD, Harrison SC. Structure of an IkappaBalpha/NF-kappaB complex. Cell. 1998;95(6):749–58. https://doi.org/10.1016/s0092-8674(00)81698-0

    Article  CAS  PubMed  Google Scholar 

  46. Tak PP, Firestein GS. NF-kappaB: a key role in inflammatory diseases. J Clin Invest. 2001;107(1):7–11. https://doi.org/10.1172/JCI11830

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Haase MG, Klawitter A, Geyer P et al. Sustained elevation of NF-kappaB DNA binding activity in radiation-induced lung damage in rats [published correction appears in Int J Radiat Biol. 2004;80(3):247]. Int J Radiat Biol. 2003;79(11):863–877. https://doi.org/10.1080/09553000310001632903

  48. Demaria O, De Gassart A, Coso S, et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci U S A. 2015;112(50):15408–13. https://doi.org/10.1073/pnas.1512832112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang H, Hu S, Chen X, et al. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci U S A. 2017;114(7):1637–42. https://doi.org/10.1073/pnas.1621363114

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. 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(8):676–84. https://doi.org/10.1158/2326-6066.CIR-17-0049

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhou X, Bao WA, Zhu X, et al. 3,3’-Diindolylmethane attenuates inflammation and fibrosis in radiation-induced lung injury by regulating NF-κB/TGF-β/Smad signaling pathways. Exp Lung Res. 2022;48(3):103–13. https://doi.org/10.1080/01902148.2022.2052208

    Article  CAS  PubMed  Google Scholar 

  52. Demaria S, Coleman CN, Formenti SC. Radiotherapy: changing the game in Immunotherapy. Trends Cancer. 2016;2(6):286–94. https://doi.org/10.1016/j.trecan.2016.05.002

    Article  PubMed  PubMed Central  Google Scholar 

  53. Hou Y, Liang H, Rao E, et al. Non-canonical NF-κB antagonizes STING sensor-mediated DNA sensing in Radiotherapy. Immunity. 2018;49(3):490–503e4. https://doi.org/10.1016/j.immuni.2018.07.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Garcia-Diaz A, Shin DS, Moreno BH et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression [published correction appears in Cell Rep. 2019;29(11):3766]. Cell Rep. 2017;19(6):1189–1201. https://doi.org/10.1016/j.celrep.2017.04.031

  55. Tang H, Liang Y, Anders RA, et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J Clin Invest. 2018;128(2):580–8. https://doi.org/10.1172/JCI96061

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

No external funding or material support was received for this work. Likewise, no funded writing assistance was provided.

Author information

Authors and Affiliations

Authors

Contributions

Chunsheng Yang and Meili Sun wrote the main manuscript text and Yan Liang and Ning Liu prepared Figs. 1 and 2. All authors reviewed the manuscript.

Corresponding author

Correspondence to Meili Sun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Manuscript is approved by all authors for publication.

Competing interests

No conflict of interest or financial ties exits in the submission of this manuscript, I would like to declare on behalf of my co-authors that the work described was original research that has not been published previously, and not under consideration for publication elsewhere, in whole or in part.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, C., Liang, Y., Liu, N. et al. Role of the cGAS-STING pathway in radiotherapy for non-small cell lung cancer. Radiat Oncol 18, 145 (2023). https://doi.org/10.1186/s13014-023-02335-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-023-02335-z

Keywords