Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Targeting STING with covalent small-molecule inhibitors

Abstract

Aberrant activation of innate immune pathways is associated with a variety of diseases. Progress in understanding the molecular mechanisms of innate immune pathways has led to the promise of targeted therapeutic approaches, but the development of drugs that act specifically on molecules of interest remains challenging. Here we report the discovery and characterization of highly potent and selective small-molecule antagonists of the stimulator of interferon genes (STING) protein, which is a central signalling component of the intracellular DNA sensing pathway1,2. Mechanistically, the identified compounds covalently target the predicted transmembrane cysteine residue 91 and thereby block the activation-induced palmitoylation of STING. Using these inhibitors, we show that the palmitoylation of STING is essential for its assembly into multimeric complexes at the Golgi apparatus and, in turn, for the recruitment of downstream signalling factors. The identified compounds and their derivatives reduce STING-mediated inflammatory cytokine production in both human and mouse cells. Furthermore, we show that these small-molecule antagonists attenuate pathological features of autoinflammatory disease in mice. In summary, our work uncovers a mechanism by which STING can be inhibited pharmacologically and demonstrates the potential of therapies that target STING for the treatment of autoinflammatory disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of small-molecule inhibitors of STING.
Fig. 2: Inhibition of STING through covalent binding of C-178 to Cys91.
Fig. 3: C-178 blocks palmitoylation-induced clustering of STING.
Fig. 4: In vivo effects of C-176.
Fig. 5: H-151 is a covalent STING antagonist.

Similar content being viewed by others

References

  1. Ishikawa, H., Ma, Z. & Barber, G. N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 461, 788–792 (2009).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  2. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP–AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  ADS  PubMed  CAS  Google Scholar 

  3. Burdette, D. L. et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature 478, 515–518 (2011).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  4. Ablasser, A. et al. cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  5. Diner, E. J. et al. The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell Reports 3, 1355–1361 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Gao, P. et al. Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP–AMP synthase. Cell 153, 1094–1107 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Wu, J. et al. Cyclic GMP–AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013).

    Article  ADS  PubMed  CAS  Google Scholar 

  8. Zhang, X. et al. Cyclic GMP–AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol. Cell 51, 226–235 (2013).

    Article  PubMed  CAS  Google Scholar 

  9. Abe, T. & Barber, G. N. Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-κB activation through TBK1. J. Virol. 88, 5328–5341 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15, 760–770 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Gall, A. et al. Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity 36, 120–131 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Jeremiah, N. et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. J. Clin. Invest. 124, 5516–5520 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Liu, Y. et al. Activated STING in a vascular and pulmonary syndrome. N. Engl. J. Med. 371, 507–518 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Crow, Y. J. & Manel, N. Aicardi–Goutières syndrome and the type I interferonopathies. Nat. Rev. Immunol. 15, 429–440 (2015).

    Article  PubMed  CAS  Google Scholar 

  15. Ahn, J., Gutman, D., Saijo, S. & Barber, G. N. STING manifests self DNA-dependent inflammatory disease. Proc. Natl Acad. Sci. USA 109, 19386–19391 (2012).

    Article  ADS  PubMed  Google Scholar 

  16. Ahn, J. et al. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 5, 5166 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. An, J. et al. Expression of cyclic GMP–AMP synthase in patients with systemic lupus erythematosus. Arthritis Rheumatol. 69, 800–807 (2017).

    Article  PubMed  CAS  Google Scholar 

  18. King, K. R. et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017).

    Article  PubMed  CAS  Google Scholar 

  19. Zeng, L. et al. ALK is a therapeutic target for lethal sepsis. Sci. Transl. Med. 9, eaan5689 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kerur, N. et al. cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat. Med. 24, 50–61 (2018).

    Article  PubMed  CAS  Google Scholar 

  21. Cavlar, T., Deimling, T., Ablasser, A., Hopfner, K. P. & Hornung, V. Species-specific detection of the antiviral small-molecule compound CMA by STING. EMBO J. 32, 1440–1450 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Conlon, J. 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. 190, 5216–5225 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Dennehy, M. K., Richards, K. A., Wernke, G. R., Shyr, Y. & Liebler, D. C. Cytosolic and nuclear protein targets of thiol-reactive electrophiles. Chem. Res. Toxicol. 19, 20–29 (2006).

    Article  PubMed  CAS  Google Scholar 

  24. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  25. Saitoh, T. et al. Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response. Proc. Natl Acad. Sci. USA 106, 20842–20846 (2009).

    Article  ADS  PubMed  Google Scholar 

  26. Mukai, K. et al. Activation of STING requires palmitoylation at the Golgi. Nat. Commun. 7, 11932 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  27. Stetson, D. B., Ko, J. S., Heidmann, T. & Medzhitov, R. Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell 134, 587–598 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Gao, D. et al. Activation of cyclic GMP–AMP synthase by self-DNA causes autoimmune diseases. Proc. Natl Acad. Sci. USA 112, E5699–E5705 (2015).

    Article  PubMed  CAS  Google Scholar 

  29. Gray, E. E., Treuting, P. M., Woodward, J. J. & Stetson, D. B. Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi–Goutières Syndrome. J. Immunol. 195, 1939–1943 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Ablasser, A. et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  31. Morita, M. et al. Gene-targeted mice lacking the Trex1 (DNase III) 3′→5′ DNA exonuclease develop inflammatory myocarditis. Mol. Cell. Biol. 24, 6719–6727 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Peschke, K. et al. Loss of Trex1 in dendritic cells is sufficient to trigger systemic autoimmunity. J. Immunol. 197, 2157–2166 (2016).

    Article  PubMed  CAS  Google Scholar 

  33. Gardeux, V., David, F. P. A., Shajkofci, A., Schwalie, P. C. & Deplancke, B. ASAP: a web-based platform for the analysis and interactive visualization of single-cell RNA-seq data. Bioinformatics 33, 3123–3125 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank N. Jordan, E. Simeoni and L. Muhandes for technical assistance and advice. We acknowledge the staff of the BSF-ACCESS screening platform at the EPFL for support, especially M. Chambon and J. Bortoli, and the staff of the ISIC Mass Spectrometry platform at the EPFL, especially N. Gasilova. We thank the following core facilities for their support: BIOp, CPG and HCF. A.A. received grants from the SNF (BSSGI0-155984, 31003A_159836), the Gebert Rüf Foundation (GRS-059_14) and the Else Kröner-Fresenius Stiftung (2014_A250). R.B. received funding from an AGS Research Award and the German Research council (DFG - BE 5877/2-1).

Reviewer information

Nature thanks Z. Chen, T. Taguchi, H. Wu and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

S.M.H., M.F.G., L.R., L.A., A.D. and M.H. designed, performed and analysed experiments, and S.M.H. and M.F.G. assisted in writing the manuscript. L.R. and A.G. synthesized chemical compounds. R.B. and M.F.G. designed, performed and analysed animal experiments. G.T. and G.F.v.d.G. provided advice. A.A. designed, performed and analysed experiments, wrote the manuscript with input from all authors, conceived the idea and supervised the study.

Corresponding author

Correspondence to Andrea Ablasser.

Ethics declarations

Competing interests

A.A. is a consultant to IFM Therapeutics, LLC. A.A., S.M.H., L.R. and the EPFL have filed provisional patent applications related to STING inhibitors. All other authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 A chemical screen identifies small-molecule inhibitors of STING.

a, Screening workflow. HEK mC-STING, HEK293T cells expressing mCherry–STING. b, Left, summary of the primary screen. Mean of normalized values for IFNβ luciferase activity in cells treated with compound versus cells treated with DMSO. Right, validation of selected candidates from the primary screen. Normalized values for IFNβ luciferase activity (light blue) induced by coexpression of cyclic di-GMP synthase (cdG-Syn) and STING are shown, in comparison to activity triggered by RIG-I (green). c, d, HEK293T cells expressing mCherry–STING were transfected with plasmids that encoded either cdG-Syn or RIG-I, as well as an IFNβ luciferase reporter, and then treated with C-178 or C-176 (1.25 μM–0.01 μM), after which IFNβ luciferase activity (c) or cell viability (using the CellTiter-Blue assay) (d) were measured. e, f, Chemical structures of derivatives of C-176 (e) and their effect (at a concentration of 0.5 μM) on IFNβ luciferase reporter activity triggered by mmSTING or RIG-I (f). Mean ± s.e.m. of n = 3 experiments (c, d, f).

Extended Data Fig. 2 Activity of C-178 against STING in distinct cells.

a, b, mRNA expression levels of Tnf in BMDMs activated with cGAMP, cyclic di-GMP (cdG), dsDNA, CMA or LPS for 5 h, after pretreatment for 1 h with C-178 (0.5 μM) or DMSO (n = 3 biological replicates). c, Heat map of RNA sequencing analysis of BMDMs treated with DMSO or CMA, in the presence or absence of C-178. The top 50 upregulated genes in cells treated with DMSO versus cells treated with CMA, in the absence of C-178, are shown for all conditions. d, Scatter plot of the dimensions PC1 versus PC2. e, Levels of expression of IFNB1 mRNA in THP-1 and WI-38 cells pretreated with C-178 (0.25 μM) or DMSO and stimulated with cGAMP for 3 h (n = 2 biological replicates). f, HEK293T cells were transfected with the construct chimaeric 1 (C1: hsSTING (amino acids 1–138)–mmSTING (amino acids 138–378)) or the construct chimaeric 2 (C2: mmSTING (amino acids 1–137)–hsSTING (amino acids 139–379)), together with cdG-Syn and IFNβ luciferase reporter, and treated with C-178 (0.5 μM) (n = 3 biological replicates). Data are mean or mean ± s.e.m. P values were determined by two-tailed t-test.

Extended Data Fig. 3 C-178 and C-176 covalently bind to Cys91.

a, BMDMs were treated with C-178 (0.25 μM) for 1 h, washed or left untreated and after 1 h were stimulated with cGAMP for 3 h. Levels of Ifnb1 mRNA were assessed by RT–qPCR. Data are mean ± s.e.m. (n = 3 biological replicates). bd, HEK293T cells expressing indicated Flag–mmSTING constructs were exposed to C-178 or C-176 (1 μM), lysed and Flag–mmSTING was immunoprecipitated. The eluted protein was analysed by intact mass spectrometry (LC-MS). b, Deconvoluted electrospray ionization mass spectrum (left), and expected and measured mass shifts after covalent binding of C-176 and C-178 (right) are shown. c, Deconvoluted electrospray ionization mass spectrum for Flag–mmSTING(C91S). d, Fragmentation map of Flag–mmSTING–C-176 analysed by top-down analysis, using LC-HCD-MS/MS (an additional 8 residues are due to N-terminal Flag). Data that were obtained with three different NCE values are combined to create a fragmentation map with assigned b- and y-fragments. Achieved sequence coverage is 15% with 20 p.p.m. mass accuracy for fragment assignment. One representative of n = 2 independent experiments is shown (b, c). For electrospray ionization spectra see Supplementary Information.

Extended Data Fig. 4 Gel-based analysis of compound interactions using clickable probes.

a, Structures of C-176-AL and C-176-AZ. b, HEK293T cells transfected with Flag–mmSTING and an IFNβ luciferase reporter were treated with C-176-AL, and luciferase activity was then assessed. Data are mean of n = 2. c, Labelling events of wild-type HEK293T cells and HEK293T with Flag–mmSTING incubated with C-176-AL (0.25 μM). d, Distinct labelling of mmSTING and hsSTING by C-176-AL (0.25 μM). e, Concentration-dependent competitor blockage of C-176-AL (0.25 μM) against C-178 and C-176-09 in HEK293T cells that express Flag–mmSTING. f, Labelling events of HEK293T cells that express Flag–mmSTING (wild-type mmSTING or mmSTING(C91S)) when exposed to iodoacetamide azide or C-176-AZ (both at 0.25 μM). g, HEK293T cells expressing Flag–mmSTING, GSTO1–Flag, MAP27K–Flag, MGMT–Flag or GSTP1–Flag were treated with C-176-AZ (0.25 μM). For in-gel fluorescence imaging, TAMRA azide (ce) or SiR alkyne (f, g) was used. Immunoblots against Flag or β-actin are shown and data are representative of n = 3 independent experiments (cg). h, Proposed reaction mechanism. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 5 C-178 and C-176 block activation-induced palmitoylation of STING.

a, Single-channel images of Flag and GM130 stainings of mouse embryonic fibroblasts that express Flag–mmSTING (control corresponding to images shown in Fig. 3), treated with CMA or DMSO. Scale bar, 20 μm. b, Protein quantification of p-TBK1, Flag–mmSTING and β-actin by immunoblot of mouse embryonic fibroblasts from a. c, [3H]-palmitate labelling of HEK293T cells that express the indicated Flag–mmSTING constructs, and which were treated with C-178, C176 (1 μM) or DMSO. d, e, [3H]-palmitate labelling of immunoprecipitated endogenous calnexin or transferrin receptor from HEK293T cells treated as indicated (compounds at 1 μM). f, HEK293T cells that express Flag–mmSTING were treated with C-178 (1 μM) or 2-bromopalmitate (2-BP) (50 μM) and stimulated with CMA for 1.5 h. Analysis of indicated proteins was performed by native PAGE or SDS–PAGE. g, Crosslinked lysates (DSS, 1 mM) of HEK293T cells that express Flag–mmSTING treated with C-178 (1 μM) and stimulated with CMA (2 h) were analysed by SDS–PAGE and immunoblotted for STING and p-TBK1. One representative of n = 3 (ac, eg) or n = 2 (d) independent experiments. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 6 In vivo effects of C-176 in wild-type mice.

a, Labelling of endogenous STING immunoprecipitaed from splenocytes of mice treated with C-176-AL, visualized by in-gel fluorescence. One representative of n = 2. b, Plasma concentration profiles of C-176 after a single-dose intraperitoneal injection into wild-type mice (n = 2 mice per condition). Data are mean of technical replicates. c, Serum levels of type I IFNs and IL-6 from wild-type mice pretreated with C-176 or vehicle 4 h after injection with CMA (n = 3 mice per condition). d, Body weight of wild-type mice during two weeks of daily DMSO and C-176 injection. eg, Mice from d were euthanized and blood samples were collected for measuring plasma levels of TNFα and IL-6 (e), blood cell counts (f), and liver and kidney parameters (g). Data are mean ± s.d. of n = 10 mice per condition (eg). P values were calculated by one-way ANOVA (c) or two-tailed t-test (eg). ns, not significant. For gel source data, see Supplementary Fig. 1. For source data, see Supplementary Table 1.

Source Data

Extended Data Fig. 7 Activity of C-178 and C-176 in TREX1-deficient cells and mice.

a, Wild-type (n = 2) or Trex1−/− (n = 7) mouse embryonic fibroblasts were treated with DMSO or C-178 (2 μM) overnight. mRNA levels of Isg15 and Cxcl10 were measured by RT–qPCR. b, c, Control mice (wild type or Trex1+/− (each n = 3)) were treated with vehicle, and Trex1−/− mice were treated with C-176 (n = 5 mice) or vehicle (n = 4 mice) for 11 days. Serum type I IFN levels (b) were measured and histological analysis of the heart (c) was performed. Scale bars, 50 μm. d, Histological analysis of distinct organs from wild-type (all n = 4) or Trex1−/− mice treated for 3 months. Smooth muscle, n = 3 (C-176) or 5 (vehicle); tongue and stomach, n = 3 (C-176) or 6 (vehicle). Representative histological images are shown. Data are mean ± s.e.m. P values were calculated using two-tailed t-test (a) or one-way ANOVA (bd). For source data, see Supplementary Table 1.

Source Data

Extended Data Fig. 8 C-170 and C-171 antagonize hsSTING.

a, Structure of C-170 and C-171. b, IFNβ luciferase reporter measurements from HEK293T cells transfected with indicated constructs (C-170 and C-171, 0.02–2 μM) (n = 3). c, d, THP-1 cells were pretreated with C-170 (0.5 μM) and stimulated with cGAMP or triphosphate RNA. IFNB1 and TNF mRNA levels were assessed by RT–qPCR (n = 5) (c), and p-TBK1 was determined by immunoblot (d). e, [3H]-palmitate labelling of HEK293T cells that express Flag–hsSTING, treated with C-170 (1 μM) or DMSO. f, HEK293T cells that express Flag–hsSTING (wild-type hsSTING or hsSTING(C91S)) were treated with C-170 (1 μM), lysed and Flag–hsSTING was analysed by intact mass measurement (LC-MS). Deconvoluted electrospray ionization mass spectrum showing intact mass indicated hsSTING constructs and treatments, are shown. Data are mean ± s.e.m. P values were calculated using two-tailed t-test. NS, not significant. One representative of n = 3 (d) and n = 2 (ef) independent experiments is shown. For gel source data, see Supplementary Fig. 1. For electrospray ionization spectra see Supplementary Information.

Extended Data Fig. 9 Mechanism of STING inhibition by H-151.

a, THP-1 cells that had been pretreated with H-151 or DMSO were stimulated with cGAMP or triphosphate RNA, or left unstimulated. IP-10 production was quantified by enzyme-linked immunosorbent assay after overnight incubation (n = 3). b, Levels of TNF mRNA assessed by RT–qPCR in THP-1 cells that had been pretreated with H-151 and stimulated with cGAMP or triphosphate RNA (n = 4). c, [3H]-palmitate labelling of immunoprecipitated endogenous calnexin or transferrin receptor from HEK293T cells that had been treated with H-151 (1 μM). d, Structure of H-151-AL. e, Competition assay of H-151-AL with H-151 in HEK293T cells that express Flag–hsSTING. Flag–hsSTING labelled with H-151-AL was visualized by in-gel fluorescence. f, HEK293T cells or HEK293T cells that express Flag–hsSTING were treated with H-151-AL, lysed and clicked to a SiR azide. Whole-cell lysates were analysed by in-gel fluorescence and by immunoblot. g, HEK293T cells that express Flag–hsSTING, GSTO1–Flag, MAP27K–Flag, MGMT–Flag or GSTP1–Flag were exposed to H-151-AL, clicked to TAMRA azide and visualized by in-gel fluorescence or immunoblot. h, Deconvoluted electrospray ionization mass spectrum showing intact mass of Flag–hsSTING(C91S) with or without H-151. i, Flag–hsSTING was purified from HEK293T cells that had been pretreated with H-151 (1 μM) and analysed by top-down analysis using LC-HCD-MS/MS (with additional 8 residues due to N-terminal Flag). Data that were obtained with three different NCE values are combined to create fragmentation map with assigned b- and y-fragments. Achieved sequence coverage is 23% with 20 p.p.m. mass accuracy for fragment assignment. Data are shown as mean ± s.e.m. P values were calculated by two-tailed t-test. NS, not significant. One of n = 2 experiments is shown (c, eh). For gel source data, see Supplementary Fig. 1. For electrospray ionization spectra, see Supplementary Information.

Extended Data Fig. 10 Activity of H-151 against mmSTING.

a, HEK293T cells were transfected with plasmids encoding mmSTING in combination with cdG-Syn or were transfected with a plasmid for TBK1, and an IFNβ luciferase reporter, and then treated with H-151 (concentration 0.02–2 μM). Reporter activity was measured after overnight incubation. Data are mean ± s.e.m. (n = 3; nonlinear regression analysis). Experiments were performed together with those that generated the data displayed in Fig. 5b. b, BMDMs were pretreated with H-151 (0.5 μM) and levels of Ifnb1 mRNA expression induced by cGAMP were assessed by RT–qPCR. Data are mean ± s.e.m. (n = 3); P value was calculated by two-tailed t-test. c, Mean plasma concentration profiles of H-151 following a single-dose intraperitoneal injection into wild-type mice (n = 3 mice per time point). d, Schematic, and in vivo detection of H-176-AL binding to mmSTING. Visualization by in-gel fluorescence was performed using SiR azide. One representative of n = 2. For gel source data, see Supplementary Fig. 1. For source data, see Supplementary Table 1.

Source Data

Extended Data Fig. 11 Mechanism of action of the identified STING antagonists.

In the absence of inhibition, ligand binding triggers the translocation of STING to the Golgi1,25, where palmitoylation occurs at cytoplasmic proximal cysteine residues (Cys88 and Cys91)26. In turn, this post-translational modification facilitates the multimerization of STING to create a platform—possibly at the lipid raft domain at the trans-Golgi network26—for the recruitment of TBK1, and thereby enables the initiation of downstream signalling. Through covalent interaction with Cys91, the compounds we describe here block the palmitoylation of STING and retain the protein in a signalling-incompetent state.

Supplementary information

Supplementary Data

This file contains the complete NMR spectra data

Reporting Summary

Supplementary Figure

This file contains Supplementary Figure 1

Supplementary Information

This file contains a Supplementary Note, which provides information detailing the palmitoylation of STING

Supplementary Information

This file contains information on chemical synthesis

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haag, S.M., Gulen, M.F., Reymond, L. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018). https://doi.org/10.1038/s41586-018-0287-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0287-8

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing