IRAK4-IN-4

Small molecule approaches to treat autoimmune and inflammatory diseases (Part II): Nucleic acid sensing antagonists and inhibitors

Xiaoqing Wang, Yafei Liu, Xingchun Han, Ge Zou, Wei Zhu, Hong Shen, HaiXia Liu
Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China

A B S T R A C T
Nucleic acid sensing pathways play an important role in the innate immune system, protecting hosts against infections. However, a large body of evidence supports a close association between aberrant activation of those pathways and autoimmune and inflammatory diseases. Part II of the digest series on small molecule approaches to autoimmune and inflammatory diseases concentrates on recent advances with respect to small molecule an- tagonists or inhibitors of the nucleic acid sensing pathways, including endosomal TLRs, NLRP3 inflammasome and cGAS-STING.

Introduction
The innate immune system of vertebrate organisms provides the first line of defense against pathogen infections by means of a diverse rangeof germline-encoded immune sensors called pattern recognition re- ceptors (PRRs).1,2 These receptors can recognize pathogen or pathogen-associated molecular patterns (PAMPs) and damage-associated molec- ular patterns (DAMPs). The major classes of PRRs are toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), nucleotide-binding oligomerization domain (NOD)-like receptors(NLRs), absent in melanoma 2 (AIM2)-like receptors (ALRs) and C-type lectin-like receptors (CLRs). The nucleic acid sensors3–7 (See Fig. 1) suchas endosomal TLRs and cytosolic RLRs, NLRs and ALRs can recognize DNA, RNA and their derivatives to trigger innate immune responses. In addition to the PRRs, guanosine monophosphate-adenosine mono- phosphate synthase (cGAS) is another cytosolic DNA sensor protein.
The activation of nucleic acid sensing pathways leads to the secretionof inflammatory molecules, particularly type I and type II interferons (IFNs)8,9 which promote a protective response against pathogens by inducing IFN-stimulated genes (ISGs) and by galvanizing a robustadaptive immune response.1 However, over-activation of the pathwaysis closely associated with many autoimmune and inflammatory diseases where a similar ISG signature is identified.10 Given their critical role in the pathogenesis of autoimmune and inflammatory diseases, nucleicacid sensing pathways have become attractive drug targets. Direct blockade of nucleic acid sensors may hold the promise of a comparable or even better efficacy than that of existing therapeutic agents such asJanus kinases (JAK) inhibitors against interferon signaling, and anti- bodies against specific cytokines, owing to a broader coverage of in- flammatory cytokines. Recent advances in understanding the structural basis of various targets in the nucleic acid sensing pathways such as TLR7, TLR8, cGAS and stimulator of interferon genes (STING) have had a profound impact on the discovery of small molecule antagonists or inhibitors in terms of druggability assessment, screening platform development and the following lead identification/optimization. In the sections below, the review focuses on the most investigated targets such as endosomal TLRs, NLRP3 and cGAS-STING, which are amenable totherapeutic targeting by small molecules. It should be noted that given these targets have previously been comprehensively reviewed,11–17 onlythe key advances from 2018 to 2020 are discussed.

Targeting endosomal TLRs
Ten TLRs (TLR1-10) have been identified in humans, among which TLR3, 7, 8 and 9 are mainly localized on the membrane of the endosome to recognize nucleic acids. TLR3 is activated by dsRNA, TLR7 and TLR8 detect ssRNA and TLR9 recognizes DNA containing an unmethylated CpG motif. TLRs comprise an extracellular domain, a transmembrane domain and an intracellular Toll/interleukin-1 receptor (TIR) domain. Upon ligand stimulation, the extracellular domain interacts with the ligand, inducing a conformational change of the TIR domain and initi- ating the recruitment of MyD88 or TRIF to trigger the production ofproinflammatory cytokines (tumor necrosis factor-α (TNF-α), IL-6 etc.) and chemokines.18 TLR7, 8, and 9 signal through a MyD88 cascade; incontrast, TLR3 engages TRIF for downstream signalling (See Fig. 1).
Mounting evidence indicates that dysregulation of the endosomal DNA- and RNA-sensing pathways is implicated in the pathogenesis of autoimmune diseases19 such as systemic lupus erythematosus (SLE)20and Sjogren’s syndrome (SS).21 Several review articles11,18,22–25 havesummarized TLR modulators and their therapeutic prospects. The recent discovery of small molecule antagonists targeting TLR7, 8 and 9 led to the identification of diverse chemical series with selective, dual- and pan-TLR antagonism, though it is unclear which TLR selectivity profileof antagonism provides optimal clinical therapeutic efficacy. The emerging co-crystal structures of TLR726 and TLR827 with small mole- cules clearly show that the antagonists’ binding hot spots are located at the interface of the ectodomain of TLR homodimers. TLR7 and TLR8share 40% sequence identity in their ectodomains, and 50% of the res- idues in their binding sites are identical.28 So, it is feasible to identifyselective or dual TLR7/8 antagonists. However, the precise TLR9 antagonist mechanism of action (MoA) remains unclear. Furthermore, the TLR9 ectodomain is related more distantly to TLR8 with 33%sequence identity overall and 25% with respect to residues in their binding sites.28 Therefore, identification of pan-TLR7/8/9 antagonists ismore challenging. Even for those alleged pan-antagonists, TLR9 potency is much weaker than that of TLR7/8. Table 1 summarizes the recently reported endosomal TLR antagonists according to their selectivities. Recent advances with respect to both clinical and preclinical endosomal TLR antagonists are covered in the following section.
At this time, there are at least four small molecules of endosomal TLR antagonists under clinical development (Fig. 2).29 The most advanced molecule, M5049 (1) from Merck KGaA, is undergoing a phase II studyto evaluate its safety and efficacy in COVID-19 pneumonia patients. Meanwhile, a phase I study to evaluate its safety, tolerability and pharmacokinetics in participants with SLE or cutaneous lupus erythe- matosus (CLE) is planned to start in 2021. In addition, BMS-986256 (2), developed by Bristol-Myers Squibb, is in phase I clinical studies to assessits safety and drug levels in subjects with CLE. The other two molecules, E6742 (structure undisclosed) from Eisai and CPG52364 (3) from Pfizer,have completed their phase I studies in healthy volunteers. While M5049,30 BMS-98625631 and E6742 are TLR7/8 dual antagonists, CPG5236432 is a TLR7/8/9 pan-antagonist.
In 2018, Yin’s group identified two distinct chemical scaffolds, pyrazolo[1,5-a]pyrimidine and 4-phenyl-1-(2H)-phthalazinone, as se- lective TLR8 inhibitors through high-throughput screening (HTS).27,47 Medicinal chemistry optimization led to the identification of CU-CPT8m (8), CU-CPT9a-b (9–10) with an IC50 within nanomolar and picomolar range, respectively, in a HEK-Blue TLR8 reporter assay. Both CU-CPT8m and CU-CPT9a doses dependently suppressed TNF-α in peripheral blood mononuclear cells (PBMCs) harvested from rheumatoid arthritis (RA)patients. Importantly, the co-crystal structures of TLR8 with CU-CPT8m and CU-CPT9b indicated that the small-molecule ligands stabilize the pre-formed TLR8 homodimer in its resting state and prevent its activa- tion by binding to a unique site at the dimeric interface. Based on the newly identified unconventional binding site and another HTS hit, they designed a triazole derivative TH1027 (11) as a selective TLR8 antag-onist with an IC50 of 30 2 nM.35 In addition, two more differentiatedchemical scaffolds were discovered with CU-72 (16) as a TLR7/8 dual antagonist and CU-115 (12) and CU-68 (13) as selective TLR8 antago- nists with relatively low potency.36
Based on the work of Shukla et al.,48–50 Karroum et al. discovered a series of compounds with the pyrazolo[1,5-a]quinoXaline scaffold as aselective TLR7 antagonist in 2019.33 The most potent compound (compound 4) showed an IC50 of 8.2 ± 1.6 μM in HEK-blue-hTLR7 cells without any TLR8 activity. According to a modeling study based on Yin’sco-crystal structure of TLR8-CU-CPTs, the difference in amino acids, Thr384 in TLR7 versus Ile403 in TLR8, plays a critical role in the selectivity of compound 4. Mukherjee et al. subsequently reported their discovery of a selective TLR7 antagonist from a well-established purine scaffold TLR7 agonist by a ‘chemical switch’, removing the O-alkylgroup at the C-2 position.34 The leading compound, compound 5, with a low micromolar IC50 and good pharmacokinetic properties was effective in ameliorating cutaneous pathology in a cutaneous autoimmune dis- ease psoriasis mouse model via oral dosing.
In 2020, Tojo et al. reported their discovery of potent and selective TLR7 antagonists.26 They successfully identified compounds 6 and 7with TLR7 IC50s of 15 and 25 nM, respectively, and high selectivity over TLR8 and TLR9, through modification of 8-oXoadenine derivatives of TLR7 agonists. More importantly, using crystallography and cryo- electron microscopy, they identified that both TLR7 agonists and an- tagonists bind to the interface of the TLR7 homodimer. The agonists induced a closed-form conformation, whereas antagonist 6 induced an open-form conformation. Interestingly, the less potent compound 7 could induce both conformations, which were in dynamic equilibrium. Moreover, compound 6 was shown to ameliorate autoimmunity in lupus-prone NZB/W F1 mice through decreasing proteinuria, increasing survival rate and reducing glomerular lesion scores in a dose-dependent manner via oral dosing.
In 2018 Ganguly’s group developed a series of potent and selectiveTLR9 antagonists.37 By modulating the substituents with varying ba- sicity, lipophilicity and flexibility at the quinazoline core, they suc-cessfully determined compound 14 to be the most selective TLR9 antagonist over TLR7 (>600-fold). By introducing lipophilic basic pro- pylpyrrolidine moiety, the compound could easily penetrate the cells and be trapped inside the acidic (pH 6.0–6.5) endosomal compartment, which increased the ligand concentration around the target. Using the same strategy, they also developed another purine series (e.g. compound15) with even more potent TLR9 antagonistic activity.38
In 2020, Novartis reported their discovery of TLR7/8 dual antagonist 17, discovered by means of a high throughput TLR8 antagonist competition assay to specifically identify binders of the ectodomain.39 Compound 17 inhibited IFN-α release in a dose-dependent manner in a ssRNA challenged mouse model through oral dosing, correlating withthe compound’s good in vitro efficacy and pharmacokinetic properties. In addition, Novartis reported two other distinct scaffolds, 1840 and 22,43 as TLR7/8 dual antagonist and TLR7/8/9 pan-antagonist,respectively, with very good in vitro activity.
In addition to the clinical TLR7/8 dual antagonist, BMS-986256, Bristol-Myers Squibb reported the discovery of compound 21 as a TLR7/8/9 pan-antagonist in 2020.28 However, the TLR9 activity wasrelatively weak in comparison to that of TLR7/8 activity. In mouse pharmacodynamics study, oral dosing of compound 21 could dose- dependently decrease agonist-induced IL-6 production. Compound 21 also dose-dependently alleviated the severity of lupus and psoriasis manifestation in MRL/lpr mice and imiquimod-induced psoriasis mice.
Recently, Roche has published several patentapplications41,42,44,45,51–55 which have disclosed several chemical scaf- folds as TLR7/8/9 pan-antagonists or TLR7/8 dual-antagonists. Some compounds such as compound 23 and compound 24 showed excellent TLR7/8/9 pan-antagonistic activity with IC50s below the assay detection limits, while other molecules such as compounds 19 and 20 exihibitedgood TLR7/8 dual-antagonistic activity with a >1000-fold selectivity over TLR9.
In 2020, Choi’s group reported the small molecule pan-TLR3/7/8/9 antagonist TAC5-a (25) that could inhibit all four of the nucleic acid sensing TLRs.46 It dose-dependently inhibited the secretion of proin- flammatory cytokines (TNF-α and IL-6) mediated by TLR3/7/8/9 in the dose range of 1–50 μM in cell-based assays without affecting other TLRs.
Although the in vitro potency was relatively weak, TAC5-a was confirmed to be effective in alleviating psoriasis and lupus symptoms and reducing disease markers in mouse models.

Targeting the NLRP3 inflammasome
Inflammasomes are multiprotein complexes consisting of NLR family members NLRP1, NLRP3 and NLRC4, and non-NLR receptors such as AIM2 and IFI16.56 The NLRP3 inflammasome is the most widelyinvestigated NLR given its important role in host defense and innateimmunity. The NLRP3 inflammasome is formed by three components: NLRP3 protein, procaspase-1 and ASC. NLRP3 protein contains PYD at the N-terminus, an array of 12 LRR at the C-terminus and the NACHT.57
PYDs interact with ASC, which contains a CARD. LRR senses microbial ligands and endogenous alarmins. NACHT contains ATPase activity and requires ATP binding for NLRP3 activation. Under normal conditions, the connection between NACHT and LRRs prevents the formation of inflammasome via inhibiting the interaction between NLRP3 protein and ASC.
NLRP3 responds to various stimuli, including viral RNA and extra- cellular ATP.58,59 The activation of NLRP3 inflammasome depends on two successive signals.58 In the initial priming step, internal and external DAMPs and PAMPs are recognized by TLRs to activate the nuclear NF-κB mediated signal, which results in the upregulation of inactive NLRP3 protein, proIL-1β and proIL-18. The subsequent activation step is trig- gered by further stimuli. Firstly, the conformation of NLRP3 protein ischanged to facilitate the interaction between PYDs in NLRP3 protein and ASC. Secondly, the procaspase-1 binds to ASC via CARD, leading to the formation of NLRP3 inflammasome. Thirdly, self-cleavage of procaspase-1 generates active caspase-1, leading to maturation of proIL- 1β and proIL-18 to IL-1β and IL-18. Then, inflammatory and immune responses are then induced (See Fig. 1). In addition, the active caspase-1 also cleaves and activates Gasdermin-D, which causes pyroptosis to formpore in the cell wall and the release of cytokines and cellular contents into extracellular space.60
There is a lot of evidence to support the hypothesis that the inap- propriate activation of NLRP3 inflammasome correlates with multiple diseases, such as autoimmune diseases61 (e.g. systemic sclerosis,IBD,61,62 RA and SLE), inflammatory diseases58 and neurodegenerativediseases.63–65 Therefore, the NLRP3 inflammasome might be a potential target to treat these diseases.
Until now, at least five NLRP3 inflammasome inhibitors have been tested in human clinical trials.66 OLT1177 (34 in Fig. 3) is an orally available and specific NLRP3 inflammasome inhibitor by directlybinding to NLRP3.67 In phase I study, OLT1177 was determined to be safe and well-tolerated in healthy volunteers receiving oral doses up to 1000 mg/day for 8 days. OLT1177 also showed a meaningful exposurewith a long half-life. Currently, it is undergoing a phase II clinical study for the treatment of acute gouty arthritis.68 NT-0167 (structure undis- closed) is under phase I clinical study to evaluate safety, tolerability,pharmacokinetics and pharmacodynamics responses. IFM-2427 wasdeveloped by IFM Tre and later acquired by Novartis. As an inhibitor of NLRP3 by directly binding to NLRP3 protein,69 IFM-2427 is now un- dergoing a phase I clinical study for autoimmune diseases. Recently,
Novartis has advanced IFM-2427 (renamed as DFV890) into phase IIor therapeutic effects on several disease models such as gouty arthritis, cryopyrin-associated autoinflammatory syndromes, and T2D.
Recently, Xu and co-workers reported a novel tetrahydroquinoline, compound 37, as a specific NLRP3 inflammasome inhibitor without affecting NLRC4 or AIM2 inflammasomes.97 The results from a biolayer interferometry (BLI) assay indicated that compound 37 directly boundto the NACHT domain of NLRP3 and inhibited NLRP3 ATPase activity. It showed an IC50 of 7.8 μM on IL-1β release in LPS/ATP-stimulated THP-M cells. Compound 37 alleviated the shortening of colon length and dose- dependently decreased the secretion of IL-1β in a dextran sulfate sodium (DSS)-induced colitis mouse model.study for COVID-19-related pneumonia.70 SomaliX and Inzomelid
Bharate and co-workers reported their discovery of a computation-(structures undisclosed) were developed by Inflazome, but were recentlyacquired by Roche.71–76 As a potent, selective, orally available and peripherally-restricted NLRP3 inflammasome inhibitor for inflamma-tory diseases, SomaliX showed excellent safety, tolerability and phar- macokinetics in healthy subjects.77 This compound is ready to enter intophase II study. Inzomelid, an orally available, brain-penetrant inhibitor used in the treatment of debilitating inflammatory diseases of the brain, showed excellent safety, tolerability and pharmacokinetic properties in healthy subjects in the completed phase I study, and also deliveredpreliminary positive results in a patient with cryopyrin-associated pe- riodic syndrome (CAPS).78,79
Structure-diverse NLRP3 inflammasome inhibitors have been re- ported (Fig. 3), including Glyburide (26),80 JC-121 (27),81 JC-171(28),82 MCC950 (29),83–85 MNS (30),86 NBC-6 (31),87 BOT-4-one(32)88 and Oridonin (33)89. Recent review articles17,66,90–94 have nicely summarized theNLRP3 activation mechanism, inhibitors and related indications. The following section focuses on non-natural se- lective NLRP3 inflammasome inhibitors that have been reported since 2018 and which have MoA of inhibiting ATPase activity, suppressing caspase-1 activation and blocking recruitment of ASC (Table 2, Fig. 4). Discovered by Deng group through modification of a HTS hit, the 4-oXo-2-thioXo-thiazolidinone derivative CY-09 (35) is a potent and se- lective inhibitor of NLRP3.95 It showed inhibitory activity with an IC50 of 2.40 0.10 μM on IL-1β release in LPS-primed bone marrow derived macrophages (BMDMs) under the stimulation of nigericin. It also dose- dependently inhibited IL-1β secretion in freshly isolated synovial fluid cells (SFCs) from patients with gout.104 CY-09 showed specific inhibitionon NLRP3 by directly binding to NACHT and inhibiting ATPase activity without affecting NLRC4 and AIM2 inflammasomes.
Tranilast (36), a previously approved anti-allergic drug, was found tospecifically suppress NLRP3 inflammasome activation without affecting NLRC4 or AIM2 inflammasomes.96 Mechanistic study indicated that Tranilast directly binds to the NACHT domain and blocks NLRP3 olig-omerization. In a dose-dependent manner, Tranilast inhibited caspase-1 activation and IL-1β production at 50 and 100 μM, respectively, in SFCs from patients with gout. Importantly, it showed remarkable preventiveally designed compound 38 by using a NLRP3 homology model.98 The inhibition IC50 of compound 38 was 5 μM on IL-1β release in ATP- stimulated J774A.1 cells. Molecular modeling indicated that com- pound 38 occupies the ATP-binding site of NLRP3 protein, the same site as MCC950 with additional cation-π interaction with Leu 171 contrib- uted by the nitro-group.
Sharma and co-workers discovered compound 39 to be a highly potent, orally bioavailable and selective NLRP3 inflammasome inhibitor by optimizing the sulfonylurea moiety of MCC950 (clogP = 3.27).99
Compound 39 has an IC50 of 6.8 ± 0.5 nM on IL-1β release in THP1 cells.
In comparison to MCC950 (IC50 of 8 0.7 nM), compound 39 has equivalent potency and lower lipophilicity (clogP 2.45). It specifically blocked recruitment of ASC during NLRP3 activation without affecting the AIM2 inflammasome. In addition, it decreased NLRP3 dependent IL- 1β release by 44% at 10 mg/kg oral dosing in LPS/ATP challenged mice.
The Zhang group reported the identification of JC-124 (40) as a se- lective NLRP3 inhibitor by inhibiting the activation of caspase-1.100 Thein vivo efficacy was demonstrated in three models: a transgenic mouse AD model by reducing AD pathology at 50 mg/kg/day;81 an AMI model by limiting the infarct size and protecting cardiac functions at 30 mg/kg;100 and a traumatic brain injury (TBI) model by decreasing inflam-matory response in an injured brain and cortical lesion volume after TBI in adult male Sprague-Dawley rats via multiple i.p. dosing.105 Encour- aged by the promising pharmacodynamics effect of JC-124, Zhang’s group further optimized the benzenesulfoamide scaffold and identified a series of JCP-124 close analogues, among which, YQ128 (41) showed an
IC50 of 0.30 ± 0.01 μM on IL-1β release in J774A.1. In addition, YQ-128 dose-dependently suppressed IL-1β release in peritoneal macrophages upon LPS/ATP challenge with an IC50 of 1.59 0.60 μM. YQ128 spe-cifically inhibited NLRP3 inflammasome without affecting NLRC4 or AIM2 inflammasome. However, pharmacokinetic studies in rats showed an extensive clearance and tissue distribution, and low oral bioavail- ability (%F 10), likely due to poor absorption and high first-pass metabolism.
The natural product parthenolide (PTL) exhibits anti-inflammatory activity, and has been reported to target the NLRP3 inflammasome;106

DNA-cGAS interaction modulators
The anti-malarial drugs hydroXychloroquine (HCQ) and quinacrine have been identified as inhibiting cGAS activity and IFN-β secretion through blocking the formation of DNA-cGAS complex.129,143 Elkon’s group further reported a series of quinacrine derivatives,144 amongwhich X6 (45) showed modest potency with an IC50 of 14 μM in THP-1cells. In Trex1—/— mice, X6 reduced cGAMP production in the heart and attenuated the expression of ISGs in the spleen and heart. In addition, X6reduced ISG expression in the PBMCs of SLE patients. Suramin (46), a drug used to treat river blindness and African sleeping sickness, was recently identified as a cGAS inhibitor through the screening of a focused library. Mechanistically, suramin displaced the bound DNA from cGAS. It dose-dependently inhibited cGAMP production and fully reduced IFN-β expression in THP-1 cells at 5 μM. Li and coworkers un- covered that acetylation at one of the three lysine residues of cGAS,K384, K394 and K414, impaired cGAS’s binding to dsDNA, therebysuppressing its enzymatic function.145 Importantly, they identified thatcatalytic domain.The binding of the initial virtual screening hits wasthe widely used drug, aspirin (47), directly acetylated those lysine res- idues and efficiently inhibited cGAS-mediated immune responses. More interestingly, they further demonstrated that aspirin significantlyreduced the ISGs in the PBMC of AGS patients and attenuated self-DNA- induced autoimmunity in the Trex1—/— mice (See Fig. 5).

cGAS dimerization modulator
Yin’s lab recently reported a new class of h-cGAS inhibitors that target the dimeric interface, presumably, to disrupt cGAS dimeriza- tion.146 A virtual screening was conducted to target the key residues Lys335 (human Lys347) and Lys382 (human Lys394) crucial for cGASoligomerization139 based on the reported mouse cGAS structure. Z918(48) was identified as a weakly active h-cGAS inhibitor. Subsequent medicinal chemistry optimization produced CU-76 (49), which inhibi- ted cGAS activity in both ATP consumption assay with an IC50 of 0.24 µM and in THP-1 cells (See Fig. 6).

cGAS active site inhibitor
PF-06928215142 and RU.521147 are the first reported cGAS in- hibitors targeting h-cGAS and mouse cGAS (m-cGAS) active sites, respectively. They usually occupy the cGAMP pocket, and picked up key stacking interactions with Agr376 and Try436. The Tuschl grouprecently reported the discovery of h-cGAS inhibitors by using a high throughput ATP-coupled luminescence-based assay.148 Medicinalchemistry optimization of two initial hits led to the identification of more potent compounds such as J014 (50) and G150 (51) with IC50 values of 0.100 µM and 0.0102 µM, respectively. J014 was active in THP-1 cells with an IC50 value of 2.63 µM, but was deprioritized due to cytotoXicity and poor selectivity. G150 showed a higher activity in THP- 1 cells with an IC50 value of 1.96 µM and good activity in primary human macrophages (IC50 less than 1 µM). The crystal structure of G150 bound to the h-cGAS apo catalytic domain demonstrated its localization in the cGAMP pocket. Zhao and coworkers reported the discovery of novel cGAS active site inhibitors by utilizing a virtual screening approachconfirmed by thermal shift assay (TSA), and the enzymatic activity was evaluated in a PPiase-coupled assay, identifying compound 52 as a moderately effective cGAS inhibitor with an IC50 of 29.88 3.20 µM. Subsequent similarity research into compound 52, followed by a second virtual screening, led to the identification of more potent compounds S2(53) and S3 (54) with IC50 values of 13.1 0.09 µM and 4.9 0.26 µM, respectively. The crystal structure of compound S2 bound to the h-cGAS catalytic domain was resolved (PDB code: 6LRL). A Bellbrook Labspatent disclosed a series of tricyclic benzofuropyrimidine analogues as cGAS inhibitors.150 The co-structure of compound 55 indicated that the tricyclic core sat in the middle of Arg376 and Try436. Compound 56 showed the best cellular potency with an IC50 of 13 µM in an IFN-β ELISA assay using THP-1 cells (See Fig. 7).

cGAS inhibitor with undisclosed mechanism
Aduro Biotech recently disclosed multiple series of cGAS inhib- itors.151–153 C1089 (57) with a pyrazolopyrimidinone core was active ina cGAS biochemical assay and in THP-1 cells with an IC50 value less than 20 µM.151 It also showed inhibitory activity against cytokines such as RANTES and monocyte chemoattractant protein-1 (MCP-1) secretion inTrex1—/— BMDM with IC50s of 1.251 µM and 6.973 µM, respectively. Triazine compounds were disclosed in another patent application.
Compound TA1065 (58) was the most potent one in THP-1 cells with an IC50 value between 20 and 100 µM.152 In addition, new compounds with an imidazopyridazinone core have been recently disclosed.153 Com- pound 59 is a representative showing an IC50 of less than 1 µM in the cGAS biochemcial assay and an IC50 of less than 10 µM in THP-1 cells. Bellbrook Labs disclosed another patent application on their cGAS in- hibitors with an undisclosed binding MoA.154 Compound 60 inhibited cGAS enzymatic activity with an IC50 of 0.391 µM, and was weakly active in THP-1 cells (See Fig. 8).

Targeting the STING
STING is an endoplasmic-reticulum protein111 consisting of an N-terminal domain (~140 aa) that contains four transmembrane helices, and a cytoplasmic C-terminal domain that includes a globular ligand binding domain (aa 153 – 340) and a C-terminal tail (aa 342 – 379). The cytoplasmic parts of STING form a butterfly-shaped dimer with the ligand binding pocket located at the dimer interface. Crystal structures of the cytosolic domain of human STING in both apo and in complexwith natural and synthetic ligands have been determined to be highly structured dimers.155–162
All the reported STING antagonists are at the discovery stage. In terms of the binding MoA, two types of STING antagonists have beenreported, including competitive binding (Fig. 9) and allosteric binding (Fig. 10). The STING competitive pocket is large and polar with highaffinity endogenous ligand cGAMP (Kd 4.6 nM),163 and as such, it would be challenging to develop a potent and orally available small molecule antagonist.164 Alternatively, inhibition of the agonist-induced,post-translational palmitoylation by covalently modifying Cys88/91 at an allosteric pocket could inhibit STING signaling,165 presumably, by blocking STING polymerization. This new MoA provides a novelapproach to identify STING signaling inhibitors.166
Merck scientists have reported the discovery of orally availableSTING antagonists that bind to the STING competitive binding site.164 To leverage the intrinsic symmetric nature of STING, the team focused on identifying small molecules that can bind to the STING homodimer ina 2:1 ratio while maintaining physicochemical properties suitable for oral drugs. Compound 61 was firstly identified as a weak hit (IC50 = 7.3 µM in HAQ STING cGAMP displacement assay), and its crystal structure indicated a 2:1 binding mode with intermolecular interaction of the twoligands. Further medicinal chemistry optimization led to the discovery of compound 62 with improved binding affinity (IC50 of 0.068 µM). Compound 62 inhibited the IFN-β production with an IC50 of 11 µM in cGAMP stimulated THP-1 cells. In addition, compound 62 demonstrated good oral bioavailability (%F 60) in a rat pharmacokinetics study.
Astin C (63), a cyclopeptide isolated from the medicinal plant Astertataricus, was identified as a STING antagonist by Wang lab through a screening of a Compositae-type cyclopeptide library.167 It was found tospecifically bind to the STING C-terminal domain by utilizing a biotin pull-down assay. Further in silico virtual simulation analysis suggested that astin C may block the cGAMP binding pocket. The binding affinity of astin C (Kd 53 nM) is comparable to that of cGAMP in an isothermal titration calorimetry (ITC) assay. In addition, astin C showed good ac- tivity in inhibiting IFN-β production in human fetal lung fibroblasts withan IC50 of 10.83 1.88 µM. Moreover, astin C significantly attenuated the IFN-β and a set of pro-inflammatory cytokines in Trex1—/— BMDMcells and mice.
GlaxoSmithKline disclosed a series of N-methyl amidobenzimida- zoles STING antagonists in a recent patent application,168 derived from a previously reported agonist scaffold that occupies the STING competi-tive pocket at the dimeric interface.160 Some antagonists exhibitedexcellent potency in both biochemical and cellular assays. For example, compound 64 showed excellent binding affinity with a pIC50 value of >9.9 in a displacement assay, and strongly inhibited IFN-β secretion inboth THP-1 cells and human PBMCs with pIC50 values of 8.9 and 7.1, respectively.
Ablasser’s group reported the identification of covalent STING an-tagonists from a cell-based chemical screening.166 The identified nitro- furan analogues, as exemplified by C-178 (65), covalently modified the transmembrane Cys91 through a nucleophilic addition of the thiol to the 4-position of the furan ring followed by an intramolecular rearrange- ment. C-178 potently and selectively inhibited STING activation inmouse BMDMs. A more soluble close analogue, C-176 (66), suppressedIFN-β production in agonist challenged mice and in Trex1—/— mice. In addition, C-176 attenuated various markers of systemic inflammation inTrex1—/— mice by means of chronic treatment. However, both C-176 and C-178 are mouse specific STING antagonists, with a following additionalchemical screen revealing a human STING antagonist H-151 (67), which similarly inhibited human STING at submicromolar level through co- valent modification of Cys91. H-151 doesn’t contain a typical cysteine reactive scaffold, and covalent product was observed in a mass spec- trometry study; however, the reaction mechanism of covalent bond formation has not been explained. H-151 exhibited notable efficacy in agonist-challenged mice and in the bioluminescent IFN-β reporterTrex1—/— mice. Notably, another two research groups have discovered that endogenous ligands such as nitro-fatty acid (68)169 and 4-HNE (69)170 inhibit STING activation in a similar fashion by forming a co-valent bond with Cys91/Cys88 and Cys88 respectively through Michael addition.
In 2020, IFM Therapeutics released a series of patent applications to protect their STING antagonists,171–178 with the clinical trial of the lead molecule expected to be conducted in 2021.179 Their antagonists seemto have been derived from the pharmacophore of H-151, containing a(n) (un)substituted (aza)indole connecting with a hydrophobic scaffold through a linker such as urea, amide, aminosulfoXimine or 3,4-diamino- cyclobut-3-ene-1,2-dione, the exact MoA of which has not been dis-closed. The compounds showed very good potency in THP-1 cells as exemplified by compounds 70,172 71,173 72,174 and 73175 with IC50s lessthan 1.0 μM (See Fig. 11).

Concluding remarks and future prospects
Unlike the various kinase inhibitors, as outlined in the first part of the review,180 which treat autoimmune and inflammatory diseases fromdownstream of either innate or adaptive immune signaling, the nucleic acid sensing antagonists/inhibitors address the diseases from the initi- ation of the pathogenic signaling, which might bring differentiated therapeutic benefit. Meanwhile, the discovery and development of nucleic acid sensing antagonists face several challenges. Firstly, auto- immune and inflammatory diseases, in particular SLE, are very hetero- geneous diseases, thus selecting the right patients is critical for clinical proof-of-concept. Secondly, the pathogenic nucleic acids may activate multiple pathways; so, to achieve satisfactory efficacy may require blocking several nucleic acid sensors simultaneously, which may not be technically achievable by a single molecule. In addition, the technical challenge of obtaining orally available small molecule antagonists/in-hibitors for certain targets is high. For example, the cGAS-STING pathway has fostered enthusiasm since the identification of cGAS141; however, progress has been relatively slow, which might be related tothe poor druggability of the protein pocket. Despite these obstacles, several small molecule antagonists/inhibitors of nucleic acid sensors such as TLR78 and NLRP3 have been advanced into clinical trials as noted beforehand. The cGAS-STING antagonists are approaching the entry into clinical development. In addition, a growing understanding of the structural basis of nucleic acid sensing will continue to accelerate the discovery process. Moreover, new technologies such as targeted protein degradation and RNA-targeted small molecules provide alternative ap- proaches to inhibiting nucleic acid sensing. Richer small molecule nucleic acid sensing antagonist/inhibitor pipelines are expected to be seen in both preclinical and clinical stages in the coming years. More importantly, the clinical outcomes of the pilot molecules will further guide preclinical compound optimization and design of clinical studies.

References
1 Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007;449(7164):819–826.
2 Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol. 2015;33:257–290.
3 Okude H, Ori D, Kawai T. Signaling through nucleic acid sensors and their roles in inflammatory diseases. Front Immunol. 2020;11, 625833.
4 Tan X, Sun L, Chen J, Chen ZJ. Detection of microbial infections through innate immune sensing of nucleic acids. Annu Rev Microbiol. 2018;72:447–478.
5 Matz KM, Guzman RM, Goodman AG. The role of nucleic acid sensing in controlling microbial and autoimmune disorders. Int Rev Cell Mol Biol. 2019;345:35–136.
6 Chen G, Shaw MH, Kim YG, Nunez G. NOD-like receptors: role in innate immunity and inflammatory disease. Annu Rev Pathol. 2009;4:365–398.
7 Xiao TS. The nucleic acid-sensing inflammasomes. Immunol Rev. 2015;265(1): 103–111.
8 Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005;23:307–336.
9 Egli A, Santer DM, O’Shea D, Tyrrell DL, Houghton M. The impact of the interferon- lambda family on the innate and adaptive immune response to viral infections. Emerg Microbes Infect. 2014;3(7), e51.
10 Junt T, Barchet W. Translating nucleic acid-sensing pathways into therapies. Nat Rev Immunol. 2015;15(9):529–544.
11 Hawkins LD. LR7, TLR8, and TLR9 antagonists: potential therapies for autoimmune diseases 2020. Medicinal Chemistry Reviews. 2020;55(9).
12 Zhu G, Xu Y, Cen X, Nandakumar KS, Liu S, Cheng K. Targeting pattern-recognition receptors to discover new small molecule immune modulators. Eur J Med Chem. 2018;144:82–92.
13 Zhang H, You QD, Xu XL. Targeting stimulator of interferon genes (STING): a medicinal chemistry perspective. J Med Chem. 2020;63(8):3785–3816.
14 Cui X, Zhang R, Cen S, Zhou J. STING modulators: predictive significance in drug discovery. Eur J Med Chem. 2019;182, 111591.
15 Sintim HO, Mikek CG, Wang M, Sooreshjani MA. Interrupting cyclic dinucleotide- cGAS-STING axis with small molecules. Medchemcomm. 2019;10(12):1999–2023.
16 Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128.
17 Zhang X, Xu A, Lv J, et al. Development of small molecule inhibitors targeting NLRP3 inflammasome pathway for inflammatory diseases. Eur J Med Chem. 2020; 185, 111822.
18 Anwar MA, Shah M, Kim J, Choi S. Recent clinical trends in Toll-like receptor targeting therapeutics. Med Res Rev. 2019;39(3):1053–1090.
19 Duffy L, O’Reilly SC. Toll-like receptors in the pathogenesis of autoimmunediseases: recent and emerging translational developments. Immunotargets Ther. 2016;5:69–80.
20 Rahman AH, Eisenberg RA. The role of toll-like receptors in systemic lupus erythematosus. Springer Semin Immun. 2006;28(2):131–143.
21 Spachidou MP, Bourazopoulou E, Maratheftis CI, et al. EXpression of functional Toll-like receptors by salivary gland epithelial cells: increased mRNA expression in
22 Federico S, Pozzetti L, Papa A, et al. Modulation of the innate immune response by targeting toll-like receptors: a perspective on their agonists and antagonists. J Med Chem. 2020;63(22):13466–13513.
23 O’Neill LAJ, Bryant CE, Doyle SL. Therapeutic targeting of toll-like receptors forinfectious and inflammatory diseases and cancer. Pharmacol Rev. 2009;61(2): 177–197.
24 Wang YB, Zhang ST, Li HY, et al. Small-molecule modulators of toll-like receptors.Accounts Chem Res. 2020;53(5):1046–1055.
25 Patinote C, Karroum NB, Moarbess G, et al. Agonist and antagonist ligands of toll- like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem. 2020;193, 112238.
26 Tojo S, Zhang ZK, Matsui H, et al. Structural analysis reveals TLR7 dynamics underlying antagonism. Nat Commun. 2020;11(1):5204.
27 Zhang ST, Hu Z, Tanji H, et al. Small-molecule inhibition of TLR8 through stabilization of its resting state. Nat Chem Biol. 2018;14(1):58–64.
28 Mussari CP, Dodd DS, Sreekantha RK, et al. Discovery of potent and orally bioavailable small molecule antagonists of toll-like receptors 7/8/9 (TLR7/8/9).Acs Med Chem Lett. 2020;11(9):1751–1758.
29 For M5049: https://clinicaltrials.gov/ct2/results?cond &term M5049&cntry &state &city &dist ; For BMS986256: https:// clinicaltrials.gov/ct2/results?cond &term BMS-986256 &cntry &state &city &dist ; For E6742: https://clinicaltrials.gov/ct2/results? cond &term E6742&cntry &state &city &dist ; For CPG52364: https:// clinicaltrials.gov/ct2/results?cond &term CPG52364&cntry &state &city &dist .
30 Vlach J, Bender AT, Przetak M, et al. Discovery of M5049: a novel selective TLR7/8 inhibitor for treatment of autoimmunity. J Pharmacol Exp Ther. 2021;376:397–409.
31 Dyckman AJ, Dodd DS, Haque TS, Lombardo LJ, Macor JE, Mussari CP, Pasunoori L, Ratna Kumar S, Sherwood TC, Posy SL, Sistla RK, Hegde S, Ramachandra A. [1,2,4]triazolo[1,5-a]pyridinyl substituted indole compounds. WO 2018/005586 A1. 2018.
32 Lipford GB, Zepp CM. Quinazoline derivative useful as toll-like receptor antagonist. WO 2008/152471 A1. 2008. Lipford GB, Zepp CM. Quinazoline derivative useful as toll-like receptor antagonist. WO 2008/152471 A1. 2008.
33 Karroum NB, Moarbess G, Guichou JF, et al. Novel and Selective TLR7 Antagonists among the Imidazo[1,2-a]pyrazines, Imidazo[1,5-a]quinoXalines, and Pyrazolo[1,5-a]quinoXalines Series. J Med Chem. 2019;62(15):7015–7031.
34 Mukherjee A, Raychaudhuri D, Sinha BP, et al. A chemical switch for transforming a purine agonist for toll-like receptor 7 to a clinically relevant antagonist. J Med Chem. 2020;63(9):4776–4789.
35 Jiang SS, Tanji H, Yin K, et al. Rationally designed small-molecule inhibitors targeting an unconventional pocket on the tlr8 protein-protein interface. J Med Chem. 2020;63(8):4117–4132.
36 Padilla-Salinas R, Anderson R, Sakaniwa K, et al. Discovery of novel small molecule dual inhibitors targeting toll-like receptors 7 and 8. J Med Chem. 2019;62(22): 10221–10244.
37 Paul B, Rahaman O, Roy S, et al. Activity-guided development of potent and selective toll-like receptor 9 antagonists. Eur J Med Chem. 2018;159:187–205.
38 Talukdar A, Ganguly D, Mukherjee A, Paul B, Rahaman O, Kundu B, Roy S, DeblinaR. Purine based compounds as toll-like receptor 9 antagonist. WO2019092739. Council of Scientific and Industry Research. 2019.
39 Knoepfel T, Nimsgern P, Jacquier S, et al. Target-based identification and optimization of 5-Indazol-5-yl pyridones as toll-like receptor 7 and 8 antagonistsusing a biochemical TLR8 antagonist competition assay. J Med Chem. 2020;63(15): 8276–8295.
40 Alpera PB, Deanea J, Betschartb C, et al. Discovery of potent, orally bioavailable in vivo efficacious antagonists of the TLR7/8 pathway. Bioorg Med Chem Lett. 2020;30 (17), 127366.
41 Dey F, Hu T, Liu H, Shen H, Zhang Z, Zhu W. Novel pyrazolopyridine compounds for the treatment of autoimmune disease. WO2020048596. F Hoffmann-La Roche AG. 2020.
42 Liu H, Wu G, Zhang W, Zhu W, Shen H, Dey F. Novel pyrrolidinyl amide compounds for the treatment of autoimmune disease. WO2020043271. F Hoffmann-La Roche AG. 2020.
43 Alper P, Deane J, Jiang S, Jiang T, Knoepfel T, Michellys P, Y. Mutnick D, Pei W, Syka P, Zhang G, Zhang Y. Compounds and compositions as inhibitors of endosomal toll-like receptors. WO2018047081. Navartis AG. 2018.
44 Dey F, Shen H, Xu H, Yun H, Zou G, Zhu W. Tetrahydro-1H-pyrazino[2,1-a] isoindolylquinoline compounds for the treatment of autoimmune disease. WO2019233941. F Hoffmann-La Roche AG. 2019.
45 Liu H, Shen H, Zhu W, Hu T, Zhang Z, Zhang Z, Dey F, Wang X. Pyridinyl heterocyclyl compounds for the treatment of autoimmune disease. WO2019238629. F Hoffmann-La Roche AG. 2019.
46 Patra MC, Achek A, Kim GY, et al. A novel small-molecule inhibitor of endosomal TLRs reduces inflammation and alleviates autoimmune disease symptoms in murine models. Cells-Basel. 2020;9(7):1648.
47 Hu ZY, Tanji H, Jiang SS, et al. Small-molecule TLR8 antagonists via structure- based rational design. Cell Chem Biol. 2018;25(10):1286–1291.
48 Shukla NM, Kimbrell MR, Malladi SS, David SA. Regioisomerism-dependent TLR7 agonism and antagonism in an imidazoquinoline. Bioorg Med Chem Lett. 2009;19 (8):2211–2214.
49 Shukla NM, Malladi SS, Day V, David SA. Preliminary evaluation of a 3H imidazoquinoline library as dual TLR7/TLR8 antagonists. Bioorgan Med Chem. 2011;19(12):3801–3811.
50 Shukla NM, Mutz CA, Malladi SS, Warshakoon HJ, Balakrishna R, David SA. Toll- like receptor (TLR)-7 and -8 modulatory activities of dimeric imidazoquinolines.J Med Chem. 2012;55(3):1106–1116.
51 Dey F, Liu H, Shen H, Wu G, Zhang W, Zhu W. Novel pyrrolidine amine compounds for the treatment of autoimmune disease. WO2020048605. F Hoffmann-La Roche AG. 2020.
52 Novel morpholinyl amine compounds for the treatment of autoimmune disease. EP3623369. F Hoffmann-La Roche AG. 2020.
53 Zhu W, Zou G, Dey F. Benzothiazole compounds for the treatment of autoimmune diseases. WO2020048583. F Hoffmann-La Roche AG. 2020.
54 Dey F, Qiu Z, Zhu W, Zou G. Novel piperazine compounds for the treatment of autoimmune disease. WO2020020800. F Hoffmann-La Roche AG. 2020.
55 Qiu Z, Shen H, Zhu W, Dey F, Zou G, Xu H. Novel cyclic amidine compounds for the treatment of autoimmune disease. WO2020048595. F Hoffmann-La Roche AG. 2020.
56 Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–687.
57 Lamkanfi M, Kanneganti TD. Nlrp3: an immune sensor of cellular stress and infection. Int J Biochem Cell Biol. 2010;42(6):792–795.
58 Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD, Latz E. Targeting theNLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discov. 2018;17(8): 588–606.
59 Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489.
60 Lieberman J, Wu H, Kagan JC. Gasdermin D activity in inflammation and host defense. Sci Immunol. 2019;4(39), eaav1447.
61 Shen HH, Yang YX, Meng X, et al. NLRP3: a promising therapeutic target for autoimmune diseases. Autoimmun Rev. 2018;17(7):694–702.
62 Shao BZ, Wang SL, Pan P, et al. Targeting NLRP3 inflammasome in inflammatory bowel disease: putting out the fire of inflammation. Inflammation. 2019;42(4): 1147–1159.
63 Ising C, Venegas C, Zhang S, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575(7784):669–673.
64 Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer’s diseaseand contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–678.
65 Gordon R, Albornoz EA, Christie DC, et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci Transl Med. 2018;10(465). eaah4066.
66 Schwaid AG, Spencer KB. Strategies for targeting the NLRP3 inflammasome in the clinical and preclinical space. J Med Chem. 2021;64(1):101–122.
67 Marchetti C, Swartzwelter B, Gamboni F, et al. OLT1177, a beta-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses themetabolic cost of inflammation. PNAS. 2018;115(7):E1530–E1539.
68 Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15(4):203–214.
69 https://www.ifmthera.com/news/ifm-therapeutics-announces-dosing-of-first- subjects-in-phase-1-healthy-volunteer-study-of-lead-nlrp3-antagonist/.
70 https://www.clinicaltrialsregister.eu/ctr-search/search?query IFM-2427.
71 O’Nell L, Coll R, Cooper M, Robertson A, Schroder K. Preparation of sulfonylureas and related compounds as inhibitors of NLRP3 inflammasome activation for the treatment of diseases WO2016131098. The university of Queensland; 2016.
72 Miller D, Macleod A, Del RG, Stratford SA. Preparation of a sodium salt of N- ((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-isopropyl-1H-pyrazole-3- sulfonamide WO2019206871. Inflazome Limited. 2019.
73 Schwizer D, Breeger S, Thom S, Alanine T. Processes for preparing N- ((1,2,3,4,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-isopropyl-1H-pyrazole-3- sulfonamide WO2020079207. Inflazome Limited. 2020.
74 Cooper M, Miller D, Macleod A, Shannon J, Thom S, Strutt I, Castagna D, Carrillo AJ, Van Wiltenburg J. Pyrazolylsulfonylureas and related compounds as NLRP3 inhibitors and their preparation WO2020104657. Inflazome Limited. 2020.
75 Cooper M, Miller D, Macleod A, Thom S, Shannon J, St-Gallay S. Preparation of sulfonylureas and sulfonylthioureas as NLRP3 inhibitors. WO2019034693. Inflazome Limited. 2019.
76 Cooper M, Miller D, Macleod A,Van Wiltenburg J, Thom S, St-Gallay S, Shannon J. Novel sulfonamidocarboXamide compounds as NLRP3 inhibitors and their preparation WO2019008025. Inflazome Limited. 2019.
77 https://www.biospace.com/article/releases/inflazome-s-somaliX-demonstrates-p ositive-safety-tolerability-and-pharmacodynamic-profile-in-its-phase-i-study/.
78 https://clinicaltrials.gov/ct2/show/NCT04015076?term=Inflazome&draw=2&r ank=3.
79 https://www.businesswire.com/news/home/20200326005102/en/Inzomelid-completes-Phase-I-studies-and-shows-positive-results-in-the-treatment-of-Cryop yrin-Associated-Periodic-Syndrome-CAPS.
80 Lamkanfi M, Mueller JL, Vitari AC, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009;187(1):61–70.
81 Marchetti C, Chojnacki J, Toldo S, et al. A novel pharmacologic inhibitor of theNLRP3 inflammasome limits myocardial injury after ischemia-reperfusion in the mouse. J Cardiovasc Pharmacol. 2014;63(4):316–322.
82 Guo C, Fulp JW, Jiang Y, et al. Development and characterization of a hydroXyl- sulfonamide analogue, 5-Chloro-N-[2-(4-hydroXysulfamoyl-phenyl)-ethyl]-2- methoXy-benzamide, as a novel NLRP3 inflammasome inhibitor for potentialtreatment of multiple sclerosis. ACS Chem Neurosci. 2017;8(10):2194–2201.
83 Coll RC, Robertson AA, Chae JJ, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3): 248–255.
84 Salla M, Butler MS, Pelingon R, et al. Identification, synthesis, and biological evaluation of the major human metabolite of NLRP3 inflammasome inhibitorMCC950. ACS Med Chem Lett. 2016;7(12):1034–1038.
85 Coll RC, Hill JR, Day CJ, et al. MCC950 directly targets the NLRP3 ATP-hydrolysis motif for inflammasome inhibition. Nat Chem Biol. 2019;15(6):556–559.
86 He Y, Varadarajan S, Munoz-Planillo R, Burberry A, Nakamura Y, Nunez G. 3,4- methylenedioXy-beta-nitrostyrene inhibits NLRP3 inflammasome activation byblocking assembly of the inflammasome. J Biol Chem. 2014;289(2):1142–1150.
87 Baldwin AG, Rivers-Auty J, Daniels MJD, White CS, Schwalbe CH, Schilling T, Hammadi H, Jaiyong P, Spencer NG, England H, Luheshi NM, Kadirvel M, Lawrence CB, Rothwell NJ, Harte MK, Bryce RA, Allan SM, Eder C, Freeman S, Brough D. Boron-Based Inhibitors of the NLRP3 Inflammasome. Cell Chem Biol.2017;24(11):1321–1335. e5.
88 Shim DW, Shin WY, Yu SH, et al. BOT-4-one attenuates NLRP3 inflammasome activation: NLRP3 alkylation leading to the regulation of its ATPase activity and ubiquitination. Sci Rep. 2017;7(1):15020.
89 He H, Jiang H, Chen Y, et al. Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat Commun. 2018;9(1):2550.
90 Zahid A, Li B, Kombe AJK, Jin T, Tao J. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol. 2019;10:2538.
91 Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20(13):3328.
92 Wang Z, Hu W, Lu C, et al. Targeting NLRP3 (nucleotide-binding domain, leucine- rich-containing family, pyrin domain-containing-3) inflammasome incardiovascular disorders. Arterioscler Thromb Vasc Biol. 2018;38(12):2765–2779.
93 Weber ANR, Bittner ZA, Shankar S, Liu X, Chang TH, Jin T, Tapia-Abell´an A. Recent insights into the regulatory networks of NLRP3 inflammasome activation. J Cell Sci. 2020;133(23):jcs248344.
94 Bertinaria M, Gastaldi S, Marini E, Giorgis M. Development of covalent NLRP3 inflammasome inhibitors: chemistry and biological activity. Arch Biochem Biophys. 2019;670:116–139.
95 Chen Y, He H, Jiang H, et al. Discovery and optimization of 4-oXo-2-thioXo-thia- zolidinones as NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inhibitors. Bioorg Med Chem Lett. 2020;30(7), 127021.
96 Huang Y, Jiang H, Chen Y, et al. Tranilast directly targets NLRP3 to treat inflammasome-driven diseases. EMBO Mol Med. 2018;10(4), e8689.
97 Dai Z, Chen XY, An LY, et al. Development of novel tetrahydroquinoline inhibitors of NLRP3 inflammasome for potential treatment of DSS-induced mouse colitis.J Med Chem. 2021;64(1):871–889.
98 Abdullaha M, Mohammed S, Ali M, Kumar A, Vishwakarma RA, Bharate SB. Discovery of quinazolin-4(3 H)-ones as NLRP3 inflammasome inhibitors:
computational design, metal-free synthesis, and in vitro biological evaluation. J Org Chem. 2019;84(9):5129–5140.
99 Agarwal S, Sasane S, Shah HA, et al. Discovery of N-Cyano-sulfoXimineurea derivatives as potent and orally bioavailable NLRP3 Inflammasome inhibitors. ACS Med Chem Lett. 2020;11(4):414–418.
100 Fulp J, He L, Toldo S, et al. Structural insights of benzenesulfonamide analogues as NLRP3 inflammasome inhibitors: design, synthesis, and biological characterization.J Med Chem. 2018;61(12):5412–5423.
101 Jiang Y, He L, Green J, et al. Discovery of second-generation NLRP3 inflammasome inhibitors: design, synthesis, and biological characterization. J Med Chem. 2019;62 (21):9718–9731.
102 Ou Y, Sun P, Wu N, et al. Synthesis and biological evaluation of parthenolide derivatives with reduced toXicity as potential inhibitors of the NLRP3 inflammasome. Bioorg Med Chem Lett. 2020;30(17), 127399.
103 Harrison D, Boutard N, Brzozka K, et al. Discovery of a series of ester-substituted NLRP3 inflammasome inhibitors. Bioorg Med Chem Lett. 2020;30(23), 127560.
104 Jiang H, He H, Chen Y, et al. Identification of a selective and direct NLRP3 inhibitor to treat inflammatory disorders. J Exp Med. 2017;214(11):3219–3238.
105 Kuwar R, Rolfe A, Di L, et al. A novel small molecular NLRP3 inflammasome inhibitor alleviates neuroinflammatory response following traumatic brain injury. J Neuroinflammation. 2019;16(1):81.
106 Juliana C, Fernandes-Alnemri T, Wu J, et al. Anti-inflammatory compounds parthenolide and Bay 11–7082 are direct inhibitors of the inflammasome. J Biol Chem. 2010;285(13):9792–9802.
107 Lesiak K, Koprowska K, Zalesna I, Nejc D, Duchler M, Czyz M. Parthenolide, a sesquiterpene lactone from the medical herb feverfew, shows anticancer activityagainst human melanoma cells in vitro. Melanoma Res. 2010;20(1):21–34.
108 Civril F, Deimling T, de Oliveira Mann CC, et al. Structural mechanism of cytosolic DNA sensing by cGAS. Nature. 2013;498(7454):332–337.
109 Gao P, Ascano M, Wu Y, et al. Cyclic [G(2’,5’)pA(3’,5’)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell. 2013;153 (5):1094–1107.
110 Wu J, Sun L, Chen X, et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science. 2013;339(6121):826–830.
111 Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455(7213):674–678.
112 Hopfner KP, Hornung V. Molecular mechanisms and cellular functions of cGAS- STING signalling. Nat Rev Mol Cell Biol. 2020;21(9):501–521.
113 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(6121):786–791.
114 Ablasser A, Chen ZJ. cGAS in action: expanding roles in immunity and inflammation. Science. 2019;363(6431).
115 Ahn J, Xia T, Konno H, Konno K, Ruiz P, Barber GN. Inflammation-driven carcinogenesis is mediated through STING. Nat Commun. 2014;5:5166.
116 Kerur N, Fukuda S, Banerjee D, et al. cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat Med. 2018;24(1):50–61.
117 Cao DJ, Schiattarella GG, Villalobos E, et al. Cytosolic DNA sensing promotes macrophage transformation and governs myocardial ischemic injury. Circulation. 2018;137(24):2613–2634.
118 King KR, Aguirre AD, Ye YX, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017;23(12):1481–1487.
119 Zeng L, Kang R, Zhu S, et al. ALK is a therapeutic target for lethal sepsis. Sci Transl Med. 2017;9(412).
120 Yu Y, Liu Y, An W, Song J, Zhang Y, Zhao X. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J Clin Invest. 2019;129(2):546–555.
121 Sliter DA, Martinez J, Hao L, et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature. 2018;561(7722):258–262.
122 McCauley ME, O’Rourke JG, Yanez A, et al. C9orf72 in myeloid cells suppressesSTING-induced inflammation. Nature. 2020;585(7823):9–101.
123 Yu CH, Davidson S, Harapas CR, et al. TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell. 2020;183(3):636–649 e618.
124 Pang W, Hu F. Cellular and physiological functions of C9ORF72 and implications for ALS/FTD. J Neurochem. 2020.
125 Lee JD, Woodruff TM. TDP-43 Puts the STING in ALS. Trends Neurosci. 2020.
126 An J, Durcan L, Karr RM, et al. EXpression of cyclic GMP-AMP synthase in patients with systemic lupus erythematosus. Arthritis Rheumatol. 2017;69(4):800–807.
127 Papinska J, Bagavant H, Gmyrek GB, et al. Activation of stimulator of interferon genes (STING) and sjogren syndrome. J Dent Res. 2018;97(8):893–900.
128 Gall A, Treuting P, Elkon KB, et al. Autoimmunity initiates in nonhematopoieticcells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity. 2012;36(1):120–131.
129 An J, Woodward JJ, Sasaki T, Minie M, Elkon KB. Cutting edge: antimalarial drugs inhibit IFN-beta production through blockade of cyclic GMP-AMP synthase-DNAinteraction. J Immunol. 2015;194(9):4089–4093.
130 Mackenzie KJ, Carroll P, Lettice L, et al. Ribonuclease H2 mutations induce a cGAS/ STING-dependent innate immune response. EMBO J. 2016;35(8):831–844.
131 Gao D, Li T, Li XD, et al. Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. PNAS. 2015;112(42):E5699–5705.
132 Liu Y, Jesus AA, Marrero B, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371(6):507–518.
133 Jeremiah N, Neven B, Gentili M, et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. J Clin Invest. 2014;124(12):5516–5520.
134 Kumar V. A STING to inflammation and autoimmunity. J Leukoc Biol. 2019;106(1): 171–185.
135 Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17(10):1142–1149.
136 Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet. 2019;20(11):657–674.
137 Jiang M, Chen P, Wang L, et al. cGAS-STING, an important pathway in cancer immunotherapy. J Hematol Oncol. 2020;13(1):81–91.
138 Ding C, Song Z, Shen A, Chen T, Zhang A. Small molecules targeting the innate immune cGAS-STING-TBK1 signaling pathway. Acta Pharm Sin B. 2020;10(12): 2272–2298.
139 Zhang X, Wu J, Du F, et al. The cytosolic DNA sensor cGAS forms an oligomericcomplex with DNA and undergoes switch-like conformational changes in the activation loop. Cell Rep. 2014;6(3):421–430.
140 Li X, Shu C, Yi G, et al. Cyclic GMP-AMP synthase is activated by double-stranded DNA-induced oligomerization. Immunity. 2013;39(6):1019–1031.
141 Sheridan C. Drug developers switch gears to inhibit STING. Nat Biotechnol. 2019;37 (3):199–201.
142 Hall J, Brault A, Vincent F, et al. Discovery of PF-06928215 as a high affinity inhibitor of cGAS enabled by a novel fluorescence polarization assay. PLoS ONE. 2017;12(9), e0184843.
143 Bose D, Su Y, Marcus A, Raulet DH, Hammond MC. An RNA-Based FluorescentBiosensor for High-Throughput Analysis of the cGAS-cGAMP-STING Pathway. Cell Chem Biol. 2016;23(12):1539–549.
144 An J, Woodward JJ, Lai W, et al. Inhibition of Cyclic GMP-AMP Synthase Using a Novel Antimalarial Drug Derivative in Trex1-Deficient Mice. Arthritis Rheumatol. 2018;70(11):1807–1819.
145 Dai J, Huang YJ, He X, et al. Acetylation Blocks cGAS Activity and Inhibits Self- DNA-Induced Autoimmunity. Cell. 2019;176(6):1447–1460 e1414.
146 Padilla-Salinas R, Sun L, Anderson R, et al. Discovery of small-molecule cyclic GMP- AMP synthase inhibitors. J Org Chem. 2020;85(3):1579–1600.
147 Vincent J, Adura C, Gao P, et al. Small molecule inhibition of cGAS reduces interferon expression in primary macrophages from autoimmune mice. Nat Commun. 2017;8(1):750–762.
148 Lama L, Adura C, Xie W, et al. Development of human cGAS-specific small-molecule inhibitors for repression of dsDNA-triggered interferon expression. Nat Commun. 2019;10(1):2261–2274.
149 Zhao W, Xiong M, Yuan X, Li M, Sun H, Xu Y. In silico screening-based discovery of novel inhibitors of human cyclic GMP-AMP synthase: a cross-validation study of molecular docking and experimental testing. J Chem Inf Model. 2020;60(6):3265–3276.
150 Lowery RG, Kumar M, BoXer M, Maloney D, Boyd S. Inhibitors of cGAS activity as therapeutic agents. WO2020142729. BellBrook Labs. 2020.
151 Ndubaku CO, Katibah GE, Roberts TC, et al. Preparation of pyrazolopyrimidinone compounds as inhibitors of the cGAS/STING pathway and of the cellular cytokine secretion and their uses in the treatment of autoimmune, inflammatory and neurodegenerative disorders. WO2019055750. Aduro Biotech Inc. 2019.
152 Katibah GE, Kim JY, Ndubaku CO, Roberts TC, Tjandra M. Triazine compounds as cGAS inhibitor and their preparation. WO2019245910. Aduro Biotech Inc. 2019.
153 Ciblat S, Johnson T, Latimer BK, Ndubaku CO, Ramtohul YK, Roberts TC. Imidazopyridazinone compounds and uses thereof. WO2020123422. Aduro BioTech Inc. 2020.
154 Lowery RG, Kumar M, BoXer M, Maloney D, Boyd S. Inhibitors of cGAS activity as therapeutic agents. WO2020142735. BellBrook Labs. 2020.
155 Huang YH, Liu XY, Du XX, Jiang ZF, Su XD. The structural basis for the sensing and binding of cyclic di-GMP by STING. Nat Struct Mol Biol. 2012;19(7):728–730.
156 Ouyang S, Song X, Wang Y, et al. Structural analysis of the STING adaptor protein reveals a hydrophobic dimer interface and mode of cyclic di-GMP binding.Immunity. 2012;36(6):1073–1086.
157 Shang G, Zhu D, Li N, et al. Crystal structures of STING protein reveal basis for recognition of cyclic di-GMP. Nat Struct Mol Biol. 2012;19(7):725–727.
158 Shu C, Yi G, Watts T, Kao CC, Li P. Structure of STING bound to cyclic di-GMP reveals the mechanism of cyclic dinucleotide recognition by the immune system.Nat Struct Mol Biol. 2012;19(7):722–724.
159 Yin Q, Tian Y, Kabaleeswaran V, et al. Cyclic di-GMP sensing via the innate immune signaling protein STING. Mol Cell. 2012;46(6):735–745.
160 Ramanjulu JM, Pesiridis GS, Yang J, et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564(7736):439–443.
161 Pan BS, Perera SA, PiesvauX JA, Presland JP, Schroeder GK, Cumming JN, Trotter BW, Altman MD, Buevich AV, Cash B, Cemerski S, Chang W, Chen Y, Dandliker PJ, Feng G, Haidle A, Henderson T, Jewell J, Kariv I, Knemeyer I, Kopinja J, Lacey BM,Laskey J, Lesburg CA, Liang R, Long BJ, Lu M, Ma Y, Minnihan EC, O’Donnell G,Otte R, Price L, Rakhilina L, Sauvagnat B, Sharma S, Tyagarajan S, Woo H, Wyss DF, Xu S, Bennett DJ, Addona GH. An orally available non-nucleotide STING agonist with antitumor activity. Science. 2020;369(6506):eaba6098.
162 Chin EN, Yu C, Vartabedian VF, et al. Antitumor activity of a systemic STING- activating non-nucleotide cGAMP mimetic. Science. 2020;369(6506):993–999.
163 Zhang X, Shi H, Wu J, et al. Cyclic GMP-AMP containing miXed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol Cell. 2013;51(2): 226–235.
164 Siu T, Altman MD, Baltus GA, et al. Discovery of a novel cGAMP competitive ligand of the inactive form of STING. ACS Med Chem Lett. 2019;10(1):92–97.
165 Mukai K, Konno H, Akiba T, et al. Activation of STING requires palmitoylation at the Golgi. Nat Commun. 2016;7:11932–11941.
166 Haag SM, Gulen MF, Reymond L, et al. Targeting STING with covalent small- molecule inhibitors. Nature. 2018;559(7713):269–273.
167 Li S, Hong Z, Wang Z, et al. The Cyclopeptide astin c specifically inhibits the innate immune CDN Sensor STING. Cell Rep. 2018;25(12):3405–3421 e3407.
168 Fosbenner DT, Graybill TL, Kang J, et al. Preparation of heterocyclic amides as modulators of stimulator of interferon genes (STING). WO2019069270. GlaxoSmithKline Intellectual Property Development Limited. 2019.
169 Hansen AL, Buchan GJ, Ruhl M, et al. Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling.PNAS. 2018;115(33):E7768–E7775.
170 Jia M, Qin D, Zhao C, et al. RedoX homeostasis maintained by GPX4 facilitates STING activation. Nat Immunol. 2020;21(7):727–735.
171 Roush WR, Venkatraman S, Glick G, Seidel HM. Compounds and compositions for treating conditions such as cancer associated with STING activity. WO2020010092. IFM Due Inc. 2020.
172 Seidel HM, Roush WR, Venkatraman S. Compounds and compositions for treating conditions associated with STING activity such as cancer using STING inhibitors. WO2020150417. IFM Due Inc. 2020.
173 Venkatraman S, Roush WR, Seidel HM. Sulfonimidamide compounds and compositions for treating conditions associated with STING activity and their preparation. WO2020106736. IFM Due Inc. 2020.
174 Venkatraman S, Katz J, Roush WR. N-heteroaryl-squaramide compounds for treating conditions associated with STING activity WO2020236586. IFM Due Inc. 2020.
175 Katz J, Venkatraman S, Rouse WR. Preparation of substituted ureas for treating conditions associated with STING activity WO2020252240. IFM Due Inc. 2020.
176 Roush WR, Seidel HM, Venkatraman S. Aromatic compounds and compositions for treating conditions associated with STING activity and their preparation. WO2020106741. IFM Due Inc. 2020.
177 Roush WR, Venkatraman S, Click G, Seidel HM. Preparation of IRAK4-IN-4 heteroaryl aryl ureas for treating conditions associated with STING activity WO2020010155. IFM Due Inc. 2020.
178 Venkatraman S, Katz J, Roush WR. Compounds and compositions for treating conditions associated with STING activity WO2020243519. IFM Due Inc. 2020.
179 https://www.genengnews.com/news/novartis-gains-option-to-acquire-second- ifm-subsidiary-in-up-to-840m-cgas-sting-collaboration/.
180 Zheng J, Wu J, Ding X, Shen HC, Zou G. Small molecule approaches to treat autoimmune and inflammatory diseases (Part I): Kinase inhibitors. Bioorg Med Chem Lett. 2021;38:127862–127875.