Journal Pre-proofs Design, synthesis and biological evaluation of Acridone Analogues as novel STING receptor agonists Shi Hou, Xiu-juan Lan, Wei Li, Xin-lin Yan, Jia-jia Chang, Xiao-hong Yang, Wei Sun, Jun-hai Xiao, Song Li PII: DOI: Reference:
S0045-2068(19)31757-2 https://doi.org/10.1016/j.bioorg.2019.103556 YBIOO 103556
To appear in:
Bioorganic Chemistry
Received Date: Revised Date: Accepted Date:
18 October 2019 23 December 2019 24 December 2019
Please cite this article as: S. Hou, X-j. Lan, W. Li, X-l. Yan, J-j. Chang, X-h. Yang, W. Sun, J-h. Xiao, S. Li, Design, synthesis and biological evaluation of Acridone Analogues as novel STING receptor agonists, Bioorganic Chemistry (2019), doi: https://doi.org/10.1016/j.bioorg.2019.103556
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2019 Published by Elsevier Inc.
Design, synthesis and biological evaluation of Acridone Analogues as novel STING receptor agonists
Shi Hou a,b,c, Xiu-juan Lan a,b,c, Wei Li b,c, Xin-lin Yan b,c, Jia-jia Chang b,c,d , Xiao-hong Yang a,*, Wei Sun a,*, Jun-hai Xiao b,c,*, Song Li b,c
a
School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China
b
National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China c
State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China d School
of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China
* Corresponding author
Xiao-hong Yang: School of Pharmaceutical Sciences, Jilin University, China; E-mail:
[email protected]
Wei Sun: School of Pharmaceutical Sciences, Jilin University, China; E-mail:
[email protected]
Jun-hai Xiao: National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, China; E-mail:
[email protected]
ABSTRACT STING (Stimulator of Interferon Genes) has become a focal point in immunology research and a target in drug discovery. The discovery of a potent human-STING agonist is expected to revolutionize current anti-virus or cancer immunotherapy. Inspired by the structure and function of murine STING-specific agonists (DMXAA and CMA), we rationally designed and synthesized four series of novel compounds for the enhancement of human sensitivity. In the cell-based assay, we identified six compounds from all the synthetic small molecules: 2g, 9g, and 12b are STING agonists that are efficacious across species, and all have the skeleton of acridone; 1b, 1c, and 12c just function in the murine STING pathway. Notably, 12b exhibits the best activity among the six agonists, and its inductions of both human and murine STING-dependent signalling are similar to that of
2’3’-cGAMP, which is a well-known STING inducer. While a protein assay indicated that 2g, 9g, and 12b could activate the pathway by directly binding human STING, 12b also displayed the strongest binding affinity. Additionally, our studies show that 12b can induce faster, more powerful, and more durable responses of assorted cytokines in a native system than 2’3’-cGAMP. Consequently, our team is the first to successfully modify murine STING agonists to obtain human sensitivity, and these results suggest that 12b is a potent direct-human-STING agonist. Additionally, the acridone analogues demonstrate tremendous potential in the treatment of tumours or viral infections. Keywords: STING; human agonists; direct bonding; induction of cytokines
1.
Introduction STING is both a pattern-recognition receptor (PRR), which directly binds cyclic dinucleotides
(CDNs) via its C-terminal domain [1], and an adaptor molecule, which can be activated by multiple cytoplasmic receptors of dsDNA [2]. STING plays a vital role in the activation of innate immune responses. Accumulated evidence has demonstrated that the cyclic GMP-AMP synthase (cGAS) is the major dsDNA cytosolic sensor allowing STING to function [3, 4]. The essential mechanism for innate immune sensing of various pathogens and tumours exists in the cGAS–STING pathway in which cGAS is activated by cytoplasmic DNA to subsequently catalyse the production of cyclic GMP-AMP (cGAMP). 2’3’-cGAMP is a non-canonical CDN that binds to STING and induces it to oligomerize [5], leading to a downstream signalling cascade including recruiting the serine/threonine-protein kinase (TBK1), phosphorylating the interferon regulatory transcription factor (IRF3) and nuclear factor-κB (NF-κB), and producing cytokines and proteins such as the type I interferons (IFNs), interleukin 6 (IL-6), and tumour necrosis factor α (TNFα) [6]. IFN-β is the signature cytokine induced by the host STING pathway, and it primes anticancer activities via diversified approach including cross-presentation of tumour antigens and mobilization of tumour-specific CD8 T cells [7]. Therefore, STING is regarded as a promising target in various fields, including immunization, autoinflammation, and cancer therapeutics [8]. To the best of our knowledge, current efforts regarding STING agonists are centred on chemically modified CDNs for improving the polarity and liability of its analogues [9]; some of these analogues strongly and
continually degrade tumours in syngeneic tumour models through intratumoural delivery and are currently in clinical trials [10]. However, CDNs analogues also exhibit the limitations of low cell-membrane permeability [11]. Substantial studies to identify more drug-like small molecule STING agonists have been conducted. According to species selectivity, we classify non-nucleotide STING agonists into three categories: murine-specific, human-specific, and nonspecific agonists. Since the studies of the Fitzgerald research team [12] and the Mitchison group [13] were reported, the species-specific of STING
has
been
attracting
great
interest.
Both
of
these
groups
considered
that
5′,6′-dimethylxanthenone-4-acetic acid (DMXAA) is a potent agonist of murine STING (mSTING) rather than human STING (hSTING). These studies clearly explained why DMXAA failed in human phase III trials against non-small-cell lung cancer (NSCLC). The structure and mechanism of 10-carboxymethyl-9-acridanone (CMA) are similar those of to DMXAA [14]. To the best of our knowledge, five chemotypes of compounds have been identified as the human STING-specific agonist [15-19]. Except for a dietary xanthone α-mangostin that directly binds and activates human STING, other small molecules share the following properties: indirect STING agonists cannot bind to STING, identification of chemical compounds by cell-based high-throughput screening (HTS), comprehensive and diverse bioactivities were lower than that of 2’3’-cGAMP in vitro, and the existence of unknown difficulties in animal experiments. In 2018, 44 scientists jointly published a new study in Nature. These scientists used STING competition binding assays to screen small molecules and found an aminobenzimidazole (ABZI) compound and its dimer (di-ABZIs) derivatives as potent and nonspecific STING agonists [20]. Although di-ABZIs have shown pre-clinical promise as STING agonists, the relatively large molecular weight and polarity, which are similar to those of 2’3’-cGAMP, probably limit the application for intratumoural injection. Analysis of various STING agonists above, the further effort should be focused on the identification of small molecules STING agonists that are efficacious across species. Furthermore, indistinct mechanisms of the STING pathway may be elucidated by them as tool molecules, and their application in animals enables to facilitate the assessment of safety.
2. Results and Discussion
2.1 Design and Synthesis of xanthenone-4-acetic acid(XAA)and acridone Analogs Undeniably, murine STING activators DMXAA and CMA have some inherent advantages: 1) They are small molecular compounds, which can mitigate the defects of CDNs [21]; 2) Both of them can bind to murine STING protein, and have confirmed crystal structures [14, 22]; 3) The seminal contributions of Patel and co-workers, which includes subtle mutations in protein sequences of STING now able to endow human STING with DMXAA sensitivity [23], inspire us to begin our study. Hence, our innovations of molecule design originate from comprehensively analysing the similarities of DMXAA and CMA, especially the relationship between molecules and proteins (Figure 1A-C).
Figure 1. Intermolecular contacts in the complex of small molecules bound to STING with the representation of hydrogen bonding as the dotted line. (A) CMA bound to murine STING [14]; (B) DMXAA bound to murine STING [22]; (C) DMXAA bound to human STING
S162A/Q266I
[23]; (D) The corresponding location between
DMXAA and mutant residues.
The following information has been revealed: maintaining the structure of three rings guarantees the interaction between two small molecules; the 5,6-dimethyl substitutes of DMXAA are highly significant since it can produce a strong hydrophobic effect with the surrounding amino acids (Figure 1D); the mutant substitutions (S162A and Q266I) of hSTING at the region of binding-pocket correspond to the positions of DMXAA at C1/C2 and C7 (Figure 1D). Previous work synthesized C7-position DMXAA derivatives that contain a hydrogen bonding donor and acceptor or a halide, but none of the compounds can bind to hSTING [24]. Thus, we designed and synthesized four types of small molecules of a relevance skeleton whose changes concentrated on assorted groups at C1/C2 and C7 positions of compounds. The structures and synthetic methods of design molecules are summarized in Figure 2 and Scheme 1–4.
Figure 2. The structures of design molecules
The DMXAA synthetic protocol could be applied to our synthesis of series Ⅰ (Scheme 1) [25].
Cross-coupling between the sodium phenolate 5a, 5b and potassium salt of Iodide 6a, 6b followed by a Friedel–Crafts acylation-type reaction produced DMXAA and other XAAs (1a–1d). The synthetic route for designed compounds 2a–2o (series Ⅱ) is outlined in Scheme 2 [26]. Thus, Ullmann condensation between phenylacetic acid derivative 7a–7e and 5-substituted-2-amino-3,4dimethyl-benzoic acid 8a–8c in the presence of Cu powder, Cu2O and K2CO3 in DMF, and synthetic compounds of benzene acetic acid were treated with Eaton’s reagent to afford the cyclized intermediate products. End-products were obtained by opening five-membered rings under alkaline condition. In Scheme 3, we show the synthesis of a series of multi-site-substituted acridones (series Ⅲ) [27]. First, acridone analogous 12a–12f could be accessed using Scheme 2 synthetic procedures. Subsequent ester compounds 13a, 13b were synthesized by coupling ethyl bromoacetate [28], and we prepared the final compounds 3a, 3b by hydrolysis and acidification. Ullmann’s reaction and Friedel-Crafts acylation were also carried out in Scheme 4 to give another kind of acridone derivative (series Ⅳ): 2-methoxy-4-carboxylic acid acridone 4. The structures of the compounds were listed in Table 1 and confirmed on the basis of MS and NMR (1H and
13C)
spectral data.
However, the number of molecules was significantly limited by the precise modification sites and the commercial availability of the origin of benzene derivatives. O
KOOC
R1
I
ONa
R1
a,b
COONa R3
5a,5b
O
R3
R2
R2
6a,6b
1a-1d
COOH
Scheme 1. The synthetic routes of 1a-1d (series Ⅰ). (a) DMSO, CuCl, TDA-1, 95°C; (b) H2SO4, H2O (9:1), 80°C. R1
O HOOC
R2
R3
COOH
R1
a,b
7a-7e
c
R2 N H 2a-2o
9a-9o
O
8a-8c
R1
R3
N
H 2N
Br
O R2
R3
COOH
Scheme 2. The synthetic routes of 2a–2o (series Ⅱ). (a) DMF, Cu, Cu 2 O, K2CO3, 100-120 °C; (b) Eaton’s reagent, 80-100 °C; (c) EtOH, H2O, NaOH, reflux.
R1
R4
COOH Br 10a,10b
R3
H 2N R2 11a-11c
a,b
R1
R4 R3 R2
N H 12a-12f
O
O
O c
R4
R1
R3
R1
R3
N R2
d
R4
O O 13a,13b
N OH
R2 O 3a,3b
Scheme 3. The synthetic routes of 3a, 3b (series Ⅲ). (a) DMF, Cu, Cu 2 O, K2CO3, 100-120 °C; (b) Eaton’s reagent, 80-100 °C; (c) BrCH2 COOC2H5, NaH, r.t.; (d) NaOH, 2 M HCl.
COOH
O
Br 10b
O
HOOC
a,b
H 2N 8a
O N H
COOH
4
Scheme 4. The synthetic routes of 4 (series Ⅳ). (a) DMF, Cu, Cu 2 O, K2CO3, 100-120 °C; (b) Eaton’s reagent, 80-100 °C.
2.2
In Vitro Screening of STING Agonists and the Structure−Activity Relationship (SAR) analysis. To thoroughly evaluate the four kinds of analogues, we elaborately selected three cell lines in
which the STING gene has been transfected or knocked out: 293T-hSTING-R232 cells, 293T-mSTING cells, and THP1-KO-STING cells. STING is either deactivated, undetectable, or not expressed in certain cell lines, such as human embryonic kidney cells (HEK293) [29]. However, 293T-STING cells are a family of reporter cells designed to study variants of STING. Recent work revealed that the sequence differences among these variants can markedly affect STING function and, consequently, impact human health. Our choice of 232R-RGR (71R-230G-293R) is the most prevalent variant known as the wild-type STING gene and accounts for approximately 70% of the human population [30]. Therefore, human or murine specific STING agonist stimulation can be respectively assessed in 293T-hSTING-R232 cells or 293T-mSTING cells by monitoring IFN-stimulated response elements and (ISRE)-induced secreted embryonic alkaline phosphatase (SEAP) production. Then, we employed THP1-KO-STING cells to confirm whether the compound exhibits the function of STING-dependent cytokine induction. The screening process was divided into two steps. First, we used three STING cell lines to screen all targeted compounds and their synthetic precursors at two concentrations (100 μM, 20 μM) to quickly identify small molecules that induced I IFN via the STING pathway. The structures of the screening compounds were displayed in Figure 3, and the first screened results were listed in Table 1.
O
O R1
R3
1a-1d
R3
N H 2a-2o
COOH
O
R1
R3 R2
O 9g
COOH
O
R4 N
N H 4
COOH 3a,3b
O
O
O
N R2
COOH
O
O R1
R2
R3
O R2
O
R1
N
N H
O
12a-12f
13b O
Figure 3. The structure of screening small molecules
Table 1 Structures and Biological grades of the XAA and acridone Analogues of the Preliminary Screening Compound
R1
R2
R3
R4
Human-STINGa
Murine-STINGb
1a(DMXAA)
H
CH3
CH3
-
-
++
1b
Br
CH3
CH3
-
-
+
1c
H
H
OCH3
-
-
+
1d
Br
H
OCH3
-
-
-
2a
H
H
H
-
-
-
2b
F
H
H
-
-
-
2c
Cl
H
H
-
-
-
2d
H
F
H
-
-
-
2e
H
Cl
H
-
-
-
2f
H
H
Br
-
-
-
2g
H
H
OCH3
-
+
+
2h
F
H
Br
-
-
-
2i
Cl
H
Br
-
-
-
2j
H
F
Br
-
-
-
2k
H
Cl
Br
-
-
-
2l
F
H
OCH3
-
-
-
2m
Cl
H
OCH3
-
-
-
2n
H
F
OCH3
-
-
-
2o
H
Cl
OCH3
-
-
-
3a(CMA)
H
H
H
-
-
+
3b
OCH3
CH3
CH3
-
-
-
4
-
-
-
-
-
-
9g
H
H
OCH3
-
+
+
12a
H
H
H
H
-
-
12b
OCH3
CH3
CH3
H
++
++
12c
OCH3
H
H
H
-
+
12d
H
CH3
CH3
H
-
-
12e
H
CH3
CH3
OCH3
-
-
12f
OCH3
CH3
CH3
OCH3
-
-
13b
OCH3
CH3
CH3
H
-
-
Measuring the OD (optical density) value of a small molecule at two concentrations (100 μM, 20 μM) and comparing with 2’3’-cGAMP (30 μg/ml), we classified the activity of small molecules into three grades: “++”, excellent (the OD value of the compound at 20 μM is approximately the 2’3’-cGAMP); “+”, good (the OD value of the compound at 100 μM is approximately the 2’3’-cGAMP); “-”, inactive (the OD value of the compound at 100 μM is much lower than the 2’3’-cGAMP). aEach result does three independent experiments in 293T-hSTING-R232 cells. bEach result does three independent experiments in 293T-mSTING cells.
Secondly, we used differentiated concentration gradients to test and verify the intensity of screened small molecules and compared their activities to that of the reference STING agonists for murine (DMXAA) and human (2’3’-cGAMP). The function of agonists in STING-dependent cytokine induction has been confirmed by using THP1-KO-STING cells. Together, six potent compounds were obtained from the various synthetic precursors and their derivatives by two-step selection. Three were non-species-specific agonists (2g, 9g, and 12b), and none of these compounds exhibited any IRF or NF-κB activities in the knockout hSTING cells, while the other compounds (1b, 1c, and 12c) just activated the murine STING pathway (Figure 4 and Table 2).
O
O
O
Br
O
HOOC
O
O
1b
1c
HOOC
O
N H 12c
O
O
O
O
O
O N H
N O 9g
N H 12b
COOH
2g
Figure 4. The structures of screened agonists.
Table 2 Biological Activities of the screened agonists of the Second Screening and of DMXAA and 2’3’-cGAMP a, b
Compound
Human-STING EC50 (μM)a
Murine-STING
Emax (%)b
EC50 (μM)a
Emax (%)b
1b
nt
nt
152.91±6.6
63.15±6.5
1c
nt
nt
115.59±6.6
69.64±3.4
2g
154.21±9.6
63.8±8.8
66.45±10.5
78.32±5.6
9g
134.59±12.1
70.3±8.5
72.33±11.1
83.39±4.1
12b
7.45±2.7
109±19.7
10.23±0.6
125.91±20.4
12c
nt
nt
134.62±5.9
60.45±5.3
DMXAA(1a)
nt
nt
8.87±1.1
118.27±15.8
CMA(3a)
nt
nt
96.21±6.6
82.39±10.4
2’3’-cGAMP
7.23±2.6
100
7.01±2.1
100
a
Each value is the mean ± SD from three independent experiments at least. b Emax values are reported as a mean percentage ± SD relative to the corresponding 2’3’-cGAMP response from three independent experiments at least.
In our study, the refinements of DMXAA also proved to be unsuccessful. Synthesized small molecules not only fail to trigger human STING pathway induction but also negatively respond to the mouse pathway with expanding or changing substituents in DMXAA. Fortunately, some of the acridone analogues we discovered were powerful and helpful, especially compound 12b, which displayed approximately similar activities to 2’3’-cGAMP at activating both pathways (human: EC50=7.45 μM, murine: EC50=10.23 μM). In acridone analogues, we mainly investigated the effects of the different substitutions at various
positions and explored and obtained some preliminary SARs as much as possible. All the screened human-STING agonists, which are typical acridone structures, consist of two benzene rings fused together with a keto-group and a nitrogen atom at the 9th and 10th position, exhibit the common substitutes of 5,6-dimethyl and verify the rationality of our plan. For the series Ⅱ compounds, we found that the modifications of the C1/C2 position were ineffective. But, 2g and 9g (the series Ⅱ compounds of C7-modification) shown better activities than CMA in murine-STING derived cells, and had STING stimulating activities in human-STING derived cells. It indicated that the methoxyl at the C7-position was crucial for inducing the STING pathway. By analysis of series Ⅲ and Ⅳ compounds, we discovered that their intermediates without carboxylic acid structure are active. Specifically, the synthetic precursors 12b differs from compounds 3b and 4 in the absence of corresponding carboxylic acids at N or C-4 position, and the addition of the substituent enormously shrank the activity of the molecules. Meanwhile, we compared 12b, 12c, and 12d noticed that the importance of the C-2-methoxyl is the key group in small molecules to the activation of the STING pathway. The structure of 5,6-dimethyl is to endow acridone compounds with the sensitivity of human STING (Figure 5).
Figure 5. Summarized SARs of the acridone analogues.
Interestingly, compound 12e, which maintains the structure of 5,6-dimethyl-acridone and only substitutes methoxyl at the C-7 position, is inactive and indicates that the C-7-methoxyl may work in coordination with groups of other positions (maybe at N and C-4). The active response of acridones increases when C-2 or C-7 is substituted by methoxyl; however, the disappearance in the induction of type I IFNs after two sites are replaced together (reference compound 12f). Accordingly, for the future design of acridone agonists, we need to maintain the structure of 12b and decorate the positions of N, C-4 and C-7 with suitable groups to strengthen the activity in vitro.
2.3
The Acridone Agonists Directly Bind and Stabilize the hSTING Protein Next, we explored the reason why acridones agonists vigorously stimulated the STING pathway
and further evaluated the biological activity of them. To verify whether acridones agonists directly activate the STING-TBK1-IRF3 pathway through STING, we first used human STING binding kits provided by Cisbio. Using a homogeneous time-resolved fluorescence (HTRF) competition format, which uses a STING ligand-d2, a 6His tagged human STING
WT
protein, and an anti-6His
Cryptate-labelled antibody, STING binders compete with the STING ligand-d2, thus preventing fluorescence resonance energy transfer (FRET) from occurring. The competitive binding data identified 12b (IC50=1.92 μM) as exhibiting the strongest ligand-human STING interaction among all our synthetic agonists. The IC50 value of 2’3’-cGAMP was 4.6 nM in agreement with the reference values provided by kits Instructions(2’3’-cGAMP Standard IC50=5 nM), and DMXAA (purple rhombuses, IC50 > 500 μM), as a negative control drug, does not compete with the binding of STING ligand-d2 to the human STING protein as expected (Figure 6A). To further cross-validate the direct binding of 12b with hSTING CTD, we conducted a universal and authoritative method called isothermal titration calorimetry (ITC) to measure the Kd value of 12b. The results of ITC were consistent with that of the binding kit, and the Kd values of 2’3’-cGAMP and 12b were 0.02 μM and 26.4 μM, respectively (Figure 6B-D).
Figure 6. The acridone agonists bind and stabilize the hSTING CTD. (A) STING competition binding assay. Dose-response curves of 2’3’-cGAMP (n = 3), 12b (n = 4), 2g (n = 3), 9g(n=3), DMXAA(n=3), Error bars represent SD of independent experiments. Relative potency of compounds was calculated using GraphPad Prism software, Mean response ± SD. The interaction of hSTINGWT CTD with buffer (B), compound 12b (C) and 2’3’-cGAMP (D) was quantified using isothermal titration calorimetry (ITC).
The great differences in binding affinity between 12b and 2'3'-cGAMP could be explained by their crystal structures of binding STING [31]: 2’3’-cGAMP bound to the symmetrical dimer of human STING by a single molecule (Figure 7A), but there are two molecules of DMXAA in their complex with the dimer of murine STING (Figure 7B). The A rings of the two DMXAA molecules overlapped each other (the distance was 3.9 Å) to form a π – π stacking interaction that kept the
structure of STING dimer (Figure 7C). For the weak interaction of the π – π stacking interaction, the binding force of DMXAA was much lower than that of 2’3’-cGAMP. Hornung research group also reported that the acridone ring moieties of both CMAs partially stack to each other in a parallel (~ 4 Å distance) [14]. Therefore, we speculated that the mechanism of compound 12b is the same as that of DMXAA and CMA.
Figure 7. The binding mechanisms of STING agonists. (A) The 2.25 Å crystal structure of 2’3’-cGAMP bound to hSTING
H232
(PDB code: 4LOH); (B) The 2.43 Å crystal structure of two molecules of DMXAA bound to
mSTING R231 (PDB code: 4LOL); (C) The distance between the A rings of the two DMXAA molecules (PDB code: 4LOL).
Similarly, the Kd values of natural STING-ligands 2'3'-cGAMP and 3'2'-cGAMP are nearly 1000 times different, but their activation levels of IFN-β are equipotent in the micro molar range[3]. Furthermore, the binding affinity is not necessarily related to the activation of small molecules in the STING pathway, and small molecules with high affinity may be the STING inhibitors. Significantly, different types of agonists have different binding mechanisms, the mechanism of compound 12b needs to be further determined. Furthermore, we have successfully demonstrated that acridone
agonists can directly bind to human STING. The binding affinity of acridone derivatives are exactly proportional to cellular effects in vitro, and this reveals that the point of design ideas about modification of this kind of derivatives can be used to enhance the affinity between small molecules and STING C-terminal domain (CTD).
2.4
Identification of an Acridone Compound That Potently Activates the cGAS−Human STING
Pathway To confirm the functional consequence of binding, we employed real-time PCR (qPCR) to examine the effects of the best compound 12b on proinflammatory-cytokine gene expression in a native system (THP-1 cells), using well-known STING binders 2’3’-cGAMP as a control. Firstly, we recognized the fitting concentration of 12b (40 μM) through preliminary experiments. Then, the experiments of time gradient (co-cultivation time) were used to further weigh the competence of two agonists in secreting cytokines (Figure 8). Interestingly, the advantage of the 2’3’-cGAMP lied only in the expression of IP-10, which is an interferon-stimulating gene, while the maximum expression of other immune factors induced by 12b were greater than that induced by 2’3’-cGAMP, especially TNF-α. Considering the impact of time, the mRNA level of most cytokines was enhanced with time by 12b and 2’3’-cGAMP, except for the induction of IFN-β by 12b, which showed a slight downward trend.
Figure 8. 2’3’-cGAMP and 12b induces diverse cytokines production in a native system. THP1 cells were stimulated with 21 μM 2’3’-cGAMP or 40 μM 12b. mRNA level of IFNβ (A), TNFα (B), IL-6 (C) and IP-10(D) were measured at 8h, 20h and 30h by real-time PCR and normalized by GAPDH expression. Error bars represent SEM of independent experiments (n=3). Significance (* P < 0.05, ** P < 0.01, *** P < 0.001) was determined by t-test relative to DMSO at a particular time.
By comprehensive analysis, we observed that 12b and 2’3’-cGAMP generally exhibited potent action in excreting cytokines. Although the former gave a more fast, powerful, and durable induction, the results can be explained by the different mechanisms between 12b and 2’3’-cGAMP. STING agonists can activate two pathways: the STING-IRF and the STING-NF-κB. STING-IRF3 pathway mainly induces the production of type Ⅰ interferons, and the pro-inflammatory cytokines are secreted by the STING-NF-κB pathway. The production of TNF-α is the marker in NF-κB-pathway [32]. We boldly presumed that 12b can induce both STING pathways, and the activation of NF-κB-pathway is more active than 2’3’-cGAMP.
3. Conclusions The past few years have witnessed the rapid advance of the novel STING agonists as anti-tumour, anti-virus, and immunostimulatory agents. However, the best agonists of CDNs
analogues were also confined to poor permeability of cell membranes in intra-tumoural drug delivery of clinical trials [11]. The present research aimed to discover novel STING agonists that may result in more drug-like properties and lead to a breakthrough at species-specificity. A promising thought is to modify the mouse-specific STING agonist DMXAA, but no derivatives have been identified in binding human STING to date [24]. To overcome these challenges, we summarized the general character of mouse-specific STING agonists (DMXAA and CMA), rationally designed and synthesized various novel XAA and acridone derivatives, and investigated the biological activity of them. Notably, compound 12b is a non-species-specific STING agonist with an acridone skeleton, exhibiting the most potent activity with the induction of type I interferons among all synthetic compounds. The levels of the activation of STING signalling in vitro are similar to those of the 2’3’-cGAMP. Additionally, some information provided by comprehensive and detailed SARs implied that removing any substituents of 12b could weaken its activity. Therefore, we need to preserve the structure and change other sites for improvement of the effects. We further demonstrated that 12b directly binds human STING. The binding capability between the acridone analogues and STING is tightly associated with its activity in vitro. In our study of the mechanism, showing its ability to lead to vigorous activation in both STING pathways of IRF3 and NF-κB, 12b exhibits the tremendous potential to be a better direct-human-STING agonist than 2’3’-cGAMP. Accordingly, improving the activity or drug properties of acridone analogues still results in many possibilities, such as adding suitable groups at positions N, C-4 and C-7 or synthesising the double-structured molecules by imitating di-ABZIs in order to increase the binding affinity between the molecule and STING.
4. Experiment Section 4.1. Chemistry All reagents and solvents were used as received from commercial sources. 1H-NMR and 13C-NMR
spectra were recorded at 400 MHz and 100 MHz on a JNM-ECA-400 instrument (JEOL
Ltd., Tokyo, Japan) in DMSO-D6. Chemical shifts are expressed in δ (ppm), with tetramethyl silane (TMS) functioning as the internal reference. Coupling constants (J) were expressed in Hz. High-resolution mass spectra were obtained using a TOF G6230A LC/MS (Agilent Technologies,
New York, NY, USA) with an ESI source. Reagents and solvents were commercially available without further purification. The 1H-NMR,
13C-NMR
and HRMS spectra of the compounds in this
article can be found in Supporting Information. 5′,6′-dimethylxanthenone-4-acetic acid (DMXAA-1a).
2-hydroxyphenylacetic acid 5a was
prepared to disodium salt and 2-iodo-3,4-dimethylbenzoic acid 6a was converted into its potassium salt. To a solution of 5a (0.62 g, 3.2 mmol), 6a (1 g, 3.2 mmol), TDA-1 (0.2 g, 0.64 mmol) and CuCl (0.06g, 0.64 mmol) in dry DMSO (5ml) was added. Then the mixture was stirred at 85°C under nitrogen for 8 h. Upon cooling to room temperature and removal of the solvents under a vacuum. The residue was dissolved with 1N NaOH (20ml) followed by filtration. The solvent was acidified to PH=7 by slowly dropping the conc.HCl before filtered to give crude 2-(2-(carboxymethyl) phenoxy)-3,4-dimethylbenzoic acid (0.73g, 76.0% yield).
After the first step, the intermediate
product was cyclodehydrated in conc.H2SO4 (4 ml) and water (1 mL) at 100°C for 2h. After pouring the reaction mixture into ice cold water (25ml), the crude 1a was collected by filtration and was crystallised from EtOAc/MeOH to give pale white solid DMXAA (0.58 g, 64.4% yield over two steps). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 8.06 (dd, J = 8.0, 1.6 Hz, 1H), 7.87 (d, J = 8.1 Hz, 1H), 7.77 (dd, J = 7.2, 1.5 Hz, 1H), 7.39 (t, J = 7.6 Hz, 1H), 7.22 (d, J = 8.1 Hz, 1H), 3.94 (s, 2H), 2.38 (s, 3H), 2.34 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 176.62, 172.41, 154.26,
153.74, 145.20, 137.10, 126.48, 125.82, 125.64, 125.13, 124.15, 123.11, 121.01, 119.26, 35.98, 20.71, 11.59. HRMS (ESI) m/z [M+ H] + calculated for C17H14O4: 283.09 found: 283.09.
(2-bromo-5,6-dimethyl-9-oxo-9H-xanthen-4-yl)-acetic acid (1b). compounds
was
similarly
to
1a.
2-hydroxyphenylacetic
The preparation of title acid
5a
was
replaced
with
2-(5-bromo-2-hydroxyphenyl) acetic acid 5b. (pale gray solid, yield: 51.3%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 12.72 (s, 1H), 8.14 (d, J = 2.5 Hz, 1H), 8.03 (d, J = 2.5 Hz, 1H), 7.92 (d, J = 8.3 Hz, 1H), 7.32 (d, J = 8.2 Hz, 1H), 4.00 (s, 2H), 2.44 (s, 3H), 2.39 (s, 3H).
13C
NMR (101 MHz,
DMSO-D6) δ (ppm): 180.62, 176.41, 158.26, 157.74, 149.20, 141.10, 137.33, 130.48, 129.82, 129.13, 128.15, 127.11, 125.01, 120.19, 39.98, 24.71, 15.59. HRMS (ESI) m/z [M+ H] + calculated for C17H13BrO4: 362.99 found: 363.00.
(6-methoxy-9-oxo-9H-xanthen-4-yl)-acetic acid (1c).
The title compound was obtained similarly
to 1a. 2-iodo-3,4-dimethylbenzoic acid 6a was replaced with 2-iodo-4-methoxybenzoic acid 6b. (yellow solid, yield: 45.8%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 12.59 (s, 1H), 8.12 – 8.07 (m, 2H), 7.78 (dd, J = 7.3, 1.7 Hz, 1H), 7.42 (t, J = 7.9, 7.3 Hz, 1H), 7.11 – 7.04 (m, 2H), 3.98 (s, 2H), 3.94 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 175.49, 172.49, 165.48, 157.78,
154.39, 136.95, 128.07, 125.29, 125.19, 124.28, 121.62, 115.17, 114.49, 100.84, 56.70, 35.20. HRMS (ESI) m/z [M+ H] + calculated for C16H12O5: 285.07 found: 285.08.
(2-bromo-6-methoxy-9-oxo-9H-xanthen-4-yl)-acetic acid (1d).
The preparation of title compound
was similarly to 1a. 2-hydroxyphenylacetic acid 5a and 2-iodo-3,4-dimethylbenzoic acid 6a were replaced with 2-(5-bromo-2-hydroxyphenyl) acetic acid 5b and 2-iodo-4-methoxybenzoic acid 6b respectively. (brown solid, yield: 40.3%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 12.79 (s, 1H), 8.10 (d, J = 2.1 Hz, 1H), 8.05 (d, J = 8.8 Hz, 1H), 7.98 (s, 1H), 7.09 – 7.01 (m, 2H), 3.97 (s, 2H), 3.93 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 174.25, 172.00, 165.73, 157.67, 153.43,
139.06, 128.36, 128.20, 127.04, 122.98, 116.34, 114.84, 114.63,100.90, 56.77, 34.73. HRMS (ESI) m/z [M+ H] + calculated for C16H11BrO5: 362.98 found: 362.99.
General Procedure for the Preparation of (5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2a).
To a magnetically stirred solution of 2-(2-bromophenyl)acetic acid 7a (0.72g,
3.3mmol) and 2-amino-3,4-dimethylbenzoic acid 8a (0.5g, 3.0mmol) in DMF was added powdered Cu (0.02g), Cu2O (0.02g) and K2CO3 (0.33g, 2.4mmol) successively. The reaction mixture was heated at 95℃ for 12h. Upon removal of the solvents, the residue was triturated with 1N NaOH solution. The whole mixture was filtered off and the alkaline solution was acidified with conc.HCl, and then collected the precipitate. The dried residue was subjected to column purification (PE/EA 2:1) to furnish the desired compound. A solution of 2-((2-(carboxymethyl) phenyl) amino)-3,4-dimethylbenzoic acid in Eaton’s reagent (P2O5 –CH3SO3H) (5 mL) at room temperature was heated to 90 °C for 1 h. After cooling to room temperature, the mixture was added slowly to the saturated
aqueous
NaHCO3
solution.
The
crude
9,10-dimethyl-6H-pyrrolo[3,2,1-de]
acridine-1,6(2H)-dione was accumulated by filtration followed by purification with silica gel column
chromatography.
Finally,
NaOH
(0.1g)
was
added
to
a
solution
of
9,10-dimethyl-6H-pyrrolo[3,2,1-de] acridine-1,6(2H)-dione in EtOH (20 mL) and water (2ml). The resulting mixture was refluxed overnight. Upon removal of the solvents, the residue was dissolved with water. Then, the solution was neutralized by conc.HCl. After filtering the mixture, the crude was obtained and purified by silica gel column chromatography to give 0.36g 2a as yellow solid in a yield of 43.8%. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.14 (s, 1H), 9.50 (s, 1H), 8.19 (dd, J = 8.1, 1.3 Hz, 1H), 8.02 (d, J = 8.2 Hz, 1H), 7.69 (dd, J = 7.2, 1.5 Hz, 1H), 7.25 (m, J = 8.0, 7.2 Hz, 1H), 7.16 (d, J = 8.3 Hz, 1H), 4.12 (s, 2H), 2.50 (s, 3H), 2.45 (s, 3H).
13C
NMR (101 MHz,
DMSO-D6) δ (ppm): 177.54 (s), 173.73 (s), 142.42 (s), 139.74 (s), 139.48 (s), 135.94 (s), 125.89 (s), 124.65 (s), 123.64 (s), 123.17 (s), 121.67 (s), 121.26 (s), 119.43 (s), 38.77 (s), 21.21 (s), 12.70 (s). HRMS (ESI) m/z [M+ H] + calculated for C17H15NO3: 282.11 found: 282.11.
(1-fluoro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2b).
The preparation of
title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a was replaced with 2-(2-bromo-4-fluorophenyl) acetic acid 7b. The pale gray solid was got, yield: 32.8%. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.17 (s, 1H), 9.43 (s, 1H), 7.95 (d, J = 8.2 Hz, 1H), 7.65 (m, J = 8.2, 5.4 Hz, 1H), 7.15 (d, J = 8.2 Hz, 1H), 6.95 (m, J = 11.7, 8.2 Hz, 1H), 4.07 (s, 2H), 2.47 (s, 3H), 2.44 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 198.95, 196.39, 185.29, 182.70, 165.35, 164.12,
161.50, 158.98, 147.73, 146.17, 145.85, 143.32, 141.64, 130.24, 61.10, 43.87, 35.35. HRMS (ESI) m/z [M+ H] + calculated for C17H14FNO3: 300.10 found: 300.10.
(1-chloro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2c).
The title compound
was obtained similarly to 2a. 2-(2-bromophenyl) acetic acid 7a was replaced with 2-(2-bromo-4-chlorophenyl) acetic acid 7c. (pale red solid, yield: 28.5%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.28 (s, 1H), 9.53 (s, 1H), 7.95 (d, J = 8.2 Hz, 1H), 7.59 (d, J = 7.9 Hz, 1H), 7.23 (d, J = 7.9 Hz, 1H), 7.15 (d, J = 8.3 Hz, 1H), 4.09 (s, 2H), 2.46 (s, 3H), 2.43 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 176.71, 173.48, 142.51, 142.02, 138.42, 135.38, 132.64, 125.03, 124.25, 123.73, 123.00, 122.71, 120.71, 117.10, 38.80, 21.10, 12.55. HRMS (ESI) m/z [M+ H] + calculated for C17H14ClNO3: 316.07 found: 316.07.
(2-fluoro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2d).
The preparation of
title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a was replaced with 2-(2-bromo-5-fluorophenyl) acetic acid 7d. The brown solid was obtained, yield: 37.6%. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.35 (s, 1H), 9.88 (s, 1H), 8.00 (d, J = 8.3 Hz, 1H), 7.82 (dd, J = 8.9, 3.1 Hz, 1H), 7.68 (dd, J = 8.9, 3.0 Hz, 1H), 7.16 (d, J = 8.4 Hz, 1H), 4.16 (s, 2H), 2.48 (s, 3H), 2.44 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 199.60, 196.05, 180.90, 178.51, 165.30,
162.29, 159.54, 149.63, 147.61, 146.80, 146.22, 144.47, 141.40, 131.91, 61.72, 43.93, 35.50. HRMS (ESI) m/z [M+ H] + calculated for C17H14FNO3: 300.10 found: 300.10.
(2-chloro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2e).
The title compound
was obtained similarly to 2a. 2-(2-bromophenyl) acetic acid 7a was replaced with 2-(2-bromo-5-chlorophenyl) acetic acid 7e. (pale brown solid, yield: 39%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.78 (s, 1H), 11.00 (s, 1H), 7.99 (d, J = 2.3 Hz, 1H), 7.97 (s, 1H), 7.55 (d, J = 2.5 Hz, 1H), 7.10 (d, J = 8.3 Hz, 1H), 3.74 (s, 2H), 2.50 (s, 3H), 2.41 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 176.68, 173.66, 142.04, 140.82, 140.22, 133.79, 130.29, 125.36, 124.55, 124.48, 123.42, 122.81, 121.92, 119.49, 45.15, 21.23, 13.49. HRMS (ESI) m/z [M+ H] + calculated for C17H14ClNO3: 316.07 found: 316.07.
(7-bromo-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid anion (2f).
The title
compound was obtained similarly to 2a. 2-amino-3,4-dimethylbenzoic acid 8a was replaced with 2-amino-5-bromo-3,4-dimethylbenzoic acid 8b. The starting material 8b was synthesized according to a method previously reported in the literature.[25] The dark brown solid was provided in 28.8% yield. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 9.69 (s, 1H), 8.25 (s, 1H), 8.17 (d, J = 8.1 Hz, 1H), 7.71 (d, J = 7.1 Hz, 1H), 7.27 (t, J = 7.6 Hz, 1H), 4.12 (s, 2H), 2.58 (s, 3H), 2.54 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 176.46, 173.80, 140.98, 139.76, 138.55, 136.29, 126.81, 126.18, 125.80, 123.50, 122.08, 121.15, 120.14, 118.71, 38.96, 21.10, 14.32. HRMS (ESI) m/z [M+ H] + calculated for C17H14BrNO3: 360.02 found: 360.02.
(7-methoxy-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic acid (2g).
The preparation of
title compound was similarly to 2a. 2-amino-3,4-dimethylbenzoic acid 8a was replaced with 2-amino-5-methoxy-3,4-dimethylbenzoic acid 8c. The starting material 8c was synthesized according to a method previously reported in the literature.[25] (gray solid, yield: 25%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 9.49 (s, 1H), 8.19 (dd, J = 8.1, 1.3 Hz, 1H), 7.67 (dd, J = 7.2, 1.3 Hz, 1H), 7.53 (s, 1H), 7.23 (dd, J = 8.0, 7.2 Hz, 1H), 4.11 (s, 2H), 3.89 (s, 3H), 2.53 (s, 3H), 2.32 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 176.85, 173.75, 153.25, 139.29, 135.45, 134.39,
133.16, 125.81, 125.23, 123.10, 121.36, 120.67, 119.38, 101.69, 55.97, 38.78, 13.40, 13.26. HRMS (ESI) m/z [M+ H] + calculated for C18H17NO4: 312.12 found: 312.14.
(7-bromo-1-fluoro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic
acid
(2h).The
title
compound
acetic
acid
and
was
obtained
similarly
to
2a.
2-(2-bromophenyl)
7a
2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-4-fluorophenyl) acetic acid 7b and 2-amino-5-bromo-3,4-dimethylbenzoic acid 8b respectively. (dark yellow solid, yield: 17.3%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.23 (s, 1H), 9.55 (s, 1H), 8.17 (s, 1H), 7.67 (dd, J = 8.2, 5.4 Hz, 1H), 6.97 (dd, J = 11.5, 8.2 Hz, 1H), 4.07 (s, 2H), 2.56 (s, 3H), 2.52 (s, 3H).
13C
NMR (101
MHz, DMSO-D6) δ (ppm): 174.91 (s), 173.75 (s), 162.40 (s), 159.80 (s), 141.15 (s), 137.80 (s), 136.68 (d, J = 10.9 Hz), 126.59 (s), 126.00 (s), 121.16 (s), 119.10 (s), 110.81 (s), 108.09 (s), 38.48 (s), 21.01 (s), 14.21 (s). HRMS (ESI) m/z [M+ H] + calculated for C17H13BrFNO3: 378.01 found: 378.01.
(7-bromo-1-chloro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic
acid
(2i).The
title
compound
acetic
acid
and
was
obtained
similarly
to
2a.
2-(2-bromophenyl)
7a
2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-4-chlorophenyl) acetic acid 7c and 2-amino-5-bromo-3,4-dimethylbenzoic acid 8b respectively. (brown solid, yield: 22.5%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.30 (s, 1H), 9.48 (s, 1H), 8.17 (s, 1H), 7.61 (d, J = 7.9 Hz, 1H), 7.25 (d, J = 7.9 Hz, 1H), 4.09 (s, 2H), 2.54 (d, J = 5.9 Hz, 3H), 2.51 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 175.55 (s), 173.44 (s), 142.01 (s), 141.25 (s), 137.57 (s), 135.83 (s), 132.73 (s), 127.02 (s), 126.03 (s), 124.68 (s), 122.82 (s), 121.44 (s), 119.15 (s), 117.12 (s), 38.57 (s), 21.05 (s),
14.19 (s). HRMS (ESI) m/z [M+ H] + calculated for C17H13BrClNO3: 395.65 found: 395.98.
(7-bromo-2-fluoro-5,6-dimethyl-9-oxo-9,10-dihydro-acridin-4-yl)-acetic
acid
(2j).The
title
compound
acetic
acid
and
was
obtained
similarly
to
2a.
2-(2-bromophenyl)
7a
2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-5-fluorophenyl) acetic acid 7d and 2-amino-5-bromo-3,4-dimethylbenzoic acid 8b respectively. (yellow solid, yield: 23.4%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 10.23 (s, 1H), 8.21 (s, 1H), 7.78 (dd, J = 8.8, 3.0 Hz, 1H), 7.68 (dd, J = 8.9, 3.0 Hz, 1H), 4.13 (s, 2H), 2.53 (s, 3H), 2.50 – 2.48 (m, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 178.95, 176.39, 165.29, 162.70, 145.35, 144.16, 141.50, 139.10, 135.98, 126.17, 123.32, 121.64, 113.56, 110.24, 41.10, 23.87, 15.35. HRMS (ESI) m/z [M+ H] + calculated for C17H13BrFNO3: 378.01 found: 378.01.
2-(7-bromo-2-chloro-5,6-dimethyl-9-oxo-9,10-dihydroacridin-4-yl) acetic acid (2k). The title compound
was
obtained
similarly
to
2a.
2-(2-bromophenyl)
acetic
acid
7a
and
2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-5-chlorophenyl) acetic acid 7e and 2-amino-5-bromo-3,4-dimethylbenzoic acid 8b respectively. (yellow solid, yield: 16.6%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 15.19 (s, 1H), 10.59(s, 1H), 8.23 (s, 1H), 7.96 (d, J = 2.4 Hz, 1H), 7.54 (d, J = 2.4 Hz, 1H), 3.67 (s, 2H), 2.62 (s, 3H), 2.52 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 176.46, 173.80, 140.98, 139.76, 138.55, 136.29, 135.21, 130.18, 129.80, 123.50, 122.08, 121.15, 120.14, 118.71, 38.96, 21.10, 14.32. HRMS (ESI) m/z [M+ H] + calculated for C17H13BrClNO3: 395.65 found: 395.98.
2-(1-fluoro-7-methoxy-5,6-dimethyl-9-oxo-9,10-dihydroacridin-4-yl)
acetic
acid
(2l).
The
preparation of title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a and 2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-4-fluorophenyl) acetic acid 7b and 2-amino-5-methoxy-3,4-dimethylbenzoic acid 8c. The dark brown solid was provided in 20.8% yield. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 9.65 (s, 1H), 7.61 (dd, J = 8.1, 5.3 Hz, 1H), 7.48 (s, 1H), 6.91 (dd, J = 11.7, 8.2 Hz, 1H), 4.04 (s, 2H), 3.88 (s, 3H), 2.50 (s, 3H), 2.30 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 175.51, 173.76, 162.48, 159.90, 153.56, 141.05, 135.68, 133.19,
125.21, 120.63, 118.97, 110.36, 107.22, 101.64, 55.98, 38.60, 13.32, 13.19. HRMS (ESI) m/z [M+ H] + calculated for C18H16FNO4: 330.11 found: 330.11.
2-(1-chloro-7-methoxy-5,6-dimethyl-9-oxo-9,10-dihydroacridin-4-yl)
acetic
acid
(2m).
The
preparation of title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a and 2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-4- chlorophenyl) acetic acid 7c and 2-amino-5-methoxy-3,4-dimethylbenzoic acid 8c. The dark brown solid was provided in 21.1% yield. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.15 (s, 1H), 9.41 (s, 1H), 7.57 (d, J = 7.9 Hz, 1H), 7.48 (s, 1H), 7.21 (d, J = 7.8 Hz, 1H), 4.10 (s, 2H), 3.88 (s, 3H), 2.50 – 2.46 (m, 3H), 2.31 (s, 3H). 13C
NMR (101 MHz, DMSO-D6) δ (ppm): 175.99, 173.42, 153.59, 141.52, 134.93, 133.22, 133.17,
132.54, 125.11, 123.94, 122.59, 120.76, 116.46, 101.94, 55.98, 38.59, 13.31, 13.14. HRMS (ESI) m/z [M+ H] + calculated for C18H16ClNO4: 346.08 found: 346.08.
2-(2-fluoro-7-methoxy-5,6-dimethyl-9-oxo-9,10-dihydroacridin-4-yl)
acetic
acid
(2n).
The
preparation of title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a and 2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-5- fluorophenyl) acetic acid 7d and 2-amino-5-methoxy-3,4-dimethylbenzoic acid 8c. The brown solid was obtained in 23.6% yield. 1H
NMR (400 MHz, DMSO-D6) δ (ppm): 10.23 (s, 1H), 8.21 (s, 1H), 7.78 (dd, J = 8.8, 3.0 Hz, 1H),
7.68 (dd, J = 8.9, 3.0 Hz, 1H), 4.13 (s, 2H), 2.53 (s, 3H), 2.48 (s, 3H).
13C
NMR (101 MHz,
DMSO-D6) δ (ppm): 176.12, 173.18, 155.61, 153.39, 136.18, 134.37, 133.47, 126.42, 125.31, 123.80, 121.02, 118.61, 109.28, 101.35, 55.97, 38.29, 13.40, 13.27. HRMS (ESI) m/z [M+ H] + calculated for C18H16FNO4: 330.11 found: 330.11.
2-(2-chloro-7-methoxy-5,6-dimethyl-9-oxo-9,10-dihydroacridin-4-yl)
acetic
acid
(2o).
The
preparation of title compound was similarly to 2a. 2-(2-bromophenyl) acetic acid 7a and 2-amino-3,4-dimethylbenzoic acid 8a were replaced with 2-(2-bromo-5- chlorophenyl) acetic acid 7e and 2-amino-5-methoxy-3,4-dimethylbenzoic acid 8c. The yellow solid was obtained in 23.1% yield. 1H
NMR (400 MHz, DMSO-D6) δ (ppm): 13.21 (s, 1H), 9.51 (s, 1H), 8.11 (s, 1H), 7.76 (s, 1H), 7.50
(s, 1H), 4.18 (s, 2H), 3.89 (s, 3H), 2.51 (s, 3H), 2.32 (s, 3H).
13C
NMR (101 MHz, DMSO-D6) δ
(ppm): 175.75, 173.21, 153.53, 137.93, 134.88, 134.23, 133.64, 126.14, 125.57, 125.46, 124.28, 121.30, 119.27, 101.56, 56.00, 38.11, 13.42, 13.27. HRMS (ESI) m/z [M+ H] + calculated for C18H16ClNO4: 346.08 found: 346.08.
10-carboxymethyl-9-acridanone (CMA, 3a).
Intermediate ethyl 2-(9-oxoacridin-10(9H)-yl)
acetate was synthesized by the method of 13b. To the solution of ethyl 2-(9-oxoacridin-10(9H)-yl) acetate (0.30g, 1.1 mmol) and NaOH (0.05 g, 1.3 mmol) in ethanol (20 ml) and water (2 ml) was added, and the mixture was heated to 60℃ for 1h. Upon removal of the solvents, the resulting residue was dissolved in water and filtered. After neutralization of the filtrate with conc.HCl, the mixture was filtrated to get the crude solid, subsequently recrystallized by methanol for 0.2g white power, the yield: 73.3%. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 13.40 (s, 1H), 8.37 (d, J = 1.6 Hz, 1H), 8.35 (d, J = 1.6 Hz, 1H), 7.82 (ddd, J = 8.7, 6.9, 1.7 Hz, 2H), 7.69 (s, 1H), 7.67 (s, 1H), 7.36 (t, J = 7.4 Hz, 2H), 5.34 (s, 2H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 177.20, 170.58, 142.65 (2C), 134.77 (2C), 127.16 (2C), 122.12 (2C), 122.06(2C), 116.31 (2C), 48.02. HRMS (ESI) m/z [M+ H] + calculated for C15H11NO3: 254.07 found: 254.08.
2-methoxy-5,6-dimethyl-10-carboxymethyl-9-acridanone (3b). similarly
to
3a.
2-(9-oxoacridin-10(9H)-yl)
The title compound was obtained
acetate
was
replaced
with
2-methoxy-5,6-dimethyl-10-(2-oxobutyl) acridin-9(10H)-one 13b. (pale red solid, yield: 78.6%). 1H NMR (400 MHz, DMSO-D6) δ (ppm): 10.59 (s, 1H), 8.03 (d, J = 8.3 Hz, 1H), 7.98 (d, J = 9.2 Hz, 1H), 7.59 (d, J = 3.0 Hz, 1H), 7.41 – 7.36 (m, 1H), 7.09 (d, J = 8.3 Hz, 1H), 3.91 (s, 2H), 3.85 (s, 3H), 2.50 (s, 3H), 2.43 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 176.81, 174.70, 154.49, 141.53, 139.71, 136.56, 124.30, 123.90, 123.64, 123.01, 121.09, 120.55, 119.00, 105.01, 60.03, 55.81, 21.23, 13.76. HRMS (ESI) m/z [M+ H] + calculated for C18H17NO4: 312.12 found: 312.13.
2-methoxy-5,6-dimethyl-9-oxo-9,10-dihydroacridine-4-carboxylic acid (4).
The title compound
was obtained similarly to 9g. The starting materials of 7a and 8c were replaced with 10b and 8a, the green product was got in the yield of 39.8%. 1H NMR (400 MHz, DMSO-D6) δ (ppm): 14.01 (s, 1H), 11.87 (s, 1H), 7.98 – 7.88 (m, 3H), 7.10 (d, J = 8.2 Hz, 1H), 3.89 (s, 3H), 2.42 (s, 3H), 2.36 (s,
3H).
13C
NMR (101 MHz, DMSO-D6) δ (ppm): 173.82, 168.67, 148.93, 138.53, 136.73, 133.57,
121.26, 120.89, 120.65, 120.00, 118.12, 117.55, 116.03, 111.70, 59.23, 18.22, 10.75. HRMS (ESI) m/z [M+ H] + calculated for C17H15NO4: 298.10 found: 298.11.
8-methoxy-9,10-dimethyl-6H-pyrrolo[3,2,1-de] acridine-1,6(2H)-dione (9g).
The starting
material 2-amino-5-methoxy-3,4-dimethyl-benzoic acid 8c was synthesized according to a method previously
reported
in
the
literature.[25]
To
a
magnetically
stirred
solution
of
2-(2-bromophenyl)acetic acid 7a (0.61g, 2.8mmol) and 8c (0.5g, 2.6mmol) in DMF was added powdered Cu (0.02g), Cu2O (0.02g) and K2CO3 (0.28g, 2mmol) successively. The reaction mixture was heated to 95℃ for 12h. Upon removal of the solvents, the residue was triturated with 1N NaOH solution. The whole mixture was filtered off and the alkaline solution was acidified with conc. HCl, and then collected the precipitate. The dried residue was subjected to column purification (PE/EA 4:1) to furnish the desired compound. A solution of 2-((2-(carboxymethyl) phenyl) amino)-5-methoxy-3,4-dimethyl-benzoic acid in Eaton’s reagent (P2O5 –CH3SO3H) (5 mL) at room temperature was heated to 90 °C for 1 h. After cooling to room temperature, the mixture was added slowly to the saturated aqueous NaHCO3 solution. The crude was accumulated by filtration followed by purification with silica gel column chromatography to give 0.3g product: the dark brown solid, yield 37.5%. 1H NMR (400 MHz, CHLOROFORM-D) δ (ppm): 8.17 (d, J = 8.0 Hz, 1H), 7.81 (s, 1H), 7.62 (dd, J = 7.1, 0.9 Hz, 1H), 7.42 – 7.36 (m, 1H), 3.99 (s, 3H), 3.92 (s, 2H), 2.39 (s, 3H), 2.37 (s, 3H). 13C NMR (101 MHz, DMSO-D 6) δ (ppm): 173.51, 161.50, 155.21, 153.01, 139.05, 134.15, 132.91, 125.57, 124.99, 122.86, 121.12, 120.42, 119.14, 101.45, 55.73, 38.54, 13.16, 13.02. HRMS (ESI) m/z [M+ H] + calculated for C18H15NO3: 294.11 found: 294.11.
General Procedure for the Preparation of acridin-9(10H)-one (12a).
To a solution of
2-bromo-benzoic acid 10a (1.29g, 6.4mmol) and aniline 11a (0.6g, 6.4mmol) in DMF (6ml) at room temperature, we subsequently added powdered Cu (0.05g), Cu2O (0.05g) and K2CO3 (0.71g, 5.1mmol) was added. The reaction mixture was heated at 110℃ for 12h. Upon removal of the solvents under a vacuum, the residue was dissolved in 1N NaOH solution(25ml). The crude product was obtained by precipitation upon acidification of the filtrate with conc.HCl. After drying, the crude
2-(phenylamino) benzoic acid was added to Eaton’s reagent (5 mL) at room temperature, then the mixture was heated to 90 °C for 1 h. The cooled reaction mixture was dropped into the saturated aqueous NaHCO3 solution. The precipitate was filtered to collect the rough product. Purification of the residue by silica gel column chromatography provided compound 12a, an off-white solid. (0.47 g, 37.6% yield). 1H-NMR (400 MHz, DMSO-D 6) δ (ppm): 11.75 (s, 1H), 8.28 – 8.20 (m, 2H), 7.74 (ddd, J = 8.4, 6.9, 1.5 Hz, 2H), 7.55 (d, J = 8.3 Hz, 2H), 7.32 – 7.23 (m, 2H). 13C NMR (101 MHz, DMSO-D 6) δ (ppm): 177.30, 141.41 (2C), 133.99 (2C), 126.54 (2C), 121.53 (2C), 121.01 (2C), 117.87 (2C). HRMS (ESI) m/z [M+ H] + calculated for C13H9NO: 196.07 found: 196.08.
2-methoxy-5,6-dimethylacridin-9(10H)-one (12b).
The preparation of title compound was
similarly to 12a. The compound 12b was obtained from 2-bromo-5-methoxybenzoic acid 10b and 2,3-dimethylaniline 11b, as a pale yellow solid in 35.5% yield. 1H-NMR (400 MHz, DMSO-D 6 ) δ (ppm): 10.44 (s, 1H), 8.03 (d, J = 8.3 Hz, 1H), 7.90 (d, J = 9.1 Hz, 1H), 7.60 (d, J = 3.0 Hz, 1H), 7.38 (dd, J = 9.1, 3.0 Hz, 1H), 7.08 (d, J = 8.3 Hz, 1H), 3.86 (s, 3H), 2.47 (s, 3H), 2.41 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ: 176.82, 154.51, 141.53, 139.73, 136.57, 124.26, 123.89, 123.65, 123.00, 121.12, 120.55, 119.03, 105.10, 55.84, 21.22, 13.75. HRMS (ESI) m/z [M+ H] + calculated for C16H15NO2: 254.11 found: 254.12.
2-methoxyacridin-9(10H)-one (12c).
The preparation of title compound was similarly to 12a.
The compound 12c was obtained from 2-bromo-5-methoxybenzoic acid 10b and aniline 11a, as a pale green solid in 40.4% yield. 1H-NMR (400 MHz, DMSO-D 6 ) δ (ppm): 11.74 (s, 1H), 8.23 (dd, J = 8.2, 1.4 Hz, 1H), 7.71 (ddd, J = 8.4, 6.9, 1.5 Hz, 1H), 7.63 (d, J = 2.9 Hz, 1H), 7.54 (d, J = 3.2 Hz, 1H), 7.52 (d, J = 2.4 Hz, 1H), 7.42 (dd, J = 9.0, 3.0 Hz, 1H), 7.24 (ddd, J = 8.0, 6.9, 1.0 Hz, 1H), 3.86 (s, 3H).
13C
NMR (101 MHz, DMSO-D 6) δ (ppm): 176.64, 154.47, 140.97, 136.24, 133.55,
126.47, 124.82, 121.50, 121.23, 120.15, 119.72, 117.83, 105.37, 55.87. HRMS (ESI) m/z [M+ H] + calculated for C14H11NO2: 226.08 found: 226.09.
3,4-dimethylacridin-9(10H)-one (12d).
The preparation of title compound was similarly to 12a.
The compound 12d was obtained from 2-bromo-benzoic acid 10a and 2,3-dimethylaniline 11b, as a
yellow-green solid in 45.3% yield. 1H-NMR (400 MHz, DMSO-D 6 ) δ (ppm): 10.47 (s, 1H), 8.20 (dd, J = 8.1, 1.3 Hz, 1H), 8.03 (d, J = 8.2 Hz, 1H), 7.93 (d, J = 8.4 Hz, 1H), 7.73 – 7.67 (m, 1H), 7.25 (ddd, J = 8.0, 7.0, 0.9 Hz, 1H), 7.11 (d, J = 8.2 Hz, 1H), 2.49 (s, 3H), 2.42 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 173.82, 138.53, 137.60, 136.73, 127.67, 121.26, 120.89, 120.65, 120.00, 119.44, 118.12, 117.55, 116.03, 18.22, 10.75. HRMS (ESI) m/z [M+ H] + calculated for C15H13NO: 224.10 found: 224.11.
2-methoxy-3,4-dimethylacridin-9(10H)-one(12e). to
12a.
The
compound
12d
was
The preparation of title compound was similarly
obtained
from
2-bromo-benzoic
acid
10a
and
4-methoxy-2,3-dimethylaniline 11c, as a dark green solid in 38.8% yield. 1H-NMR (400 MHz, DMSO-D 6 ) δ (ppm): 10.46 (s, 1H), 8.20 (d, J = 7.9 Hz, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.68 (t, J = 7.6 Hz, 1H), 7.55 (s, 1H), 7.22 (t, J = 7.2 Hz, 1H), 3.88 (s, 3H), 2.52 (s, 3H), 2.30 (s, 3H). 13C NMR (101 MHz, DMSO-D 6) δ (ppm): 176.66, 152.83, 141.21, 134.90, 133.01, 132.79, 126.01, 125.24, 121.25, 119.91, 119.60, 118.64, 101.72, 55.90, 14.27, 13.35. HRMS (ESI) m/z [M+ H] + calculated for C16H15NO2: 254.11 found: 254.12.
2,7-dimethoxy-3,4-dimethylacridin-9(10H)-one(12f).
The preparation of title compound was
similarly to 12a. The compound 12d was obtained from 2-bromo-5-methoxybenzoic acid 10b and 4-methoxy-2,3-dimethylaniline 11c, as a black-green solid in 31.7% yield. 1H-NMR (400 MHz, DMSO-D 6 ) δ (ppm): 10.48 (s, 1H), 7.89 (d, J = 9.1 Hz, 1H), 7.58 (d, J = 2.9 Hz, 1H), 7.53 (s, 1H), 7.36 (dd, J = 9.1, 3.0 Hz, 1H), 3.87 (s, 3H), 3.85 (s, 3H), 2.50 (s, 3H), 2.30 (s, 3H). 13C NMR (101 MHz, DMSO-D6) δ (ppm): 175.84, 154.26, 152.69, 136.11, 134.63, 132.46, 125.10, 124.02, 120.53, 120.30, 118.73, 104.50, 101.37, 55.89, 55.76, 14.24, 13.33. HRMS (ESI) m/z [M+ H] + calculated for C17H17NO3: 284.12 found: 284.13.
2-methoxy-5,6-dimethyl-10-(2-oxobutyl) acridin-9(10H)-one(13b).
To a solution of NaH
(1.43mmol) and compound 12b (0.33g, 1.3mmol) in DMF (5ml) at room temperature was stirred for 1 h and then cooled to 5–7℃. The ethyl bromoacetate (0.43ml, 2.6mmol) was added to the resulting mixture and continuously stirred at room temperature for 20 h. After completion of the reaction
(TLC), the reaction mixture was poured into ice water (15ml). The resulting precipitates were filtered off, dried, and then extracted with chloroform. Evaporation of the solvent gave crude ester which was purified by recrystallization. As a green solid (0.33g) in yield of 76%. 1H NMR (400 MHz, DMSO-D6) δ 8.12 (d, J = 8.8 Hz, 1H), 8.06 (d, J = 9.4 Hz, 1H), 7.63 (d, J = 2.8 Hz, 1H), 7.50–7.43 (m, 2H), 5.05 (s, 2H), 4.25 (q, J = 7.1 Hz, 2H), 3.97 (s, 3H), 2.77 (s, 3H), 2.51 (s, 3H), 1.26 (d, J = 7.1 Hz, 3H).
13C
NMR (101 MHz, DMSO-D 6) δ (ppm): 169.40, 158.26, 156.85, 147.94, 146.48,
136.71, 133.75, 131.85, 129.78, 125.47, 119.97, 119.40, 118.75, 98.58, 72.24, 61.45, 56.04, 20.84, 14.58, 13.90. HRMS (ESI) m/z [M+ H] + calculated for C20H21NO4: 340.15 found: 340.16.
4.2.
Biological Evaluations
4.2.1.
In vitro cell-based Assay
The following reporter cell lines and positive compound were obtained from InvivoGen: 293T-hSTING-R232 Cells, 293T-mSTING Cells, THP1-KO-STING and 2′,3′-cGAMP. The setting of concentration gradient as follows: the targeted small molecule was prepared for ten different concentrations (3.5 μM, 5.2 μM, 7.8 μM, 11.7 μM, 17.6 μM, 26.3 μM, 39.5 μM, 59.2 μM, 88.8 μM, 133.3 μM, and 200 μM), and 2′,3′-cGAMP was set into eight different concentrations (0.27 μM, 0.54 μM, 1.08 μM, 2.16 μM, 4.32 μM, 8.64 μM, 17.3 μM, and 34.6 μM). The general procedure for screening of targeted compounds for cytokine induction activity: Firstly, 180μL of 293T cells (∼50000 cells per well) or THP-1 cells (∼100000 cells per well) were stimulated with 20μL of either saline or a saline solution of a test compound for 48 h at 37 °C in a 5% CO2 incubator. Then, the inductions of cytokines were indirectly quantified using QUANTI-Blue or QUANTI-Luc, which were prepared and used according to the manufacturer’s instructions. Finally, EC50 values were calculated using software Origin 8.0, and Emax values were normalized to the response induced by 2′,3′-cGAMP in each assay. The results are expressed as the mean ± SD from three independent experiments.
4.2.2. HTRF ® Human STING binding competitive Assay By using the human STING binding kits purchased from Cisbio, the human STING binding
competitive assay was performed. According to the instructions, the following solutions were successively added to each well in the 384-well plate: 5 μL of the detection compounds or 2′,3′-cGAMP (positive control) with different concentration or diluent (negative control), 5 μL of human STING 6His-tagged protein (negative control well was added to detection buffer), and 10 µL of STING ligand-d2 and Anti 6His-Tb3+ premixed working solution. After sealed cell plate and incubated at room temperature for 3 hours, the fluorescence values at 665nm and 620nm were respectively read by using Infinite® F200 PRO that equipped with the HTRF® module. The ratio of the two fluorescence intensities (665nm/620nm) was considered to estimate the binding potency of agonists. The IC50 (50% inhibitory concentration) was calculated using software GraphPad Prism.
4.2.3. Protein Expression and Purification The sequences corresponding to residues aa 137-379 of hSTING were inserted into a modified pET-24a (Novagen), in which the target protein was separated from the preceding His6-SUMO tag by an Enterokinase (EK) cleavage site. The gene sequences were subsequently confirmed by sequencing. The fusion proteins were expressed in BL21 (DE3) RIL cell strain. The cells were grown at 37 °C until OD600 reached approx. 0.8. The temperature was then shifted to 18 °C and the cells were induced by addition of isopropyl β-D-1-thiogalactopyranoside (IPTG) to the culture medium at a final concentration of 0.5 mM. After induction, the cells were grown for 16h. The fusion proteins were purified over a Ni-NTA affinity column. The His6-SUMO tag was removed by EK cleavage during dialysis against buffer containing 50 mM Tris-HCl, 150 mM NaCl, pH 7.5. After dialysis, the His6-SUMO tag was removed. The final sample of hSTING contains about 1 mg/ml protein, 0.1M PBS pH 7.5.
4.2.4. Isothermal Titration Calorimetry Binding Assay Human STING (137-379) was expressed and purified as described above. The dissociation constants (Kd) of small molecules (2’3’-cGAMP and 12b) with the purified hSTING protein were measured by isothermal titration calorimetry using a MicroCal ITC200 calorimeter at 25°C. The protein sample was diluted with working buffer 0.01 M PBS, pH 7.5 and the lyophilized compound was dissolved in the working buffer. 2 µL sample was added to the solution of sample cell
containing the protein, the titration needs to be successively performed 20 times (spaced 120s per time). Data were analyzed with the MicroCal analysis of launcher software.
4.2.5. Analysis of Cytokine Expression (Real-Time PCR) The diverse cytokines of mRNAs expression were measured using total cellular RNA via quantitative RT-PCR (qPCR) assays. Total RNA was isolated from THP-1 cells using Trizol provided by Invitrogen. Following the instruction, the cDNA was synthesized by the reverse transcription synthesis kit (Sino Biological Inc.) using 2μg of total RNA. Then, the detection of qPCR was performed in triplicate with Roche LightCycler® 480. Relative expression was normalized to the levels of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Finally, the various genes expressions of the sample were calculated using the 2–ΔΔCT method. The tested samples were limited by the conditions of testing time (0h, 8h, 20h, and 30h) and different concentrations (2’3’-cGAMP at 15 μg/ml and compound 12b at 8 μM, 16 μM, 40 μM, and 100 μM).
Conflicts of interest The authors are certain that this content of this article involves no conflict of interest.
Acknowledgement We are grateful for the financial supports of the National Science and Technology Major Projects for "Major New Drugs Innovation and Development" (2018ZX09711003) of China. References
1.
Chen Q, Sun L, Chen ZJ: Regulation and function of the cGAS–STING pathway of cytosolic DNA sensing. Nature Immunology 2016, 17:1142.
2.
Kondo T, Kobayashi J, Saitoh T, Maruyama K, Ishii KJ, Barber GN, Komatsu K, Akira S, Kawai T: DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon
by regulating STING trafficking. Proceedings of the National Academy of Sciences 2013, 110(8):2969. 3.
Zhang X, Shi H, Wu J, Zhang X, Sun L, Chen C, Chen Zhijian J: Cyclic GMP-AMP Containing Mixed Phosphodiester Linkages Is An Endogenous High-Affinity Ligand for STING. Molecular Cell 2013, 51(2):226-235.
4.
Wu J, Sun L, Chen X, Du F, Shi H, Chen C, Chen ZJ: Cyclic GMP-AMP Is an Endogenous Second Messenger in Innate Immune Signaling by Cytosolic DNA. Science 2013, 339(6121):826.
5.
Zhao B, Du F, Xu P, Shu C, Sankaran B, Bell SL, Liu M, Lei Y, Gao X, Fu X et al: A conserved PLPLRT/SD motif of STING mediates the recruitment and activation of TBK1. Nature 2019, 569(7758):718-722.
6.
Woo S-R, Fuertes Mercedes B, Corrales L, Spranger S, Furdyna Michael J, Leung Michael YK, Duggan R, Wang Y, Barber Glen N, Fitzgerald Katherine A et al: STING-Dependent Cytosolic DNA Sensing Mediates Innate Immune Recognition of Immunogenic Tumors. Immunity 2014, 41(5):830-842.
7.
Fuertes MB, Kacha AK, Kline J, Woo S-R, Kranz DM, Murphy KM, Gajewski TF: Host type I IFN signals are required for antitumor CD8 T cell responses through CD8α dendritic cells. The Journal of
Experimental Medicine 2011, 208(10):2005. 8.
Li T, Chen ZJ: The cGAS–cGAMP–STING pathway connects DNA damage to inflammation, senescence, and cancer. The Journal of Experimental Medicine 2018, 215(5):1287.
9.
Lioux T, Mauny M-A, Lamoureux A, Bascoul N, Hays M, Vernejoul F, Baudru A-S, Boularan C, Lopes-Vicente J, Qushair G et al: Design, Synthesis, and Biological Evaluation of Novel Cyclic Adenosine–Inosine Monophosphate (cAIMP) Analogs That Activate Stimulator of Interferon Genes (STING). Journal of Medicinal Chemistry 2016, 59(22):10253-10267.
10.
Corrales L, Glickman Laura H, McWhirter Sarah M, Kanne David B, Sivick Kelsey E, Katibah George E, Woo S-R, Lemmens E, Banda T, Leong Justin J et al: Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell
Reports 2015, 11(7):1018-1030. 11.
Li L, Yin Q, Kuss P, Maliga Z, Millán JL, Wu H, Mitchison TJ: Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nature Chemical Biology 2014, 10:1043.
12.
Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, Rathinam VAK, Monks B, Jin T, Xiao TS et al: Mouse, but not Human STING, Binds and Signals in Response to the Vascular Disrupting Agent 5,6-Dimethylxanthenone-4-Acetic Acid. The Journal of Immunology 2013, 190(10):5216.
13.
Kim S, Li L, Maliga Z, Yin Q, Wu H, Mitchison TJ: Anticancer Flavonoids Are Mouse-Selective STING Agonists. ACS Chemical Biology 2013, 8(7):1396-1401.
14.
Cavlar T, Deimling T, Ablasser A, Hopfner K-P, Hornung V: Species-specific detection of the antiviral small-molecule compound CMA by STING. The EMBO Journal 2013, 32(10):1440-1450.
15.
Sali TM, Pryke KM, Abraham J, Liu A, Archer I, Broeckel R, Staverosky JA, Smith JL, Al-Shammari A, Amsler L et al: Characterization of a Novel Human-Specific STING Agonist that Elicits Antiviral Activity Against Emerging Alphaviruses. PLOS Pathogens 2015, 11(12):e1005324.
16.
Liu B, Tang L, Zhang X, Ma J, Sehgal M, Cheng J, Zhang X, Zhou Y, Du Y, Kulp J et al: A cell-based high throughput screening assay for the discovery of cGAS-STING pathway agonists. Antiviral
Research 2017, 147:37-46. 17.
Pryke KM, Abraham J, Sali TM, Gall BJ, Archer I, Liu A, Bambina S, Baird J, Gough M, Chakhtoura M
et al: A Novel Agonist of the TRIF Pathway Induces a Cellular State Refractory to Replication of Zika,
Chikungunya, and Dengue Viruses. mBio 2017, 8(3):e00452-00417. 18.
Zhang Y, Sun Z, Pei J, Luo Q, Zeng X, Li Q, Yang Z, Quan J: Identification of α-Mangostin as an Agonist of Human STING. ChemMedChem 2018, 13(19):2057-2064.
19.
Zhang X, Liu B, Tang L, Su Q, Hwang N, Sehgal M, Cheng J, Ma J, Zhang X, Tan Y et al: Discovery and Mechanistic Study of a Novel Human-Stimulator-of-Interferon-Genes Agonist. ACS Infectious
Diseases 2019, 5(7):1139-1149. 20.
Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang S-Y, Tran J-L, Moore P, Lehmann S, Eberl HC et al: Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 2018, 564(7736):439-443.
21.
Cui X, Zhang R, Cen S, Zhou J: STING modulators: Predictive significance in drug discovery.
European Journal of Medicinal Chemistry 2019, 182:111591. 22.
Gao P, Ascano M, Zillinger T, Wang W, Dai P, Serganov Artem A, Gaffney Barbara L, Shuman S, Jones Roger A, Deng L et al: Structure-Function Analysis of STING Activation by c[G(2′,5′)pA(3′,5′)p] and Targeting by Antiviral DMXAA. Cell 2013, 154(4):748-762.
23.
Gao P, Zillinger T, Wang W, Ascano M, Dai P, Hartmann G, Tuschl T, Deng L, Barchet W, Patel Dinshaw J: Binding-Pocket and Lid-Region Substitutions Render Human STING Sensitive to the Species-Specific Drug DMXAA. Cell Reports 2014, 8(6):1668-1676.
24.
Hwang J, Kang T, Lee J, Choi B-S, Han S: Design, synthesis, and biological evaluation of C7-functionalized DMXAA derivatives as potential human-STING agonists. Organic & Biomolecular
Chemistry 2019, 17(7):1869-1874. 25.
Rewcastle GW, Atwell GJ, Zhuang L, Baguley BC, Denny WA: Potential antitumor agents. 61. Structure-activity
relationships
for
in
vivo
colon
38
activity
among
disubstituted
9-oxo-9H-xanthene-4-acetic acids. Journal of Medicinal Chemistry 1991, 34(1):217-222. 26.
Pal C, Kundu MK, Bandyopadhyay U, Adhikari S: ChemInform Abstract: Synthesis of Novel Heme-Interacting Acridone Derivatives to Prevent Free Heme-Mediated Protein Oxidation and Degradation. ChemInform 2011, 42(45).
27.
Duvedi DR, Bahia M, Silakari O: Synthesis, cytotoxic activity, and computational analysis of N10-substituted acridone analogs. Medicinal Chemistry Research 2014, 24.
28.
Szulc Z, Palus J: Synthesis of carbocyclic derivatives of 9 (10 H)-acridinone, 9H-carbazole and 10 H-phenothiazine 5,5-dioxide as potential immunomodulating agents. Journal für Praktische Chemie 1988, 330.
29.
Burdette D, E Vance R: STING and the innate immune response to nucleic acids in the cytosol.
Nature immunology 2013, 14:19-26. 30.
Yi G, P Brendel V, Shu C, Li P, Palanathan S, Cheng Kao C: Single Nucleotide Polymorphisms of Human STING Can Affect Innate Immune Response to Cyclic Dinucleotides. PloS one 2013, 8:e77846.
31.
Gao P, Ascano M, Zillinger T, Wang W, Dai P, Serganov AA, Gaffney BL, Shuman S, Jones RA, Deng L et al: Structure-function analysis of STING activation by c[G(2',5')pA(3',5')p] and targeting by antiviral DMXAA. Cell 2013, 154(4):748-762.
32.
Abe T, Barber GN: Cytosolic-DNA-Mediated, STING-Dependent Proinflammatory Gene Induction Necessitates Canonical NF-κB Activation through TBK1. Journal of Virology 2014, 88(10):5328.
Conflict of interest
We declare that we have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Highlights •
We successfully obtained three human STING agonists by modification of the murine STING
agonists, and the best compound 12b displayed similar activities in both human and murine cells with the natural STING inducer (2’3’-cGAMP). •
Compound 2g, 9g, and12b were determined as potent human-STING agonists by directly binding
STING, and we profoundly discussed the Structure-Activity Relationship (SAR) of synthetic compounds. •
Compound 12b can induce faster, more powerful, and more durable responses of assorted
cytokines in a native system than 2’3’-cGAMP.