Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 May 15.
Published in final edited form as: Bioorg Med Chem Lett. 2022 Mar 19;64:128696. doi: 10.1016/j.bmcl.2022.128696

Development of BRD4 inhibitors as anti-inflammatory agents and antidotes for arsenicals

Marina Fosso Yatchang a, Bini Mathew a, Ritesh K Srivastava b, Jasim Khan b, Suhail Muzaffar b, Sixue Zhang a, Mousheng Wu a, Ling Zhai a, Pedro Ruiz a, Anupam Agarwal b,c, James R Bostwick a, Mark J Suto a, Mohammad Athar b,*, Corinne E Augelli-Szafran a,*
PMCID: PMC9017782  NIHMSID: NIHMS1791901  PMID: 35318165

Abstract

Arsenicals belong to the class of chemical warfare agents known as vesicants, which are highly reactive, toxic and cause robust inflammatory response. Cutaneous exposure to arsenicals causes a wide range of systemic organ damage, beginning with cutaneous injuries, and later manifest multi-organ damage and death. Thus, the development of suitable antidotes that can effectively block injury following exposure to these agents is of great importance. Bromodomain 4 (BRD4), a member of the bromodomain and extra terminal domain (BET) family, plays crucial role in regulating transcription of inflammatory, proliferation and cell cycle genes. In this context, the development of potent small molecule inhibitors of BRD4 could serve as potential antidotes for arsenicals. Herein, we describe the synthesis and biological evaluation of a series of compounds.

Keywords: arsenicals, antidotes, Bromodomain 4, inflammation

Graphical Abstract

graphic file with name nihms-1791901-f0001.jpg


Arsenicals are a series of highly toxic arsenic compounds that are built around a chloroarsine molecule (Figure 1). Some arsenicals have been developed as organic compounds and used as chemical warfare agents including Lewisite, methyldichloroarsine, ethyldichloroarsine, diphenylchloroarsine, and diphenylcyanoarsine (Figure 1).13 As vesicants, these arsenicals cause painful irritation and blistering of the skin, eyes, and respiratory system and may manifest delayed deleterious effects to human health.4,5 We have recently developed animal models to define their pathogenesis.6 Since these chemicals are highly toxic and cannot be used under usual laboratory conditions for investigating their mechanism of action. We also developed a surrogate arsenical, phenylarsine oxide (PAO), which is less toxic and can be used under safety regulation available in laboratories.7 We showed that PAO manifests similar pathophysiological alterations in the skin and other organs that are caused by other warfare arsenicals, however, at different dose regimens.79 Thus, similar to lewisite, it also causes an early robust inflammatory response in the skin, which follows blistering and severe cutaneous injury.7 Although efforts to cease the use of these agents have been put forth by many countries, stockpiles of chemical weapons still exist and have recently been used against civilian populations.10 Their accidental exposure has also been reported.11,12 The only approved medical countermeasure against arsenical-induced toxicity is 2,3-dimercapto-1-propanol or British Anti-Lewisite (BAL), which works topically and systemically as a chelating agent of arsenic.4 However, its related toxicity and low therapeutic index are considerable limitations that demand the development of more effective and less toxic antidotes.13

Figure 1.

Figure 1.

Examples of arsenicals use as chemical warfare agents

The bromodomain-containing protein 4 (BRD4) is a member of the bromodomain and extra-terminal (BET) protein family. As such, its structure consists of two bromodomains that are arranged in a conserved bundle of four α-helices and connected by two loop segments. Each bromodomain comprises an active acetyl-lysine-binding pocket allowing for the specific recognition of ε-N-acetylated lysine residues on target proteins, including histones.14 Role of BRD4 has been shown in various pathophysiological processes. In this regard, BRD4 was particularly shown to play key role in cell proliferation and inflammation.15 More recently, its involvement in complex inflammatory disorders such as sepsis and COVID-19 are revealed.16,17 BRD4 has been shown to play an important role in the regulation of inflammatory genes.18,19 As a result, inhibiting the activity of BRD4 has served as a strategy to suppress inflammation,2022 which also can be measured by a decrease of interleukin-6 (IL-6), cytokine produced in response to tissue damage and inflammation.23 The inflammatory response caused by exposure to arsenicals is highly complex and is associated with induction of various cytokines and chemokines including IL-6.6,24,25 In light of these observations, it becomes apparent that developing BRD4 inhibitors to attenuate arsenical-induced inflammation could pave the way for the development of a new class of arsenical antidotes. BRD4 is a druggable target and a wide range of its inhibitors have recently been synthesized. Some of these molecules have moved for clinical trials against Myelofibrosis, Multiple Myeloma, Neuroblastoma, Prostate Adenocarcinoma, Acute myeloid leukemia, Non-Hodgkin Lymphoma, etc.26

2-Methoxy-N-(3-methyl-2-oxo-1, 2, 3,4-tetrahydroquinazolin-6-yl)benzenesulfonamide (PFI-1, 1) is a potent and highly selective BET bromodomain inhibitor with an IC50 value of 220 nM against BRD4 in the AlphaScreen Assay.27 Crystallographic studies have revealed that the binding mode of PFI-1 to the BRD4 active site is achieved through the quinazolinone moiety.27 The quinazolinone carbonyl and nitrogen form hydrogen bonding with the conserved asparagine N140. Furthermore, the quinazolinone moiety forms hydrophobic contacts at a hydrophobic region surrounded by residues such as V87, L92, L94, and I146 (Figure 2). Based on these studies, a series of PFI-1 analogs were synthesized by modifying the quinazolinone core, 2-methoxybenzene moiety and sulfonamide linker, and tested in the BRD4 inhibitory assay. Furthermore, active compounds were tested for their ability to inhibit IL6 activity and our results are presented here.

Figure 2.

Figure 2.

(a) Structure of PFI-1. (b) Co-crystal structure of PFI-1 and BRD4 (PDB ID: 4E96). Black and orange dashed lines indicate hydrogen bonds and hydrophobic contacts, respectively. Residues that are important for ligand binding are colored in purple. Direction that faces solvent is indicated by blue shaded area. Co-crystallized water molecules adjacent to the ligand are displayed. Hydrogen atoms have been added by Schrödinger Small Molecule Drug Discovery Suite.

In an effort to optimize the drug-like properties of the quinazolinone scaffold while retaining or improving potency, a series of analogs of the quinazolinone scaffold had been proposed. According to ligand-based in silico predictions, the parent compound 1 had moderate solubility (>10 μM) with up to three metabolic labile spots (Figure 3). Based on co-crystal structures and computational docking results, the left-hand side 2-methoxybenzene moiety, which largely resided in the solvent side, had flexible binding orientations and did not form fixed interactions at the binding site. Namely, its effect on the BRD4 inhibition potency may not be directly predicted from co-crystals or docking studies. Thus, various modifications on that moiety were experimentally explored to see whether they would retain the potency. Since the 2-methoxyl group on that moiety was a metabolic labile spot, structural modifications were focused on substituting the 2-methoxyl group to improve metabolic stability (e.g., 5a to 5n). In contrast, the right-hand side quinazolinone core was predicted to form hydrophobic contact and hydrogen bonds that were critical for ligand binding. Consistent with the docking results, analogs with the quinazolinone core shrinking to a benzimidazolinone while retaining the hydrogen bonds with residue N140 (21a and 21b) were still active in the BRD4 and IL-6 assays. Moreover, the best analog (5i) of the series was successfully co-crystallized with BRD4, which displayed the same binding pose as predicted by computational docking (for details, see section ‘Binding of compound 5i to BRD4’).

Figure 3.

Figure 3.

Brief summary of in silico predictions. Text in blue: conclusion from ligand-based solubility predictions. Text in red: conclusion from ligand-based metabolic stability predictions. Text in purple: conclusion from computational docking studies of BRD4.

Investigation of the left-hand side of PFI-1 started with the synthesis of target compounds 5a-5n as outlined in Scheme 1. Starting from the commercially available 3-methyl-1,4-dihydroquinazolin-2-one 2, treatment with potassium nitrate in the presence of concentrated sulfuric acid gave 3-methyl-6-nitro-1,4-dihydroquinazolin-2-one 3 (Scheme 1a).28 Reduction of the nitro group to the corresponding amine by hydrogenation in the presence of Raney Nickel generated the intermediate 4,28 which was subsequently treated with the corresponding sulfonyl or carbonyl chlorides to afford the quinazolinone derivatives 5a-5l.29 Target compound 5m, the N-methylated analogue of compound 5a, was obtained by methylation of intermediate 3 to give compound 6,30 which was subjected to hydrogenation to afford compound 7,28 and finally reacted with 2-fluoro-5-(trifluoromethyl)benzenesulfonyl chloride.29 Target compound 5n was prepared in two steps by the reaction of 4-(bromomethyl)benzenesulfonyl fluoride 8 with 1-ethylpiperazine to give the sulfonyl fluoride 9, followed by treatment with intermediate quinazolinone 4 (Scheme 1b).31

Scheme 1.

Scheme 1.

Synthesis of quinazolinone derivatives 5a-5n.

Target compounds 11a and 11b, which are reverse sulfonamides of PFI-1 (1) and the quinazolinone derivative 5a, respectively, were also synthesized from the commercially available 3-methyl-1,4-dihydroquinazolin-2-one 2 (Scheme 2).29 Reaction with chlorosulfonic acid afforded the sulfonyl chloride 10, which was further reacted with the corresponding anilines to give the desired compounds 11a and 11b.29

Scheme 2.

Scheme 2.

Synthesis of reverse quinazolinone sulfonamide derivatives 11a-11b.

Compounds 16a and 16b are 2,4-dioxo-quinazoline analogues of PFI-1 (1) and 5a, respectively. Their preparation started with the amidation of 2-amino-5-nitro benzoic acid 12 in presence of HATU to give compound 13,32 which upon treatment with CDI gave the cyclized intermediate 14 (Scheme 3).33 Reduction of the nitro group in the presence of iron and acetic acid gave the amine 15,34 which was reacted with the corresponding sulfonyl chlorides to give the desired compounds 16a and 16b.29

Scheme 3.

Scheme 3.

Synthesis of 2,4-dioxo-quinazoline derivatives 16a-16b.

Finally, the benzimidazolinones 21a and 21b, analogous to PFI-1 and 5a, respectively, were synthesized in four steps from starting reagent 17 (Scheme 4). Regioselective N-methylation,35 cyclization in the presence of CDI,36 acid-catalyzed nitro reduction,34 and reaction with the corresponding sulfonyl chlorides29 yielded the desired targets 21a and 21b.

Scheme 4.

Scheme 4.

Synthesis of benzimidazolinone derivatives 21a-21b.

The compounds (5a-5n, 11a-11b, 16a-16b and 21a-21b) were screened for their ability to inhibit BRD4 and IC50 results are summarized in Tables 1. JQ1, a known BRD4 inhibitor, was used as the positive control for this screening. Furthermore, active compounds from the primary assay (BRD4 Alpha Screen assay) were evaluated in vitro for their potential inhibitory activity against IL-6 (ELISA assay) and was served as the secondary assay for this study (Table 1). Additionally, selected compounds were tested for metabolic stability in rodent and human liver microsomes and for solubility (Table 2). 5i, A compound with excellent in vitro activity (BRD4 and IL6), and ADME properties advanced to pharmacokinetic (PK) studies (Table 3) and in vivo studies.

Table 1.

IC50 values of compounds 5a-5n, 11a-11b, 16a-16b and 21a-21b in the BRD4 and IL-6 assays

Compounds BRD4 IC50 (μM) IL-6 IC50 (μM) Compounds BRD4 IC50 (μM) IL-6 IC50 (μM)
5a 0.17 2.16 5l 9.60 >50
5b 0.38 4.15 5m 17.81 9.60
5c 0.38 4.95 5n 4.98 >50
5d 0.86 16.20 11a 6.23 5.91
5e 5.76 36.28 11b >40 N.T.
5f 24.5 >50 16a 3.60 5.45
5g 14.22 14.39 16b >40 N.T.
5h 0.22 0.48 21a 0.60 3.09
5i 0.33 0.83 21b 2.40 9.82
5j 0.44 1.94 PFI-1 (1) 0.041 0.43
5k 3.48 16.33 JQ-1 0.009 0.56

N.T. = not tested

Table 2.

ADME properties of compounds 5h and 5i

Compounds Microsomal Stability* (minutes) Solubility (μM) log D
MLM HLM§
5h 6.8 39.6 65.4 1.5
5i 15.4 88.8 69.9 2.5
*

Diclofenac was used as a positive control,

Estradiol (haloperidol) was used as a control standard and data were normalized,

HLM = Human Liver Microsome,

§

MLM = Mouse Liver Microsome

Table 3:

PK results of 5i

Compound IP administration
t1/2* (h) tmax (h) Cmax (ng/mL) AUClast§ (h*ng/mL)
5i 3.71 1.17 858 4438
*

t½ = half-life,

tmax = time of peak plasma concentration,

Cmax = maximum concentration,

§

AUClast = Area under the curve from the time of dosing to the last measurable concentration.

Modifications of the 2-OMe-benzene ring of PFI-1 afforded compounds with a wide range of IC50 values. While the hit compound PFI-1 remained the most potent in the BRD4 assay (IC50 = 41 nM), compounds 5a-5d and 5h-5j also displayed excellent inhibitory activities with sub-micromolar IC50 values (0.17–0.86 μM). Subtle variations on the benzene ring, as in 5a (2-F, 5-CF3; IC50 = 0.17 μM), 5c (2,3,5-triF; IC50 = 0.38 μM), 5i (3-Me, 4-F; IC50 = 0.33 μM), and 5j (4-NH2; IC50 = 0.44 μM), appeared to minimally disrupt the inhibitory potency in the BRD4 assay. Furthermore, introducing a short aliphatic chain between the benzene ring and the sulfonyl group (5d; IC50 = 0.86 μM) or simply fusing a heterocyclic ring to the benzene ring (5b; IC50 = 0.38 μM/5h; IC50 = 0.22 μM) did not make any change in the activity. Indeed, as the 2-OMe benzene ring of PFI-1 resides in the hydrophobic region, minimal substitutions or those favoring pi-pi stacking are predicted to be well tolerated. As a result, 5k (IC50 = 3.48 μM), 5l (IC50 = 9.60 μM) and 5n (IC50 = 4.98 μM), which all bear hydrophilic substitutions, only displayed moderate inhibitory activity.

When the sulfonamide linker in compound 5d (IC50 = 0.86 μM) was replaced by an amide in compound 5e, we observed around seven-fold decrease in activity (IC50 = 5.76 μM). The carbonyl group in the amide moiety thus appears to be of lesser efficacy, which was further observed in compound 5f (IC50 = 24.5 μM) and compound 5g (IC50 = 14.22 μM). For compounds 11a and 11b, sulfonamide linker in PFI-1 (IC50 = 0.041 μM) and 5a (IC50 = 0.17 μM) was replaced with a reverse sulfonamide linker and both compounds showed decreased activity (11a, IC50 = 6.23 μM and 11b. IC50 > 40 μM) by >150-fold which may have been due to the disruption of the interactions of the sulfonyl group in the BRD4 active site.

Following these modifications of the left-hand side aryl ring and the sulfonamide linker, compound 5a displayed the most effective inhibitory activity in the BRD4 assay. PFI-1 and compound 5a were thus chosen to further investigate variations of the quinazolinone moiety. When the NH group on the quinazolinone ring in compound 5a (IC50 = 0.17 μM) was converted to N-Me group as in compound 5m (IC50 = 17.81 μM), a 100-fold increase in IC50 value was observed, resulting in a significant loss of inhibitory activity. This is in accordance with the crystallographic data, which revealed that the NH group on the quinazolinone ring participates as a donor in the hydrogen bonding in the Kac site of BRD4.

Modifications of the quinazolinone moiety in PFI-1 and compound 5a by adding a carbonyl group at the C4-position to form the 2,4-dioxo-quinazoline derivatives 16a and 16b, respectively, or by completely removing the carbon atom at the C4-position to give the benzimidazolinone derivatives 21a and 21b, respectively, did not enhance the BRD4 inhibitory activity, although compound 21a still showed excellent activity. We observed a good BRD4 inhibitory activity for compound 16a (IC50 = 3.60 μM) and a complete loss of activity for compound 16b (IC50 >40 μM). Compound 21a (IC50 = 0.60 μM) exhibited excellent inhibitory activity with sub-micro molar IC50 values while compound 21b (IC50 = 2.40 μM) displayed good activity.

The selected BRD4 inhibitors were also evaluated in the cell-based IL-6 assay (Table 1) and the observation made in the BRD4 assay translated well into the IL-6 assay. PFI-1 and compounds 5a-5c, 5h-5j, and 21a, which all displayed excellent inhibitory activities with sub-molar IC50 values (0.041–0.60 μM) in BRD4 assay, also showed good to excellent efficacy in the IL-6 assay (IC50 = 0.43–4.95 μM). The only exception is compound 5d, which had an excellent IC50 value of 0.86 μM in BRD4 assay but a moderate efficacy in the IL-6 assay with IC50 value of 16.2 μM. Also, all other synthesized compounds that had IC50 values >1μM in BRD4 assay only had moderate to poor efficacy in the IL-6 assay.

Compounds 5h (BRD4 IC50 = 0.22 μM; IL-6 IC50 = 0.48 μM) and 5i (BRD4 IC50 = 0.33 μM; IL-6 IC50 = 0.83 μM) thus appeared to be the most promising compounds with sub-micro molar IC50 values in both assays. Furthermore, the ADME (Absorption, Distribution, Metabolism, and Excretion) properties (Table 2) of compound 5i compared to 5h made the former a better candidate to advance to further studies such as pharmacokinetics (PK), binding and in vivo studies.

Pharmacokinetic (PK) studies:

The compound 5i was advanced to pharmacokinetic (PK) studies to evaluate the drug-like properties in vivo. This compound was administered IP (Intraperitoneal) and the results are presented in Table 3. It exhibited a half-life (t½) of 3.7 hours with a Cmax (maximum concentration) of 858 ng/mL.

Binding of compound 5i to BRD4:

The binding of compound 5i to BRD4 was confirmed by isothermal titration calorimetry (ITC) (Figure 4a). The binding affinity (Kd) of the compound is about 1 μM. To understand the binding details of compound 5i to BRD4, the co-crystal structure of BRD4 with compound 5i was determined. Compound 5i binds to the acetyl lysine binding site on BRD4 in the structure. The quinazoline ring is sandwiched in the hydrophobic activity formed by Cys136, Phe83, Ile146, Pro82, Tyr139, Tyr97, Leu94 and Val87. Asn140 forms two hydrogen bonds with oxo and NH groups on the quinazoline ring. Sulfonamide group does not interact with any residues and is exposed to the solvent area. The fluoro methyl phenyl group forms hydrophobic interactions with Ile146, Met149 and Trp81 (Figure 4b). The location of compound 5i is similar to PFI-1 (1) in the BRD4 structure.27 The phenyl ring is shifted upward due to the location of methyl in the hydrophobic packet (Figure 4c). The ITC experiment and co-crystal structure confirmed that compound 5i specifically targets BRD4.

Figure 4.

Figure 4.

Binding affinity of 5i to BRD4 and co-crystal structure of 5i and BRD4. a) Isothermal titration calorimetry (ITC) measurement of 5i binding to BRD4. The ITC titration curves are showed in the upper panel. The reference titration curve in which BRD4 protein was titrated into buffer is above the BRD4-5i titration curve. The ITC data fitting is shown in the bottom panel. The final parameters of the analysis are shown in the box. b) The binding of compound 5i in the crystal structure of BRD4 (PDB ID 7T3F). BRD4 structure is shown in gray cartoon. Compound 5i is shown in green sticks. All residues of BRD4 involved in the binding are shown in stick. c) Comparison of PFI and compound 5i in the BRD4 crystal structure. PFI is shown in yellow stick while compound 5i is in green stick.

In vitro and in vivo evaluation of 5i in a model of arsenical-mediated cutaneous BRD4 induction and consequent inflammatory response:

To demonstrate the arsenical-induced BRD4 expression in skin keratinocytes, we treated HaCaT cells, which are immortalized human skin keratinocytes, with PAO (0.5μM, 6h). As can be seen in Fig. 5a, PAO induces nuclear accumulation of BRD4. This can also be confirmed from Western blot analysis of PAO-treated keratinocytes. Co-treatment of these cells with 5i (1μM) diminished nuclear accumulation of BRD4 as well as its expression (Figure 5a). The decrease in BRD4 was accompanied by the decrease in H4 histone acetylation at H4k5ac (Figure 5b). Furthermore, 5i co-treatment also attenuated the PAO-induced BRD4-regulated transcripts of the inflammatory genes. Data shown here, demonstrate a significant decrease in Tfe3, Nfe2l2 and Nlrp3 (Figure 5c).

Figure 5.

Figure 5.

Effects of 5i on PAO-induced BRD4 and its inflammatory target genes in HaCaT cells. (a) Immunofluorescence staining of BRD4 (green) in HaCaT cells treated with vehicle control, PAO or PAO+5i. Yellow arrows indicate nuclear localization of BRD4. Nuclei were stained with DAPI (blue). (b) Western blot analysis of BRD4 and H4K5ac. β-actin and H4 was used as endogenous loading control for BRD4 and H4k5ac respectively. (c) RT-PCR analysis of BRD4 target inflammatory genes. p<0.05*, p<0.01** and p<0.001*** show significant levels.

After demonstrating ability of 5i to target BRD4 in our in vitro assay, we confirmed its in vivo potential to inhibit PAO-induced BRD4 expression, transcription activity and regulated inflammatory responses. For this the topical treatment with 5i was done 10 minutes following challenge of Ptch1+/−/SKH1 mouse skin to PAO as shown in Figure 6a. We observed that 5i treatment attenuated PAO-induced inflammatory response dose-dependently. Significant decrease in skin bi-fold thickness, a marker of inflammation could be observed as early as 3 h following PAO challenge, which was maintained up to 6 h of observation (Figure 6b). This, in vivo decrease in cutaneous inflammation was associated with decrease in the expression of BRD4 and its H3 histone acetylation marks namely H3k9ac and H3k18ac (Figure 6c). However, H4 histone mark H4k5ac was unaffected at this time of analysis. The lower doses of 5i (0.5mg and 1.0mg) were highly effective in reducing both BRD4 expression and its histone marks as further increase in its dose did not add any significant benefit. The decrease in the BRD4-dependent inflammatory response was also confirmed by assessing the impact of 5i on the BRD4-regulated inflammatory transcripts namely Nlrp3, Nfe2l2 and Il6 (Figure 6d). A consistent and highly significant decrease in these transcription target genes suggest that our novel BRD4 inhibitor is highly effective in reducing arsenical-induced cutaneous inflammation in a BRD4-dependent manner.

Figure 6.

Figure 6.

Effects of 5i on PAO-induced BRD4 and its inflammatory target genes in mice skin. (a) Exposure paradigm showing treatment protocol. Mouse photographs showing gross changes in the skin at 6 h fallowing PAO (upper) and PAO+5i treatment (lower). (b) % Change in skin bi-fold thickness at 3 and 6 h. (c) Western blot analysis of BRD4, H3k9ac, H3k18ac and H4k5ac at 6 h. β-actin, H3 and H4 were used as endogenous controls. Histogram showing densitometry analysis of band intensity, are shown in % change. (d) RT-PCR analysis of BRD4 target inflammatory genes. p<0.05*, p<0.01**, p<0.001*** and p<0.0001**** show significant levels. N=3/sex/group.

Using in silico studies and synthetic structural modifications, we have designed and prepared a series of compounds, of which eight displayed strong BRD4 inhibitory effects with IC50 values in the sub-microlar range and two compounds exhibited IL-6 inhibition below 1 μM. Compound 5i, in particular, exhibited a potency (IC50) of 0.33 μM for BRD4 and IC50 = 0.83 μM in the IL-6 assay, and both the ITC and crystallographic studies confirmed a specific and tight interaction of 5i with BRD4. Furthermore, compound 5i was advanced to in vivo pharmacokinetic studies and evaluated in vivo for its ability to suppress inflammation resulting from exposure to arsenical. 5i was highly effective in reducing arsenical-induced BRD4 expression, nuclear localization and transcription of genes regulating cutaneous inflammation both in vitro in human skin keratinocytes and in vivo in arsenical challenged murine skin. Overall, our results from this study revealed that the active compounds can lead to chemically distinct scaffolds for the treatment of arsenical-induced inflammation.

Supplementary Material

1

Acknowledgements

We acknowledge Dr. Donghui Bao, Dr. Robert Deimler, and Raleigh Godsey from the Bioanalytical Core at Southern Research for their assistance with the ADME evaluation of compounds.

Funding

This work was supported by a grant from National institute of health (Award No. 5U54ES030246-02).

Abbreviations:

BRD4

Bromodomain 4

BET

Bromodomain and extra terminal domain

IL-6

interleukin-6

IC50

Half-maximal inhibitory concentration

CDI

Carbonyldiimidazole

HATU

1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate

ADME

Absorption, Distribution, Metabolism, and Excretion

HLM

Human Liver Microsome

MLM

MLM, Mouse Liver Microsome

PK

Pharmacokinetic

IP

Intraperetonial

Cmax

Maximum concentration

t1/2

Half-life

tmax

Time of peak plasma concentration

AUClast

Area under the curve from the time of dosing to the last measurable concentration

μM

Micromolar

ITC

Isothermal titration calorimetry

ELISA

Enzyme-linked immunoassay

PAO

phenylarsine oxide

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

Supplementary materials

Supplementary materials contain all experimental procedure.

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Animal research conducted only after appropriate ethical consideration.

References and Notes

  • 1.Flora SJ. Arsenicals: toxicity, their use as chemical warfare agents, and possible remedial measures. In: Handbook of toxicology of chemical warfare agents London: Academic Press; 2015:171–191. [Google Scholar]
  • 2.Ishizaki M, Yanaoka T, Nakamura M, Hakuta T, Ueno S, Komuro M, Shibata M, Kitamura T, Honda A, Doy M, Ishii K, Tamaoka A, Shimojo N, Ogata T, Nagasawa E, Hanaoka S. Detection of bis(diphenylarsine)oxide, diphenylarsinic acid and phenylarsonic acid, compounds probably derived from chemical warfare agents, in drinking well water. J Health Sci 2005;51(2):130–137. [Google Scholar]
  • 3.Henriksson J, Johannisson A, Bergqvist PA, Norrgren L. The toxicity of organoarsenic-based warfare agents: in vitro and in vivo studies. Arch Environ Contam Toxicol 1996;30(2):213–219. [DOI] [PubMed] [Google Scholar]
  • 4.Mcmanus J, Huebner K. Vesicants. Crit Care Clin 2005;21(4):707–718. [DOI] [PubMed] [Google Scholar]
  • 5.Ganesan K, Raza SK, Vijayaraghavan R. Chemical warfare agents. J Pharm Bioallied Sci 2010;2(3):166–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li C, Srivastava RK, Weng Z, Croutch CR, Agarwal A, Elmets CA, Afaq F, Athar M. Molecular Mechanism Underlying Pathogenesis of Lewisite-Induced Cutaneous Blistering and Inflammation: Chemical Chaperones as Potential Novel Antidotes. Am J Pathol 2016;186(10):2637–2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Srivastava RK, Li C, Weng Z, Agarwal A, Elmets CA, Afaq F, Athar M. Defining cutaneous molecular pathobiology of arsenicals using phenylarsine oxide as a prototype. Sci Rep 2016;6:34865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Srivastava RK, Muzaffar S, Khan J, Traylor AM, Zmijewski JW, Curtis LM, George JF, Ahmad A, Antony VB, Agarwal A, Athar M. Protective role of HO-1 against acute kidney injury caused by cutaneous exposure to arsenicals. Ann N Y Acad Sci 2020;1480(1):155–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Surolia R, Li FJ, Wang Z, Kashyap M, Srivastava RK, Traylor AM, Singh P, Dsouza KG, Kim H, Pittet JF, Zmijewski JW, Agarwal A, Athar M, Ahmad A, Antony VB. NETosis in the pathogenesis of acute lung injury following cutaneous chemical burns. JCI Insight 2021;6(10):e147564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rothbacher D Real-time detection of Chemical Warfare Agents at clearance decontamination levels for surface contamination: a challenge for civilian authorities. Eur. Phys. J. Plus 2021;136:519. [Google Scholar]
  • 11.Isono O, Kituda A, Fujii M, Yoshinaka T, Nakagawa G, Suzuki Y. Long-term neurological and neuropsychological complications of sulfur mustard and Lewisite mixture poisoning in Chinese victims exposed to chemical warfare agents abandoned at the end of WWII. Toxicol Lett 2018;293:9–15. [DOI] [PubMed] [Google Scholar]
  • 12.Hanaoka S, Nomura K, Wada T. Determination of mustard and lewisite related compounds in abandoned chemical weapons (Yellow shells) from sources in China and Japan. J Chromatogr A 2006;1101(1–2):268–277. [DOI] [PubMed] [Google Scholar]
  • 13.Mückter H, Liebl B, Reichl FX, Hunder G, Walther U, Fichtl B. Are we ready to replace dimercaprol (BAL) as an arsenic antidote? Hum Exp Toxicol 1997;16(8):460–465. [DOI] [PubMed] [Google Scholar]
  • 14.Chung C-w Tough DF. Bromodomains: a new target class for small molecule drug discovery. Drug Discov. Today Ther. Strat 2012;9(2–3):e111–e120. [Google Scholar]
  • 15.Gilan O, Rioja I, Knezevic K, Bell MJ, Yeung MM, Harker NR, Lam EYN, Chung CW, Bamborough P, Petretich M, Urh M, Atkinson SJ, Bassil AK, Roberts EJ, Vassiliadis D, Burr ML, Preston AGS, Wellaway C, Werner T, Gray JR, Michon AM, Gobbetti T, Kumar V, Soden PE, Haynes A, Vappiani J, Tough DF, Taylor S, Dawson SJ, Bantscheff M, Lindon M, Drewes G, Demont EH, Daniels DL, Grandi P, Prinjha RK, Dawson MA. Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science 2020;368(6489):387–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mills RJ, Humphrey SJ, Fortuna PRJ, Lor M, Foster SR, Quaife-Ryan GA, Johnston RL, Dumenil T, Bishop C, Rudraraju R, Rawle DJ, Le T, Zhao W, Lee L, Mackenzie-Kludas C, Mehdiabadi NR, Halliday C, Gilham D, Fu L, Nicholls SJ, Johansson J, Sweeney M, Wong NCW, Kulikowski E, Sokolowski KA, Tse BWC, Devilée L, Voges HK, Reynolds LT, Krumeich S, Mathieson E, Abu-Bonsrah D, Karavendzas K, Griffen B, Titmarsh D, Elliott DA, McMahon J, Suhrbier A, Subbarao K, Porrello ER, Smyth MJ, Engwerda CR, MacDonald KPA, Bald T, James DE, Hudson JE. BET inhibition blocks inflammation-induced cardiac dysfunction and SARS-CoV-2 infection. Cell 2021;184(8):2167–2182.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H, White J, Kirilovsky J, Rice CM, Lora JM, Prinjha RK, Lee K, Tarakhovsky A. Suppression of inflammation by a synthetic histone mimic. Nature 2010;468(7327):1119–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Xu Y, Vakoc CR. BRD4 is on the move during inflammation. Trends Cell. Biol 2014;24(11):615–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Khan YM, Kirkham P, Barnes PJ, Adcock, IM. Brd4 is essential for IL-1β-induced inflammation in human airway epithelial cells. PLOS One 2014;9(4):e95051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung C-W, Chandwani R, Marazzi I, Wilson P, Coste H, White J, Kirilovsky J, Rice CM, Lora JM, Prinjha RK, Lee K, Tarakhovsky A. Suppression of inflammation by a synthetic histone mimic. Nature 2010;468(7327):1119–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hajmirza A, Emadali A, Gauthier A, Casasnovas O, Gressin R, Callanan MB. BET family protein BRD4: an emerging actor in NFκB signaling in inflammation and cancer. Biomedicines 2018;6(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Dey A, Yang W, Gegonne A, Nishiyama A, Pan R, Yagi R, Grinberg A, Finkelman FD, Pfeifer K, Zhu J, Singer D, Zhu J, Ozato K. BRD4 directs hematopoietic stem cell development and modulates macrophage inflammatory responses. EMBO J 2019;38(7):e100293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tanaka T, Narazaki M, Kishimoto T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harb Perspect Biol 2014;6:a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Manzoor S, Mariappan N, Zafar I, Wei CC, Ahmad A, Surolia R, Foote JB, Agarwal A, Ahmad S, Athar M, Antony VB, Ahmad A. Cutaneous lewisite exposure causes acute lung injury. Ann N Y Acad Sci 2020;1479(1):210–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Nguon N, Cléry-Barraud C, Vallet V, Elbakdouri N, Wartelle J, Mouret S, Bertoni M, Dorandeu F, Boudry I. Time course of lewisite-induced skin lesions and inflammatory response in the SKH-1 hairless mouse model. Wound Repair Regen 2014;22(2):272–80. [DOI] [PubMed] [Google Scholar]
  • 26.Alqahtani A, Choucair K, Ashraf M, Hammouda DM, Alloghbi A, Khan T, Senzer N, Nemunaitis J. Bromodomain and extra-terminal motif inhibitors: a review of preclinical and clinical advances in cancer therapy. Future Sci OA 2019;5(3):FSO372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Picaud S, Da Costa D, Thanasopoulou A, Filippakopoulos P, Fish PV, Philpott M, Fedorov O, Brennan P, Bunnage ME, Owen DR, Bradner JE, Taniere P, O’Sullivan B, Müller S, Schwaller J, Stankovic T, Knapp S. PFI-1, a highly selective protein interaction inhibitor, targeting BET bromodomains. Cancer Res 2013;73(11):3336–3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Fish PV, Cook AS, Phillips C, Bent AF, Mills JEJ, Sciammetta N. Preparation of novel heterocyclic compounds as bromodomain inhibitors Patent WO 2013027168, 28 Feb 2013.
  • 29.Fish PV, Filippakopoulos P, Bish G, Brennan PE, Bunnage ME, Cook AS, Federov O, Gerstenberger BS, Jones H, Knapp S, Marsden B, Nocka K, Owen DR, Philpott M, Picaud S, Primiano MJ, Ralph MJ, Sciammetta N, Trzupek JD. Identification of a chemical probe for bromo and extra C-terminal bromodomain inhibition through optimization of a fragment-derived hit. J. Med. Chem 2012;55(22):9831–9837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kamino T, Namba M, Asakura T. Arylcarboxamide derivative having sulfamoyl group as long-chain fatty acid elongase (Elovl6) inhibitor, and use thereof. Patent WO 2010123139, 28 Oct 2010.
  • 31.Mahapatra S, Woroch CP, Butler TW, Carneiro SN, Kwan SC, Khasnavis SR, Gu J, Butra JK, Vetelino BC, Bellenger J, Am Ende CW, Ball ND. SuFEx activation with Ca(NTf2)2: a unified strategy to access sulfamines, sulfamates, and sulfonamides from S(VI) fluorides. Org. Lett 2010;22(11):4389–4394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Carpino LA. 1-Hydroxy-7-Azabenzotriazole - an Efficient Peptide Coupling Additive. Journal of the American Chemical Society 1993;115:4397–4398. [Google Scholar]
  • 33.Yamamoto S, Shirai J, Fukase Y, Sato A, Kono M, Tomata Y, Ochida A, Yonemori K, Oda T, Imada T, Yukawa T. Preparation of fused nitrogen heterocyclic compounds as inhibitors of retinoid-related orphan receptor (ROR)yt Patent PCT Int. Appl. 2014142255, 18 Sep 2014.
  • 34.Shen C, Wu C, Liu Y, Gong Z, Li J, Chen S. Thienodiazepine derivatives as BET bromodomain inhibitors and their preparation, pharmaceutical compositions and use in the treatment of cancer Patent PCT Int. Appl. 2019056950, 28 Mar 2019.
  • 35.Binch H, Young S, Davis C, Mortimore M, Golec J, Studley J, Robinson D, Jimenez J-M. Preparation of aminopyrimidines as kinase inhibitors Patent PCT Int. Appl. 2008128009, 23 Oct 2008.
  • 36.Fuerstner C, Ackerstaff J, Straub A, Meier H, Tinel H, Zimmermann K, Tersteegen A, Zubov D, Kast R, Schamberger J, Schaefer M, Boerngen K. Preparation of bicyclically substituted uracils useful in pharmaceuticals for treatment and/or prophylaxis of disease Patent PCT Int. Appl. 2013167495, 14 Nov 2013.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES