Abstract
Activating NRF2-driven transcription with non-electrophilic small molecules represents an attractive strategy to therapeutically target disease states associated with oxidative stress and inflammation. In this study, we describe a campaign to optimize the potency and efficacy of a previously identified bis-sulfone based non-electrophilic ARE activator 2. This work identifies the efficacious analog 17, a compound with a non-cytotoxic profile in IMR32 cells, as well as ARE activators 18 and 22, analogs with improved cellular potency. In silico drug-likeness prediction suggested the optimized bis-sulfones 17, 18, and 22 will likely be of pharmacological utility.
Keywords: ARE activator, NRF2, Non-electrophilic, PGK1 inhibitor, Drug-likeness
Continual oxidative insult is a characteristic of several age-related conditions, including autoimmune disease and neurodegeneration.1,2 To combat runaway inflammatory damage during oxidative challenge, the mammalian cell has evolved nuclear factor erythroid 2-related factor 2 (NFE2L2, NRF2 throughout), a transcription factor that induces a pro-protective transcriptional program of gene products containing antioxidant response element (ARE) sites within their genomic loci.3 In normal physiological states, NRF2 is continually ubiquitinated by cytoplasmic repressor and CUL3 adaptor protein, Kelch-like ECH-associated protein 1 (KEAP1), which promotes its proteasomal degradation. However, in response to oxidative challenge, several nucleophilic cysteine sensor residues within KEAP1 can be oxidized or alkylated, resulting in the accumulation and transcriptional activity of NRF2.4
Forced activation of NRF2 has been shown to be ameliorative in numerous animal models of disease. Consequently, an expansive set of small molecules that activate NRF2 have been described in the literature.5–10 Most of these molecules act by covalently modifying key sensor cysteines on KEAP1. Although selective covalent drugs have been used to great effect clinically,11 NRF2-activating molecules, such as the clinical candidate bardoxolone methyl, are unselective12 and cytotoxic13 and have failed to display durable clinical efficacy. Indeed, no approved drug that activates NRF2 as its primary mechanism is available, necessitating the development of alternative strategies to activate this central protective transcriptional program.
In addition to responding to exogenously derived oxidative stressors, KEAP1 also responds to altered levels of endogenous metabolites. For example, the intrinsically reactive mitochondrial metabolites, fumarate14 and itaconate15, covalently modify KEAP1 residues and alter cellular physiology by activating NRF2. As such, one potential mechanism for pharmacologically activating NRF2 involves co-opting natural signaling mechanisms by augmenting the levels of naturally occurring KEAP1-reactive metabolites. We recently described the identification of CBR-470–1, a bis-sulfone containing small molecule that inhibits the glycolytic enzyme phosphoglycerate kinase (PGK1).16 Decreased flux through glycolysis in this context results in the accumulation of a glycolytic degradation product methylglyoxal (MGO), a dicarbonyl that modifies cysteine and arginine residues to crosslink adjacent KEAP1 molecules.16 Despite altering a central metabolic pathway, CBR-470–1 treatment reportedly promotes cellular survival in several models of oxidative challenge.17,18
In an initial attempt to characterize bis-sulfone series, we recently identified non-cytotoxic and potent activators of ARE transcriptional activity, indicating that targeting PGK1 through this mechanism does not result in obligate cytotoxicity to cells.18 One plausible explanation for this observation comes from the metabolism of molecules like 2, which like other molecules of this series, including 1, is oxidatively dealkylated by CYP enzymes, yielding 3 (Fig. 1). 3 displays considerably decreased potency in activating NRF2 and is considerably more cytotoxic than other analogs. As such, optimizing the substituents of the aminoalkyl moiety may afford analogs with decreased metabolism and improved cellular activity. To further explore this hypothesis and to improve the pharmacological properties of the scaffold, an additional optimization of 2 was performed.
Fig. 1.

Fine-tuning strategy for 3-aminotetrahydrothiophene 1,1-dioxide-based NRF2 activators.
Linkage-modified analogs 10 and 12 were successfully obtained following the strategy outlined in Scheme 1. For the efficient synthesis of the desired compounds, the common intermediate 5 was constructed first by the Michael addition of isobutylamine with commercially available methyl 3-sulfolene-3-carboxylate (4). Protection of the secondary amine of 5 with a Boc group, followed by reduction of the protected methyl ester 6 afforded aldehyde 7. Compound 7 reacted with commercial 3,4-dichlorophenylmagnesium bromide to provide secondary alcohol 8, which was subsequently oxidized by Dess–Martin oxidation to obtain ketone 9. Lastly, deprotection of 9 with TFA successfully produced the desired ketone 10. Amide derivative 12 was derived from the same common intermediate 5. After hydrolysis of 5 and subsequent HATU coupling of the resulting acid 11 with 3,4-dichloroaniline in the presence of Hünig’s base, the final compound 12 was readily synthesized.
Scheme 1.

Synthesis of analogs 10 and 12. Reagents and conditions: (a) i-BuNH2, CH3CN, rt, 91%; (b) Boc2O, DMAP, CH2Cl2, rt, 92%; (c) DIBAL-H, CH2Cl2, −78 °C, 90%; (d) 3,4-dichlorophenylmagnesium bromide, THF, 0 °C; (e) DMP, CH2Cl2, 0 °C, 46% for 2 steps; (f) TFA, CH2Cl2, rt, 73%; (g) LiOH·H2O, THF/H2O (2:1), 0 °C, 95%; (h) 3,4-dichloroaniline, HATU, i-Pr2NEt, DMF, rt, 35%.
Subsequent structural modifications focused on the metabolically labile aminoalkyl groups of 2. The synthesis of bis-sulfone analogs 17–28 was achieved according to our previously established procedure, as depicted in Scheme 2.18 Briefly, commercially available 3-sulfolene (13) was reacted with 3,4-dichlorobenzenethiol in the presence of N-bromosuccinimide (NBS) to produce β-bromosulfide 14. Treatment of 14 with pyridine in refluxing CH2Cl2 gave the dehydrobrominated product 15. Consecutive mCPBA oxidation of 15, followed by conjugate addition with diverse amines under basic or neutral conditions, successfully afforded the desired bis-sulfones 17–28.
Scheme 2.

Synthesis of analogs 17–28. Reagents and conditions: (a) 3,4-dichlorobenzenethiol, NBS, CH2Cl2, rt, 50%; (b) pyridine, CH2Cl2, 70 C, 95%; (c) mCPBA, CH2Cl2, rt; (d) 3,3,3-trifluoropropylamine, 1,4-diaminobutane, or HMDA, CH3CN, rt, 21–78% for 2 steps (17, 23, and 24); (e) β-alanine, 1 N NaOH, CH3CN, rt, 30% for 2 steps (18); (f) γ-aminobutyric acid or aminocaproic acid, 2 N NaOH, CH3CN, rt, 16–23% for 2 steps (19 and 20); (g) 3-amino-2,2-difluoropropanoic acid hydrochloride, β-alanine methyl ester hydrochloride, 3-aminopropanamide hydrochloride, 2-aminoethanesulfonic acid, 2-aminoethanesulfonamide hydrochloride, or 2-aminoethylphosphonic acid, i-Pr2NEt, CH3CN, rt, 11–29% for 2 steps (21, 22, and 25–28).
The fourteen compounds 10, 12, and 17–28 obtained from the described synthetic route were evaluated for potency (EC50) and efficacy (Emax) at inducing NRF2-dependent ARE-LUC reporter activity in IMR32 cells (Table 1). The cytotoxicity of the analogs against IMR32 cells after 24 h of treatment was also evaluated using a CellTiter-Glo-based assay. First, the effect of altering the linkage between the sulfolane and the aromatic ring was evaluated to assess the essentiality of the bis-sulfone core structure. As expected, the replacement of sulfone with a ketone displayed decreased potency and efficacy of ARE induction (Table 1, 10). A bioisosteric change of the sulfone to an amide also resulted in complete loss of ARE-LUC activity (Table 1, 12).19,20 Together, these brief data confirm that the bis-sulfone core is essential for ARE activation.
Table 1.
ARE-LUC inducing activity and cytotoxicity of compounds 10, 12, and 17–28.
| Comp. | ARE-LUC EC50 [μM]a (95% CI) | ARE-LUC Emax [FI]b | IMR32 cytotoxicity IC50 [μM]c |
|---|---|---|---|
| 2 | 3.6 (0.9–5.6) | 20.0 | 5.6 |
| 10 | 4.5 (0.9–5.8) | 5.6 | 6.8 |
| 12 | > 20 | 1.7 | > 20 |
| 17 | 1.5 (1.0–4.1) | 70.7 | > 20 |
| 18 | 0.4 (0.3–2.0) | 61.5 | 5.2 |
| 19 | > 20 | 10.4 | > 20 |
| 20 | > 20 | 3.6 | > 20 |
| 21 | 2.9 (0.8–5.7) | 31.2 | 1.3 |
| 22 | 0.4 (0.3–0.5) | 31.9 | 7.4 |
| 23 | 0.9 (0.4–1.8) | 37.3 | 8.0 |
| 24 | 0.9 (0.8–3.1) | 38.5 | 7.4 |
| 25 | 0.9 (0.4–1.3) | 53.3 | 10.0 |
| 26 | 4.2 (2.1–5.6) | 18.4 | > 20 |
| 27 | 1.9 (1.6–5.1) | 27.5 | > 20 |
| 28 | 4.2 (1.4–4.8) | 43.0 | 0.04 |
EC50 values are the mean of three experiments and correspond to the concentration resulting in half-maximal induction for each compound.
FI, fold induction relative to a DMSO neutral stimulation control.
IC50 values are the mean of three experiments and correspond to the concentration of each compound which results in 50% cytotoxicity.
The effect of altering the substituents on the aminoalkyl group was then analyzed (Table 1, 17–28). The designed analogs generally showed similar or enhanced potency compared to 2, except for compounds 19 and 20. The most efficacious analog was 17, a 3,3,3-trifluoropropylamine substituted compound, demonstrating 3.5 times higher FI (70.7) than that of 2. Of note, 17 displayed no cytotoxicity toward IMR32 cells (< 20 μM). Replacement of the isobutylamine group of 2 with β-alanine resulted in an approximately 10-fold improvement of ARE-inducing potency (Table 1, 18). In contrast, substitution with γ-aminobutyric acid or aminocaproic acid led to the inability to activate ARE-driven transcription (Table 1, 19 and 20). For terminal carboxylic acids, an optimal alkyl chain length seems to exist. Among the tested compounds, five analogs, 18 and 22–25, possessed submicromolar potency for ARE activation. 22 is anticipated to be hydrolyzed to the carboxylic acid 18 in cells, which we attribute to the observed equipotency of these analogs. Like 17, the active analogs 26 and 27 also had no cytotoxic potential below 20 μM. Together, these data support the notion that cytoprotective NRF2 activation can be induced in cells by modulating PGK1 activity without obligate cytotoxicity.18
To investigate the likelihood of the bis-sulfones as candidates for therapeutic development, the most potent and efficacious compounds 17, 18, and 22 were evaluated using the web-based SwissADME tool for their in silico physicochemical and pharmacokinetic properties and drug-likeness (Table 2).21 The calculated scores forecasted that compounds 17, 18, and 22 possess favorable drug-likeness. They contain 7–8 hydrogen bond acceptors (HBA), 0 or 1 hydrogen bond donor (HBD), and 6–7 rotatable bonds (RB), which are within the range of lead-likeness criteria (HBA ≤ 9, HBD ≤ 5, and RB ≤ 10).22 Then, the topological polar surface area (TPSA) value and Abbot bioavailability score (ABS) were evaluated.23 The tested analogs met the criteria of TPSA (≤ 140 Å), and their ABS was determined as 0.55, indicating a high potential of oral bioavailability.23–25 The investigated compounds had reasonable physicochemical properties of ClogP and LogS values within the lead-likeness range22 and were moderately soluble (MS) based on Ali solubility class. Moreover, the analogs were estimated to have a high plausibility of gastrointestinal absorption (GA) and to not be p-glyco-protein substrates. Plotting these results within Bioavailability Radar plots enabled the visualization of these properties (Fig. 2). The optimal range for six physicochemical properties, including lipophilicity (LIPO, 0.7 < XLOGP3 < +5.0), size (SIZE, 150 < MW < 500 g/mol), polarity (POLAR, 20 < TPSA < 130 Å), solubility (INSOLU, 6 < log S < 0), saturation (INSATU, 0.25 < Fsp3 < 1), and flexibility (FLEX, 0 < rotatable bonds < 9), are depicted as the pink area. Although only the polarity of 18 was slightly out of the radar range, the plots of all compounds were constrained within the hexagonal area and thus considered to be drug-like.24 Lastly, analogs 17, 18, and 22 were evaluated for drug-likeness (Lipinski and Veber) and Pan Assay Interference Structures (PAINS).27,28 No such structural alerts were found in these analogs.
Table 2.
SwissADME physicochemical, pharmacokinetic, and drug-likeness prediction for compounds 17, 18, and 22.
| Comp. | HBA and HBDa | RBb | TPSAc (Å2) | ABSd | CLogPe | Ali LogSf | Ali class | GAg | Pgph | Drug-likeness (# viol.)i | PAINS |
|---|---|---|---|---|---|---|---|---|---|---|---|
| 17 | 8 and 1 | 6 | 97.07 | 0.55 | 3.32 | −5.00 | MS | High | No | Yes(0) | 0 alerts |
| 18 | 7 and 2 | 7 | 134.37 | 0.55 | 1.13 | −1.38 | MS | High | No | Yes (0) | 0 alerts |
| 22 | 7 and 1 | 7 | 123.37 | 0.55 | 2.09 | −3.84 | MS | High | No | Yes (0) | 0 alerts |
HBA: number of hydrogen bond acceptors; HBD: number of hydrogen bond donors.
Number of rotatable bonds.
Topological polar surface area.
Abbot bioavailability score.
Consensus LogP calculated by average of iLOGP, XLOGP3, WLOGP, MLOGP, and SILICOS-IT Log P.
Ali topographical method LogS.26
Gastrointestinal absorption.
P-glycoprotein substrates.
Number of violations of Lipinski and Veber filters.
Fig. 2.

Bioavailability Radar plot of compounds 17, 18, and 22.
In summary, a series of 3-aminotetrahydrothiophene 1,1-dioxides was designed, synthesized, and biologically evaluated as a part of campaign to efficiently target NRF2 with non-electrophilic small molecules. The systematic studies on scaffold modification confirmed that the sulfone linkage is essential for ARE-inducing activity. Structural fine-tuning of the aminoalkyl moiety of 2 allowed us to identify the most efficacious and potent analogs 17, 18, and 22 in this chemical series. The drug-likeness of these advanced analogs was evaluated using the in silico SwissADME tool, and the results suggested that these analogs likely provide a chemical basis for future preclinical development, providing a roadmap for further optimization of this series and the chemical matter for in vivo investigation of therapeutic activity.
Supplementary Material
Acknowledgments
This research was supported by Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare (HN21C0153000021 to HY; HN22C0120000022 to HY) and NIH (GM146865 to MJB; AG046495 to RLW).
Footnotes
Declaration of Competing Interest
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.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.bmcl.2023.129306.
Data availability
No data was used for the research described in the article.
References
- 1.Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. Clinical Interventions in Aging. 2018;13:757–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sies H, Berndt C, Jones DP. Oxidative Stress. Annual Review of Biochemistry. 2017;86: 715–748. [DOI] [PubMed] [Google Scholar]
- 3.Tonelli C, Chio IIC, Tuveson DA. Transcriptional Regulation by Nrf2. Antioxidants & Redox Signaling. 2018;29(17):1727–1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ma Q Role of nrf2 in oxidative stress and toxicity. Annual Review of Pharmacology and Toxicology. 2013;53:401–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hur W, Gray NS. Small molecule modulators of antioxidant response pathway. Current Opinion in Chemical Biology. 2011;15(1):162–173. [DOI] [PubMed] [Google Scholar]
- 6.Wu KC, McDonald PR, Liu J, Klaassen CD. Screening of natural compounds as activators of the keap1-nrf2 pathway. Planta Medica. 2014;80(1):97–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang Y, Talalay P, Cho CG, Posner GH. A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(6): 2399–2403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kumar V, Kumar S, Hassan M, et al. Novel chalcone derivatives as potent Nrf2 activators in mice and human lung epithelial cells. Journal of Medicinal Chemistry. 2011;54(12):4147–4159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wattenberg LW, Bueding E. Inhibitory effects of 5-(2-pyrazinyl)-4-methyl-1,2-dithiol-3-thione (Oltipraz) on carcinogenesis induced by benzo[a]pyrene, diethylnitrosamine and uracil mustard. Carcinogenesis. 1986;7(8):1379–1381. [DOI] [PubMed] [Google Scholar]
- 10.Rosarda JD, Baron KR, Nutsch K, et al. Metabolically Activated Proteostasis Regulators Protect against Glutamate Toxicity by Activating NRF2. ACS Chemical Biology. 2021;16(12):2852–2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nature Reviews. Cancer 2009;9(1):28–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yore MM, Kettenbach AN, Sporn MB, Gerber SA, Liby KT. Proteomic analysis shows synthetic oleanane triterpenoid binds to mTOR. PLoS One1. 2011;6(7):e22862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hermann C, Lang S, Popp T, et al. Bardoxolone-Methyl (CDDO-Me) Impairs Tumor Growth and Induces Radiosensitization of Oral Squamous Cell Carcinoma Cells. Frontiers in Pharmacology. 2020;11, 607580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ooi A, Wong JC, Petillo D, et al. An antioxidant response phenotype shared between hereditary and sporadic type 2 papillary renal cell carcinoma. Cancer Cell. 2011;20 (4):511–523. [DOI] [PubMed] [Google Scholar]
- 15.Mills EL, Ryan DG, Prag HA, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556(7699):113–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bollong MJ, Lee G, Coukos JS, et al. A metabolite-derived protein modification integrates glycolysis with KEAP1-NRF2 signalling. Nature. 2018;562(7728):600–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Liang J, Zhang XY, Zhen YF, et al. PGK1 depletion activates Nrf2 signaling to protect human osteoblasts from dexamethasone. Cell Death & Disease. 2019;10(12):888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jo J, Ibrahim L, Iaconelli J, et al. Discovery and SAR studies of 3-amino-4-(phenyl-sulfonyl)tetrahydrothiophene 1,1-dioxides as non-electrophilic antioxidant response element (ARE) activators. Bioorganic Chemistry. 2021;108, 104614. [DOI] [PubMed] [Google Scholar]
- 19.Yang H, Zhang R, Li Z, Maienfisch P, Xu X. Design, synthesis and nematicidal activitives of trifluorobutene amide derivatives against Meloidogyne incognita. Bioorganic & Medicinal Chemistry Letters. 2021;40, 127917. [DOI] [PubMed] [Google Scholar]
- 20.Cao X, Yang H, Liu C, Zhang R, Maienfisch P, Xu X. Bioisosterism and Scaffold Hopping in Modern Nematicide Research. Journal of Agricultural and Food Chemistry. 2022. [DOI] [PubMed] [Google Scholar]
- 21.Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports. 2017;7:42717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hann MM, Oprea TI. Pursuing the leadlikeness concept in pharmaceutical research. Current Opinion in Chemical Biology. 2004;8(3):255–263. [DOI] [PubMed] [Google Scholar]
- 23.Martin YC. A bioavailability score. Journal of Medicinal Chemistry. 2005;48(9): 3164–3170. [DOI] [PubMed] [Google Scholar]
- 24.Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW, Kopple KD. Molecular properties that influence the oral bioavailability of drug candidates. Journal of Medicinal Chemistry. 2002;45(12):2615–2623. [DOI] [PubMed] [Google Scholar]
- 25.Toolabi M, Moghimi S, Bakhshaiesh TO, et al. 6-Cinnamoyl-4-arylaminothienopyrimidines as highly potent cytotoxic agents: Design, synthesis and structure-activity relationship studies. European Journal of Medicinal Chemistry. 2020; 185. [DOI] [PubMed] [Google Scholar]
- 26.Ali J, Camilleri P, Brown MB, Hutt AJ, Kirton SB. In Silico Prediction of Aqueous Solubility Using Simple QSPR Models: The Importance of Phenol and Phenol-like Moieties. Journal of Chemical Information and Modeling. 2012;52(11):2950–2957. [DOI] [PubMed] [Google Scholar]
- 27.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews. 2001;46(1–3):3–26. [DOI] [PubMed] [Google Scholar]
- 28.Baell J, Walters MA. Chemistry: Chemical con artists foil drug discovery. Nature. 2014;513(7519):481–483. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
No data was used for the research described in the article.
