Next Article in Journal
Versatile Fluorine-Containing Building Blocks: β-CF3-1,3-enynes
Next Article in Special Issue
Biological Activity of Amidino-Substituted Imidazo [4,5-b]pyridines
Previous Article in Journal
Simple Detection of Pigment Red 53 as a Hazardous Substance in Cosmetic Preparation Using a Polymer Combination of Polystyrene (PS) and Polymethylmethacrylate (PMMA)
Previous Article in Special Issue
Natural Product 2-Oxokolavenol Is a Novel FXR Agonist
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of Novel and Selective G-Protein Coupled Receptor 120 (GPR120) Agonists for the Treatment of Type 2 Diabetes Mellitus

School of Pharmaceutical Sciences, Liaocheng University, 1 Hunan Street, Liaocheng 252059, China
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(24), 9018; https://doi.org/10.3390/molecules27249018
Submission received: 29 October 2022 / Revised: 11 December 2022 / Accepted: 13 December 2022 / Published: 17 December 2022
(This article belongs to the Special Issue Exploring Bioactive Organic Compounds for Drug Discovery)

Abstract

:
Diabetes mellitus (DM), a chronic metabolic disorder characterized by high blood glucose, not only poses a serious threat to human life and health, but also places an economic burden on society. Currently available antidiabetic pharmacological agents have some adverse effects, which have stimulated researchers to explore novel antidiabetic agents with different mechanisms of action. G-protein Coupled Receptor 120 (GPR120), also known as free fatty acid receptor 4 (FFAR4), which is activated by medium-chain and long-chain fatty acids, has emerged as an interesting potential target for the treatment of metabolic disorders. Herein, we designed and synthesized a series of novel GPR120 agonists based on the structure of TUG-891, which is susceptible to β-oxidation and loses its GPR120 agonistic activity in vivo. Among the designed compounds, 14d showed excellent agonistic activity and selectivity and could improve glucose tolerance in normal mice in a dose-dependent manner. In addition, the compound 14d displayed good antidiabetic effects in diet-induced obese (DIO) mice and elevated insulin levels. Molecular simulations illustrated that compound 14d could enter the active site of GPR120 and interact with ARG99, which plays an important role in GPR120 activation. Based on these observations, compound 14d may be a promising lead compound deserving of further biological evaluation and structural modifications.

1. Introduction

Diabetes mellitus (DM), a chronic metabolic disorder characterized by high blood glucose levels, not only poses a serious threat to human life and health, but also places an economic burden on society. According to the latest data from the International Diabetes Federation, approximately 537 million adults (20–79 years) worldwide have diabetes, and this number is projected to increase to 643 million by 2030 and to 783 million by 2045. In parallel, the total global health expenditure on DM is estimated at USD 966 billion [1]. Several types of drugs that reduce blood glucose levels (insulin secretagogues, insulin sensitizers, α-glucosidase inhibitors, glucagon-like peptide-1 analogs, and dipeptidyl peptidase-4 inhibitors) have been developed for treating DM. However, currently available antidiabetic agents have some adverse effects, such as hypoglycemia, weight gain, gastrointestinal discomfort (disorder), nausea, diarrhea, liver function disorder, jaundice, and heart failure, which have prompted researchers to explore novel antidiabetic agents with different mechanisms of action [2].
Free fatty acid receptors (FFARs), such as GPR40 (also known as FFAR1), GPR41 (also known as FFAR3), GPR43 (also known as FFAR2), GPR119, and GPR120 (also known as FFAR4), which are activated by free fatty acids (FFAs), play key roles in regulating various physiological responses, including insulin secretion [3]. Several studies have reported that GPR41 and GPR43 are activated by short-chain FFAs [4,5,6], GPR40 is activated by medium- and long-chain FFAs [7,8], and GPR120 is activated by long-chain FFAs [9,10]. GPR120 is abundantly expressed in the intestines, lungs, adipose tissue, and pro-inflammatory macrophages and can stimulate glucagon-like peptide-1 (GLP-1) release [9,11,12]. GLP-1, a 36 amino acid peptide hormone secreted by endocrine L-cells in the small intestine and colon, plays an essential role in blood glucose regulation. In the pancreas, GLP-1 stimulates insulin secretion from β-cells in a blood glucose-dependent manner and inhibits glucagon secretion from α-cells [13,14]. Therefore, selective GPR120 agonists have therapeutic potential for the treatment of metabolic diseases, such as obesity and type 2 diabetes mellitus (T2DM).
Only a few GPR120 agonists have been reported in the literature, and no GPR120 agonists have entered clinical trials (Figure 1) [15,16,17,18,19,20,21,22,23,24,25,26]. Further research is needed to discover potent, selective, and orally bioavailable small molecule GPR120 agonists. TUG-891, with an EC50 value of 43.7 nM against human GPR120, is the first GPR120 agonist with high selectivity and activity, and has been widely used to explore the physiological function of GPR120 [15,27]. However, TUG-891 displayed high plasma clearance and a short half-life in vivo, possibly because phenylpropanoic acid moiety can undergo β-oxidation to generate benzoic acid derivatives. Therefore, we sought ways to modify the structure of TUG-891 to improve its pharmacokinetic profile, while retaining or improving GPR120 potency and selectivity. Based on a structure–activity relationship (SAR) study of TUG-891, our strategy to accomplish this goal was to replace the β-carbon with oxygen, which is the bio-isostere of carbon (Figure 2).
In this study, we first designed compounds 10a10l by replacing the β-carbon with oxygen, exchanging the position of carbon and oxygen, introducing substituents on the benzene ring, and prolonging the distance between the carboxylic acid group and benzene ring to improve the metabolic stability of the target compounds. After GPR120 agonistic activity and selectivity evaluation in vitro, compounds with good activity and selectivity were selected as lead compounds for SAR studies (14a14n) by introducing substituents on the terminal benzene ring (Figure 2). Compound 14d, with its excellent agonistic activity and selectivity in vitro, was suitable for further development. The results in rodents showed that compound 14d had low clearance and long half-life and could improve glucose tolerance in normal mice in a dose-dependent manner, decreasing blood glucose levels in diet-induced obese (DIO) mice. These results indicated that compound 14d may be a promising lead compound deserving of further biological evaluation and structural modifications.

2. Results and Discussion

2.1. Chemistry

Target compounds 10a10l were obtained according to the synthetic route summarized in Scheme 1. Substituted or unsubstituted p-hydroxybenzaldehyde 1a1c were reacted with commercially available halogenated hydrocarbons in the presence of potassium carbonate using the Williamson reaction to produce the corresponding aldehydes 2a2l in 62–78% yield. Aldehydes 2a2l were reduced by NaBH4 to give the corresponding alcohols 3a3l, which were then converted to benzyl bromides 4a4l by substituting the hydroxyl with PBr3 in 48–56% yield in two steps. Coupling of 2-bromo-5-fluorophenol 5a with benzyl bromide in the presence of K2CO3 in refluxing acetone to produce 6a in 82% yield. Suzuki coupling of 6a and 4-tolylboronic acid by treatment with Pd(PPh3)4 and Na2CO3 produced arylated adduct 7a in 75% yield, which was then deprotected with H2/Pa-C to produce phenol 8a in 77% yield. Benzyl bromides 4a4l were coupled with phenol 8a in the presence of K2CO3, followed by hydrolysis of the esters to produce the target compounds 10a10l in 51–58% yield in two steps. The structures of the target compounds 10a10l were confirmed using 1H-NMR, 13C-NMR, and high resolution mass spectrometry (HRMS).
Compound 10k, a fluorine-containing phenoxybutyric acid derivative, exhibited excellent GPR120 agonist activity and selectivity in in vitro screening. Based on the structure of 10k, fluorine-containing phenoxybutyric acid derivatives 14a14n were designed by introducing substituents on the terminal benzene ring. Target compounds 14a14n were obtained according to the synthetic route summarized in Scheme 2. Suzuki coupling of 6a and various phenylboronic acids by treatment with Pd(PPh3)4 and Na2CO3 produced the corresponding arylated adducts 11a11n in 70–76% yield, which were then deprotected by H2/Pa-C to produce phenols 12a12n in 66–78% yield. The coupling of phenols 12a12n with methyl 4-(4-(bromomethyl)-2-fluorophenoxy)butanoate under Williamson ether conditions produced esters 13a13n, which were used in the next step without further purification. The esters 13a–13n were hydrolyzed by sodium hydroxide to give the target compounds 14a14n in 53–62% yield in two steps. The structures of the target compounds 14a14n were confirmed using 1H-NMR, 13C-NMR, and HRMS.

2.2. Pharmacology

2.2.1. GPR120 Agonistic Activity and Selectivity

Compounds were screened against human GPR120 (hGPR120) transfected CHO cells using a calcium flux assay and TUG-891 was used as a positive control. We began our SAR studies by modifying the phenylpropionic acid moiety. Compound 10a, obtained by replacing the β-carbon atom of phenylpropionic acid with an oxygen atom and exchanging carbon and oxygen between benzene rings, maintained the agonistic activity of TUG-891. Compound 10b was obtained by extending the distance between the carboxyl group and the oxygen atom (n = 2), resulting in a significant decrease in agonistic activity, while compounds 10c (n = 3) and 10d (n = 4) slightly increased the agonistic activity compared to 10b. The introduction of an electron-donating methyl group at the ortho position of the benzene ring (10e) slightly reduced the agonistic activity compared to 10a, and extending the distance between carboxyl and oxygen further decreased the activity (10f, 10g, 10h vs. 10e). Introduction of the electron-withdrawing group fluorine at the ortho position of benzene ring had a beneficial effect on hGPR120 activity, as indicated by EC50 values of 77.2, 112.2, 57.6 and 96.8 nM for 10i, 10j, 10k and 10l, respectively, in the calcium flux assay. Compounds with good hGPR120 agonistic activity (EC50 < 0.2 μM) were examined for human GPR40 (hGPR40) agonistic activity. The results revealed that the EC50 of the compounds to hGPR40 was >70 µM, indicating that the compounds had excellent selectivity (Table 1). This study aimed to improve the metabolic stability of TUG-891 cells in vivo. The pharmacokinetics of compound 10k were determined in normal C57BL/6 mice, and compound 10k exhibited higher metabolic stability than TUG-891 (Figure S1, see Supporting Materials). Compound 10k, which exhibited potent hGPR120 activity, selectivity and metabolic stability, was selected for further optimization.
Compounds 14a14n were designed based on the phenoxybutanoic acid head of compound 10k by altering the substituents on the terminal benzene ring. The monomethyl substitution of the terminal benzene ring, whether at the 2-position or the 3-position, maintained the agonistic activity of compound 10k (EC50 values of 90.4 and 82.5 nM for 14a and 14b, respectively). Substitution of the 4-methyl group with fluorine significantly increased the agonistic activity, whereas that with 2-fluorine decreased the agonistic activity (EC50 values of 168.5 and 37.5 nM for 14c and 14d, respectively). Compound 14e with a methoxy group at the 4-position was fourfold less potent against hGRP120 than 4-methyl-substituted analog 10k, and the introduction of a methyl group (14f), chlorine(14g), or fluorine (14h) atom at the 2-position did not increase the agonistic activity. Compounds substituted by two of the same groups on the terminal benzene ring showed comparable agonistic activity, while the effects were weaker than that of compound 10k (EC50 values of 158.6, 150.3, 125.6, and 136.5 nM for 14i, 14j, 14k, and 14l, respectively). The introduction of an electron-withdrawing group at the 2-position of compound 10k could also reduce the agonistic activity (EC50 values of 187.3 and 312.7 nM for 14m and 14m, respectively). All results indicated that the disubstitution of the terminal benzene ring was detrimental to the agonistic activity. Compounds with good hGPR120 agonistic activity (EC50 < 0.2 μM) were examined for hGPR40 agonistic activity. The results revealed that the EC50 of the compounds to hGPR40 was >60 µM, indicating that the compounds had excellent selectivity (Table 2). The in vitro activity of compound 14d was examined on CHO cells expressing mouse GPR120 (mGPR120) and mouse GPR40 (mGPR40). The results indicated that the agonistic activity of compound 14d was slightly reduced against mGPR120 (EC50 = 83.2 nM), while the agonistic activity against mGPR40 was slightly increased (EC50 = 12.7 μM). As a result, the selectivity of compound 14d was somewhat reduced (Table 3). The dose-dependent curves of compound 14d and TUG-891 for hGPR120 and mGPR120 indicated that compound 14d was a full agonist just like TUG-891 (Figure S2 and Table S1, see Supporting Materials). Compound 14d, which exhibited potent GPR120 activity and 152.6-fold selectivity for mGPR120 over mGPR40, was selected for further evaluation in vivo.

2.2.2. Pharmacokinetic Evaluation of Compounds 10k and 14d

The pharmacokinetic profiles of 10k and 14d were obtained from C57BL/6 mice. The results showed that compounds 10k and 14d displayed excellent metabolic stability. Compound 10k obtained by replacing phenylpropionic acid with phenoxybutyric acid reached Cmax after 30 min of gavage administration at a dose of 10 mg/kg and exhibited better pharmacokinetic properties than TUG-891, indicating that 2-fluoro-substituted phenoxybutyric acid has a metabolically stable chemical structure. The 2-fluoro-substituted phenoxybutyric acid analog 14d displayed a favorable mouse pharmacokinetic profile, characterized by a higher maximum plasma concentration, longer half-life, and higher exposure, indicating that compound 14d had a suitable profile to investigate the in vivo effects (Table 4). The pharmacokinetic profiles traces were shown in the Supplementary Materials (Figure S1, see Supporting Materials).

2.2.3. Oral Glucose Tolerance Test (OGTT) of Compound 14d in Normal C57BL/6 Mice

GPR120 agonists have been reported to increase GLP-1 secretion and improve glucose tolerance in vivo [28]. Thus, the glucose tolerance effect of compound 14d was evaluated in C57BL/6 mice, and TUG-891 was selected as a positive control. Compound 14d was orally administered at doses of 3, 10, 30, and 100 mg/kg, 30 min before the oral glucose challenge at a dose volume of 3 g/kg. Glucose levels were measured from 30 min before glucose challenge to 120 min after the glucose challenge. The blood glucose levels reached a maximum after 30 min of the glucose challenge, and compound 14d exhibited a significant reduction in blood glucose at a dose of 30 mg/kg, which was superior to that of TUG-891. Furthermore, compound 14d showed a dose-dependent area for lowering glucose and overall lowering of glucose under the curve (AUC). Compound 14d at 30 mg/kg reduced plasma glucose levels with an AUC of 25% (p < 0.001) after 120 min compared with glucose alone, and no hypoglycemia was observed during the experiment (Figure 3).

2.2.4. Anti-Hyperglycemic Effects of Compound 14d in DIO Mice

Thereafter, the anti-hyperglycemic and insulinotropic effects of compound 14d were investigated in DIO mice. Compound 14d and TUG-891 were orally administered at a dose of 20 mg/kg 30 min before the oral glucose challenge at a dose volume of 2 g/kg. The results showed that the glucose AUC0–120 min of compound 14d and TUG-891-treated DIO mice were significantly lower than that of vehicle-treated mice, and compound 14d was superior to the positive control TUG-891. Additionally, insulin concentration was also tested 30 min after the glucose challenge; 14d-treated mice showed significantly increased insulin levels compared to vehicle-treated mice, which was better than TUG-891 (Figure 4). These results indicated that compound 14d could exert antidiabetic effects by promoting insulin secretion.

2.2.5. Molecular Modeling

Molecular docking studies were performed to explore the mode of binding of compound 14d to the GPR120 receptor. As the crystal structure of GPR120 has not been obtained experimentally, we first constructed the protein structure of GPR120 with homology modeling. To ensure the accuracy of homology modeling, the crystal structures of turkey β1 adrenoceptor 20 (PDB code 6IBL) [29], neurotensin receptor21 (PDB code 4XES) [30], and β2-adrenoceptor (PDB code 3P0G) [31] with good homology were selected as templates. The model structure with the lowest discrete optimized protein energy (DOPE) score was optimized and evaluated, and used for molecular docking. The docking results showed that compound 14d interacted with GPR120 in a manner similar to that of TUG-891. Compound 14d also bound excellently to the active site of GPR120 and formed hydrogen-bonding interactions with ARG99, which played a vital role in stabilizing its binding mode [32,33]. Additionally, the ring moieties of 14d exhibited several π-π and π-alkyl interactions with pocket amino acids of the GPR120 receptor, which was advantageous for stabilizing the complex formed by 14d and GPR120 (Figure 5). The docking results further indicated that compound 14d regulated blood glucose through agonist of GPR120.

3. Materials and Methods

3.1. Synthesis

All commercially available materials and reagents were used without purification unless otherwise indicated. Purification via column chromatography was performed using silica gel (200–300 mesh). The melting points of the target compounds 10a10l and 14a14n were determined using an x-5 micro melting point apparatus, which was uncorrected. The purity and characterization of the target compounds were established using a combination of high-performance liquid chromatography and NMR analytical techniques, and the purity was >95% for all test compounds. NMR spectra (500 MHz for 1H NMR and 125 MHz for 13C NMR spectra) were recorded on a Bruker AVANCE NEO 500 instrument, and were to be determined in CDCl3 or DMSO-d6. Chemical shifts were reported in ppm relative to tetramethylsilane (0.00 ppm) or solvent peaks as the internal reference. Splitting patterns are indicated as follows: s, singlet; d, doublet; t, triplet; m, multiplet. Coupling constants (J values) are given in hertz (Hz). High resolution mass spectrometry was conducted using a UPLC G2-XS QTOF spectrometer (Waters) with the electrospray ionization Fourier transform ion cyclotron resonance technique. The NMR and HRMS spectra of compounds 10a10l and 14a-14n are presented in Figures S3–S80.

3.1.1. General Synthetic Procedure for Intermediates 2a2l

A mixture of p-hydroxybenzaldehyde derivatives 1a1c (10.0 mmol), methyl bromoacetate, methyl bromopropionate, methyl bromobutyrate, or methyl bromovalerate (10.0 mmol), and K2CO3 (20.0 mmol) in DMF (15 mL) was stirred at room temperature for 12 h. The reaction was quenched with H2O (30 mL), and extracted with ethyl acetate (3 × 30 mL). The combined organic layer was washed with H2O (4 × 50 mL), and DMF dissolved in water was separated. The organic layer was dried over anhydrous Na2SO4, concentrated with rotary evaporation, and purified with silica gel column chromatography to obtain the desired compounds 2a2l.

3.1.2. General Synthetic Procedure for Intermediates 3a3l

To a solution of aldehyde derivatives 2a2l (5.0 mmol) in THF (20 mL) and MeOH (15 mL) was added borohydride (5.0 mmol) portion wise at 0 °C. The mixture was stirred at 0 °C for 1 h and quenched with 1 N HCl after completion of the reaction (TLC examination). The mixture was extracted with ethyl acetate (3 × 30 mL), and organic layers were combined and washed with H2O (2 × 30 mL) and saturated brine (2 × 30 mL) prior to drying over anhydrous Na2SO4. After filtration and concentration using a rotary evaporator under reduced pressure, a residue of 3a3l was obtained, which was used in the next step without further purification.

3.1.3. General Synthetic Procedure for Intermediates 4a4l

To the crude intermediates 4a4l (5.0 mmol) in dichloromethane (30 mL) was added phosphorus tribromide (2.5 mmol) dissolved in dichloromethane (5 mL) at −5 °C. The mixture was stirred at 0 °C for 1 h and quenched with cold water (20 mL) after the reaction completion (TLC examination). The mixture was stirred for 2 h at room temperature and extracted with dichloromethane (3 × 20 mL). The combined organic layers were washed with saturated brine (2 × 30 mL) and dried over anhydrous Na2SO4. After filtration and concentration using a rotary evaporator under reduced pressure, the residue was purified with silica gel column chromatography to obtain the intermediates 4a4l.

3.1.4. Synthetic Procedure for Intermediate 4-fluoro-4′-methyl-[1,1′-biphenyl]-2-ol (8a)

A mixture of 2-bromo-5-fluorophenol 5a (10.0 mmol), benzyl bromide (10.0 mmol), and K2CO3 (20.0 mmol) in acetone (30 mL) was stirred at 60 °C for 12 h. The reaction was cooled, and the insoluble material was filtered after the reaction completion (TLC examination). The filtrate was evaporated under reduced pressure and the residues was purified with silica gel column chromatography to obtain the intermediate 6a (2.3 g, 82%).
Intermediate 6a (5.0 mmol) and 4-tolylboronic acids (5.85 mmol) were dissolved in a mixture of 1 N sodium carbonate aq. (20 mL), ethanol (10 mL), and toluene (20 mL). After nitrogen substitution, Pd(PPh3)4 (0.5 mmol) was added as a catalyst. The reaction mixture was stirred at 80 °C under a nitrogen atmosphere for 12 h. After the reaction was complete (TLC examination), the reaction mixture was cooled, and diluted with ethyl acetate (30 mL). The insoluble material of the mixture was filtered off through celite. The organic layer of the filtrate was washed with water (2 × 30 mL) and brine (2 × 30 mL), dried over anhydrous Na2SO4, and concentrated under reduced pressure. The residue was purified with silica gel column chromatography to afford the product 7a (1.1 g, 75%) as a solid.
To a solution of 7a (2.5 mmol) in methanol was add Pd-C (0.25 mmol) as a catalyst, and the mixture was stirred under hydrogen atmosphere at room temperature for 24 h. After the reaction was complete (TLC examination), the insoluble material of the mixture was filtered off through celite. The filtrate was evaporated under reduced pressure and the residues was purified with silica gel column chromatography to obtain the intermediate 8a (0.39 g, 77%) as a solid. 1H NMR (500 MHz, CDCl3) 7.33 (d, J = 7.8 Hz, 2H), 7.29–7.25 (m, 1H), 7.14 (d, J = 7.7 Hz, 2H), 7.07 (dd, J = 11.4, 2.3 Hz, 1H), 6.81 (td, J = 8.3, 2.3 Hz, 1H), 2.30 (s, 2H).

3.1.5. General Synthetic Procedure for Target Compounds 10a10l

A mixture of 8a (1.0 mmol), intermediates 4a4l (1.0 mmol), and K2CO3 (2.0 mmol) in acetone (15 mL) was stirred at 60 °C for 12 h. The reaction was cooled, and the insoluble material was filtered after the reaction completion (TLC examination). The filtrate was evaporated under reduced pressure and the residues 9a9l were used in the next step without further purification.
To a solution of comprising compound 9a9l (1.0 mmol) in THF (10 mL), CH3OH (5 mL), and H2O (5 mL) was added NaOH solution (2 N, 2.0 mmol) at room temperature. The reaction mixture was stirred for 2 h and acidified with HCl (1 N) to a pH of 3 after hydrolysis was complete (TLC examination). The mixture was extracted with ethyl acetate (3 × 20 mL) and the combined organic layers were washed with H2O (2 × 30 mL) and saturated brine (2 × 30 mL). The organic layer was dried over anhydrous Na2SO4, concentrated by rotary evaporation, and purified with silica gel column chromatography to obtain the target compounds 10a10l.
2-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)acetic acid (10a): Colorless solid 0.20 g, yield 53% of two steps; m.p. 164~166 °C; 1H NMR (500 MHz, DMSO-d6) δ 13.01 (s, 1H), 7.36 (d, J = 7.7 Hz, 2H), 7.34–7.26 (m, 4H), 7.18 (d, J = 7.6 Hz, 2H), 7.09 (d, J = 11.3 Hz, 1H), 6.89 (d, J = 8.2 Hz, 2H), 6.84 (t, J = 8.2 Hz, 1H), 5.05 (s, 2H), 4.66 (s, 2H), 2.31 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 170.60, 163.59, 157.92, 156.82, 136.53, 134.84, 131.86, 129.56, 129.06, 126.94, 114.83, 107.69, 101.78, 70.12, 64.97, 21.18; HRMS (ES+) for C22H19FO4 (M + Na)+: calcd 389.1165; found, 389.1161.
3-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)propanoic acid (10b): Colorless solid 0.19 g, yield 51% of two steps; m.p. 120~122 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.37 (s, 1H), 7.36 (d, J = 8.0 Hz, 2H), 7.33–7.26 (m, 3H), 7.17 (d, J = 7.9 Hz, 2H), 7.09 (dd, J = 11.5, 2.4 Hz, 1H), 6.91 (d, J = 8.6 Hz, 2H), 6.84 (td, J = 8.4, 2.4 Hz, 1H), 5.05 (s, 1H), 4.16 (t, J = 6.0 Hz, 1H), 2.68 (t, J = 6.0 Hz, 1H), 2.31 (s, 2H); 13C NMR (125 MHz, DMSO-d6) δ 172.69, 163.60, 161.67, 158.52, 156.85, 136.53, 134.84, 131.83, 129.69, 129.07, 126.92, 114.78, 107.50, 101.76, 101.56, 70.19, 64.06, 34.57, 21.17; HRMS (ES+) for C23H21FO4 (M + Na)+: calcd 403.1322; found, 403.1328.
4-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (10c): Colorless solid 0.22 g, yield 57% of two steps; m.p. 106~108 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.13 (s, 1H), 7.36 (d, J = 8.0 Hz, 2H), 7.32–7.26 (m, 3H), 7.17 (d, J = 8.0 Hz, 2H), 7.09 (dd, J = 11.5, 2.4 Hz, 1H), 6.91 (d, J = 8.6 Hz, 2H), 6.84 (td, J = 8.4, 2.4 Hz, 1H), 5.05 (s, 2H), 3.97 (t, J = 6.4 Hz, 2H), 2.37 (t, J = 7.3 Hz, 2H), 2.31 (s, 3H), 1.98–1.89 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.54, 163.59, 161.66, 158.69, 156.85, 136.52, 134.85, 131.83, 129.68, 129.04, 126.93, 114.80, 107.66, 107.50, 101.77, 70.21, 67.04, 30.58, 24.71, 21.17; HRMS (ES+) for C24H23FO4 (M + Na)+: calcd 417.1478; found, 417.1480.
5-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)pentanoic acid (10d): Colorless solid 0.21 g, yield 52% of two steps; m.p. 124~126 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.03 (s, 1H), 7.36 (d, J = 8.0 Hz, 2H), 7.33–7.26 (m, 3H), 7.17 (d, J = 8.0 Hz, 2H), 7.09 (dd, J = 11.5, 2.4 Hz, 1H), 6.90 (d, J = 8.6 Hz, 2H), 6.84 (td, J = 8.4, 2.4 Hz, 1H), 5.04 (s, 2H), 3.95 (t, J = 6.2 Hz, 2H), 2.31 (s, 3H), 2.28 (t, J = 7.3 Hz, 2H), 1.77–1.69 (m, 2H), 1.68–1.60 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.82, 163.59, 161.66, 158.81, 156.85, 136.52, 134.86, 131.83, 129.67, 129.04, 128.80, 126.93, 114.78, 114.61, 107.65, 101.76, 70.22, 67.58, 33.76, 28.59, 21.69, 21.17; HRMS (ES+) for C25H25FO4 (M + Na)+: calcd 431.1635; found, 431.1637.
2-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-methylphenoxy)acetic acid (10e): Colorless solid 0.20 g, yield 52% of two steps; m.p. 128~130 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.97 (s, 1H), 7.37 (d, J = 8.0 Hz, 2H), 7.29 (dd, J = 8.3, 7.2 Hz, 1H), 7.18 (d, J = 8.0 Hz, 2H), 7.14 (d, J = 10.1 Hz, 2H), 7.08 (dd, J = 11.4, 2.4 Hz, 1H), 6.84 (td, J = 8.4, 2.4 Hz, 1H), 6.79 (d, J = 8.2 Hz, 1H), 5.01 (s, 2H), 4.68 (s, 2H), 2.32 (s, 3H), 2.17 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 170.71, 163.60, 161.66, 156.89, 156.02, 136.54, 134.87, 131.81, 130.60, 129.61, 129.07, 129.03, 127.03, 126.70, 126.34, 111.54, 107.71, 101.85, 70.27, 65.22, 21.18, 16.51; HRMS (ES+) for C23H21FO4 (M + Na)+: calcd 403.1322; found, 403.1328.
3-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-methylphenoxy)propanoic acid (10f): Colorless solid 0.20 g, yield 52% of two steps; m.p. 125~127 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.34 (s, 1H), 7.37 (d, J = 7.5 Hz, 2H), 7.29 (t, J = 7.5 Hz, 1H), 7.23–7.12 (m, 4H), 7.08 (d, J = 10.9 Hz, 1H), 6.91 (d, J = 8.1 Hz, 1H), 6.84 (t, J = 7.6 Hz, 1H), 5.01 (s, 2H), 4.16 (t, J = 5.2 Hz, 2H), 2.69 (t, J = 5.0 Hz, 2H), 2.31 (s, 3H), 2.09 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 172.75, 163.60, 161.66, 156.91, 156.59, 136.54, 134.86, 131.79, 130.51, 129.60, 129.02, 128.70, 127.01, 126.94, 126.25, 111.72, 107.69, 101.84, 70.35, 64.38, 34.75, 21.17, 16.27; HRMS (ES+) for C24H23FO4 (M + Na)+: calcd 417.1478; found, 417.1482.
4-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-methylphenoxy)butanoic acid (10g): Colorless solid 0.22 g, yield 55% of two steps; m.p. 99~101 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.13 (s, 1H), 7.41–7.34 (m, 2H), 7.29 (dd, J = 8.5, 7.0 Hz, 1H), 7.18 (d, J = 7.8 Hz, 2H), 7.14 (d, J = 8.8 Hz, 2H), 7.08 (dd, J = 11.5, 2.5 Hz, 1H), 6.88 (d, J = 8.2 Hz, 1H), 6.84 (td, J = 8.4, 2.5 Hz, 1H), 5.01 (s, 2H), 3.97 (t, J = 6.2 Hz, 2H), 2.40 (t, J = 7.3 Hz, 2H), 2.32 (s, 3H), 2.12 (s, 3H), 2.00–1.91 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.60, 156.79, 136.54, 134.87, 131.77, 130.53, 129.61, 129.03, 128.45, 126.98, 126.18, 111.41, 107.60, 101.74, 70.36, 67.11, 30.74, 24.82, 21.18, 16.40; HRMS (ES+) for C25H25FO4 (M + Na)+: calcd 431.1635; found, 431.1637.
5-(4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-methylphenoxy)pentanoic acid (10h): Colorless solid 0.24 g, yield 57% of two steps; m.p. 114~116 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.03 (s, 1H), 7.36 (d, J = 8.0 Hz, 2H), 7.28 (dd, J = 8.3, 7.1 Hz, 1H), 7.18 (d, J = 7.9 Hz, 2H), 7.14 (d, J = 10.1 Hz, 2H), 7.08 (dd, J = 11.4, 2.4 Hz, 1H), 6.88 (d, J = 8.2 Hz, 1H), 6.83 (td, J = 8.4, 2.5 Hz, 1H), 5.01 (s, 2H), 3.95 (t, J = 6.1 Hz, 2H), 2.31 (s, 3H), 2.29 (t, J = 7.3 Hz, 2H), 2.12 (s, 3H), 1.78–1.70 (m, 2H), 1.70–1.63 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.85, 163.59, 161.66, 156.92, 156.81, 136.53, 134.88, 131.79, 130.51, 129.60, 129.02, 128.35, 127.02, 126.94, 126.12, 111.45, 107.67, 101.84, 70.38, 67.66, 33.79, 28.70, 21.77, 21.17, 16.40; HRMS (ES+) for C26H27FO4 (M + Na)+: calcd 445.1791; found, 445.1792.
2-(2-fluoro-4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)acetic acid (10i): Colorless solid 0.22 g, yield 58% of two steps; m.p. 90~92 °C; 1H NMR (500 MHz, DMSO-d6) δ 13.09 (s, 1H), 7.37 (d, J = 8.0 Hz, 2H), 7.32–7.28 (m, 1H), 7.26–7.17 (m, 3H), 7.12 (d, J = 8.7 Hz, 1H), 7.11–7.03 (m, 2H), 6.86 (td, J = 8.4, 2.3 Hz, 1H), 5.06 (s, 2H), 4.77 (s, 2H), 2.32 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 170.22, 163.59, 161.65, 156.63, 152.67, 150.73, 145.72, 136.61, 134.78, 131.89, 130.53,129.59, 129.06, 127.03, 124.17, 115.94, 115.12, 107.89, 101.82, 69.52, 65.49, 21.18; HRMS (ES+) for C22H18F2O4 (M + Na)+: calcd 407.1071; found, 407.1072.
3-(2-fluoro-4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)propanoic acid (10g): Colorless solid 0.21 g, yield 54% of two steps; m.p. 94~96 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.43 (s, 1H), 7.37 (d, J = 7.9 Hz, 2H), 7.33–7.27 (m, 1H), 7.21 (d, J = 11.3 Hz, 2H), 7.16 (m, 3H), 7.09 (m, J = 11.3, 2.0 Hz, 1H), 6.86 (td, J = 8.4, 2.1 Hz, 1H), 5.06 (s, 2H), 4.23 (t, J = 6.0 Hz, 2H), 2.72 (t, J = 6.0 Hz, 2H), 2.32 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 172.51, 163.59, 161.65, 156.63, 152.72, 150.78, 146.31, 136.61, 134.76, 131.87, 130.16, 129.59, 129.05, 126.98, 124.48, 115.89, 115.19, 107.86, 101.78, 69.55, 65.24, 34.46, 21.17; HRMS (ES+) for C23H20F2O4 (M + Na)+: calcd 421.1227; found, 421.1233.
4-(2-fluoro-4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (10k): Colorless solid 0.23 g, yield 57% of two steps; m.p. 130~132 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.16 (s, 1H), 7.37 (d, J = 8.0 Hz, 2H), 7.33–7.27 (m, 1H), 7.25–7.17 (m, 3H), 7.17–7.11 (m, 2H), 7.08 (dd, J = 11.4, 2.3 Hz, 1H), 6.86 (td, J = 8.4, 2.4 Hz, 1H), 5.06 (s, 2H), 4.05 (t, J = 6.4 Hz, 2H), 2.39 (t, J = 7.3 Hz, 2H), 2.32 (s, 3H), 2.03–1.91 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.44, 163.59, 161.65, 156.65, 152.89, 150.95, 146.45, 136.61, 134.78, 131.87, 130.04, 129.59, 129.05, 127.02, 124.43, 115.86, 115.31, 107.87, 101.81, 69.59, 68.32, 30.41, 24.63, 21.17; HRMS (ES+) for C24H22F2O4 (M + Na)+: calcd 435.1384; found, 435.1385.
5-(2-fluoro-4-(((4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)pentanoic acid (10l): Colorless solid 0.24 g, yield 56% of two steps; m.p. 112~114 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.04 (s, 1H), 7.37 (d, J = 8.1 Hz, 2H), 7.30 (dd, J = 8.4, 7.1 Hz, 1H), 7.24–7.17 (m, 3H), 7.16–7.11 (m, 2H), 7.08 (dd, J = 11.4, 2.4 Hz, 1H), 5.06 (s, 2H), 4.03 (t, J = 6.3 Hz, 2H), 2.32 (s, 3H), 2.29 (t, J = 7.3 Hz, 2H), 1.82–1.70 (m, 2H), 1.70–1.61 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.80, 163.59, 161.65, 156.65, 152.87, 150.93, 146.58, 136.60, 134.79, 131.87, 129.88, 129.59, 129.05, 127.02, 124.43, 115.84, 115.23, 107.86, 101.81, 69.60, 68.84, 33.70, 28.50, 21.56, 21.17; HRMS (ES+) for C25H24F2O4 (M + Na)+: calcd 449.1540; found, 449.1548.

3.1.6. General Synthetic Procedure for Intermediates 12a12l

The intermediates 12a12n were obtained as described for intermediate 8a.
4-fluoro-2′-methyl-[1,1′-biphenyl]-2-ol (12a): 1H NMR (500 MHz, CDCl3) δ 7.33–7.30 (m, 2H), 7.27 (dd, J = 7.7, 4.5 Hz, 1H), 7.19 (dt, J = 7.1, 1.2 Hz, 1H), 7.04 (dd, J = 8.4, 6.5 Hz, 1H), 6.74–6.65 (m, 2H), 2.15 (s, 3H).
4-fluoro-3′-methyl-[1,1′-biphenyl]-2-ol (12b): 1H NMR (500 MHz, CDCl3) δ 7.36 (td, J = 7.3, 1.0 Hz, 1H), 7.22–7.17 (m, 3H), 7.15 (dd, J = 8.3, 6.6 Hz, 1H), 6.72–6.65 (m, 2H), 2.40 (s, 3H).
2′,4-difluoro-[1,1′-biphenyl]-2-ol (12c): 1H NMR (500 MHz, CDCl3) δ 7.41–7.36 (m, 1H), 7.34 (td, J = 7.5, 1.9 Hz, 1H), 7.27–7.22 (m, 1H), 7.21–7.15 (m, 2H), 6.72 (dtd, J = 9.3, 4.0, 2.6 Hz, 2H).
4,4′-difluoro-[1,1′-biphenyl]-2-ol (12d): 1H NMR (500 MHz, CDCl3) δ 7.43–7.37 (m, 2H), 7.21–7.13 (m, 3H), 6.73–6.68 (m, 2H).
4-fluoro-4′-methoxy-[1,1′-biphenyl]-2-ol (12e): 1H NMR (500 MHz, CDCl3) δ 7.36–7.31 (m, 2H), 7.14 (dd, J = 8.3, 6.5 Hz, 1H), 7.05–6.99 (m, 2H), 6.73–6.66 (m, 2H), 3.86 (s, 3H).
4-fluoro-2′,4′-dimethyl-[1,1′-biphenyl]-2-ol (12f): 1H NMR (500 MHz, CDCl3) δ 9.80 (s, 1H), 7.04 (d, J = 1.8 Hz, 1H), 7.02–6.97 (m, 2H), 6.95 (d, J = 7.7 Hz, 1H), 6.69 (dd, J = 10.9, 2.6 Hz, 1H), 6.65 (td, J = 8.5, 2.6 Hz, 1H), 2.29 (s, 3H), 2.06 (s, 3H).
4-fluoro-3′,4′-dimethyl-[1,1′-biphenyl]-2-ol (12g): 1H NMR (500 MHz, CDCl3) δ 7.23 (d, J = 7.6 Hz, 1H), 7.16 (d, J = 2.0 Hz, 1H), 7.15–7.10 (m, 2H), 6.70–6.64 (m, 2H), 2.30 (s, 6H).
4-fluoro-3′,5′-dimethyl-[1,1′-biphenyl]-2-ol (12h): 1H NMR (500 MHz, CDCl3) δ 7.12 (dd, J = 8.3, 6.5 Hz, 1H), 7.02–6.99 (m, 1H), 6.99 (d, J = 1.4 Hz, 2H), 6.69–6.62 (m, 2H), 2.33 (s, 6H).
2′,4′-dichloro-4-fluoro-[1,1′-biphenyl]-2-ol (12i): 1H NMR (500 MHz, CDCl3) δ 7.53 (d, J = 2.1 Hz, 1H), 7.33 (dd, J = 8.2, 2.1 Hz, 1H), 7.25 (d, J = 8.2 Hz, 1H), 7.11–7.06 (m, 1H), 6.75–6.67 (m, 2H).
4-fluoro-4′-methoxy-2′-methyl-[1,1′-biphenyl]-2-ol (12j): 1H NMR (500 MHz, CDCl3) δ 9.69 (d, J = 3.3 Hz, 1H), 7.67 (dd, J = 8.8, 2.7 Hz, 1H), 7.36–7.26 (m, 2H), 7.08 (d, J = 8.3 Hz, 1H), 6.85 (d, J = 2.7 Hz, 1H), 6.81 (dd, J = 8.3, 2.7 Hz, 1H), 3.85 (s, 3H), 2.08 (s, 3H).
2′-chloro-4-fluoro-4′-methoxy-[1,1′-biphenyl]-2-ol (12k): 1H NMR (500 MHz, CDCl3) δ 7.22 (d, J = 8.5 Hz, 1H), 7.11–7.06 (m, 2H), 6.91 (dd, J = 8.5, 2.6 Hz, 1H), 6.74–6.67 (m, 2H), 3.85 (s, 3H).
2′,4-difluoro-4′-methoxy-[1,1′-biphenyl]-2-ol (12l): 1H NMR (500 MHz, CDCl3) δ 7.24 (t, J = 8.6 Hz, 1H), 7.19–7.12 (m, 1H), 6.81 (dd, J = 8.5, 2.6 Hz, 1H), 6.76 (dd, J = 11.6, 2.5 Hz, 1H), 6.74–6.68 (m, 2H), 3.85 (s, 3H).
2′-chloro-4-fluoro-4′-methyl-[1,1′-biphenyl]-2-ol (12m): 1H NMR (500 MHz, CDCl3) δ 7.35 (s, 1H), 7.20 (d, J = 7.7 Hz, 1H), 7.17 (dd, J = 7.9, 1.7 Hz, 1H), 7.10 (dd, J = 8.3, 6.5 Hz, 1H), 6.74–6.68 (m, 2H), 2.39 (s, 3H).
2′,4-difluoro-4′-methyl-[1,1′-biphenyl]-2-ol (12n): 1H NMR (500 MHz, CDCl3) δ 7.20 (t, J = 7.8 Hz, 1H), 7.17–7.13 (m, 1H), 7.04 (ddd, J = 7.7, 1.7, 0.8 Hz, 1H), 7.02–6.97 (m, 1H), 6.72–6.67 (m, 2H), 2.38 (s, 3H).

3.1.7. General Synthetic Procedure for Intermediates 13a13n

The intermediates 13a13n were obtained as described for intermediates 9a9l, which were used in the next step without further purification.

3.1.8. General Synthetic Procedure for Target Compounds 14a14n

The target compounds 14a14n were obtained as described for target compounds 10a10l.
4-(2-fluoro-4-(((4-fluoro-2′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14a): Colorless solid 0.23 g, yield 57% of two steps; m.p. 83~85 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.25 (d, J = 4.4 Hz, 2H), 7.22–7.18 (m, 1H), 7.14 (d, J = 7.7 Hz, 1H), 7.12–7.06 (m, 3H), 7.01 (t, J = 8.3 Hz, 2H), 6.85 (td, J = 8.4, 2.5 Hz, 1H), 5.03 (s, 2H), 4.02 (t, J = 6.5 Hz, 2H), 2.36 (t, J = 7.3 Hz, 2H), 2.05 (s, 3H), 1.96–1.88 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.07, 163.29, 161.35, 156.26, 152.39, 150.45, 145.92, 137.45, 136.19, 131.60, 130.04, 129.57, 127.41, 126.83, 125.51, 123.74, 115.06, 114.81, 107.06, 101.05, 68.88, 67.85, 29.99, 24.18, 19.69; HRMS (ES+) for C24H22F2O4 (M + Na)+: calcd 435.1384; found, 435.1388.
4-(2-fluoro-4-(((4-fluoro-3′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14b): Colorless solid 0.22 g, yield 54% of two steps; m.p. 87~89 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.38–7.30 (m, 2H), 7.29–7.19 (m, 3H), 7.18–7.06 (m, 4H), 6.87 (dt, J = 9.2, 4.7 Hz, 1H), 5.06 (s, 2H), 4.05 (t, J = 6.5 Hz, 2H), 2.38 (t, J = 7.2 Hz, 2H), 2.32 (s, 3H), 2.03–1.90 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.55, 163.76, 161.82, 156.68, 152.97, 151.03, 146.43, 137.46, 131.92, 130.63, 130.11, 128.45, 128.10, 127.21, 126.83, 124.39, 115.78, 115.33, 107.88, 101.80, 69.62, 68.38, 30.46, 24.66, 21.48; HRMS (ES+) for C24H22F2O4 (M + Na)+: calcd 435.1384; found, 435.1386.
4-(4-(((2′,4-difluoro-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14c): Colorless solid 0.23 g, yield 55% of two steps; m.p. 94~96 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.44–7.38 (m, 1H), 7.36 (td, J = 7.6, 1.8 Hz, 1H), 7.33–7.22 (m, 3H), 7.22–7.05 (m, 4H), 6.89 (td, J = 8.4, 2.5 Hz, 1H), 5.07 (s, 2H), 4.04 (t, J = 6.5 Hz, 2H), 2.38 (t, J = 7.3 Hz, 2H), 2.05–1.89 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.48, 164.28, 162.34, 160.89, 158.93, 157.07, 152.86, 150.92, 146.35, 132.42, 130.06, 125.39, 124.70, 124.11, 121.44, 115.74, 115.42, 115.24, 107.66, 101.55, 69.42, 68.28, 30.40, 24.60; HRMS (ES+) for C23H19F3O4 (M + Na)+: calcd 439.1133; found, 439.1130.
4-(4-(((4,4′-difluoro-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14d): Colorless solid 0.24 g, yield 57% of two steps; m.p. 89~91 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.56–7.48 (m, 1H), 7.33 (dd, J = 8.4, 6.9 Hz, 1H), 7.25–7.16 (m, 3H), 7.16–7.09 (m, 3H), 6.88 (td, J = 8.4, 2.5 Hz, 1H), 5.07 (s, 2H), 4.05 (t, J = 6.4 Hz, 2H), 2.38 (t, J = 7.3 Hz, 2H), 2.00–1.90 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.46, 163.80, 162.75, 161.86, 160.81, 156.56, 152.89, 150.95, 146.41, 133.99, 132.00, 131.70, 129.93, 126.02, 124.39, 115.73, 115.48, 107.89, 101.82, 69.65, 68.32, 30.41, 24.62; HRMS (ES+) for C23H19F3O4 (M + Na)+: calcd 439.1133; found, 439.1140.
4-(2-fluoro-4-(((4-fluoro-4′-methoxy-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14e): Colorless solid 0.22 g, yield 53% of two steps; m.p. 103~105 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.14 (s, 1H), 7.44–7.37 (m, 2H), 7.28 (dd, J = 8.4, 6.9 Hz, 1H), 7.22–7.16 (m, 1H), 7.15–7.09 (m, 2H), 7.06 (dd, J = 11.4, 2.5 Hz, 1H), 6.97–6.91 (m, 2H), 6.84 (td, J = 8.3, 2.5 Hz, 1H), 5.05 (s, 2H), 4.04 (t, J = 6.4 Hz, 2H), 3.77 (s, 3H), 2.37 (t, J = 7.3 Hz, 2H), 2.03–1.89 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.46, 163.43, 161.49, 158.80, 156.53, 152.91, 150.97, 146.39, 131.71, 130.85, 130.08, 129.94, 126.80, 124.37, 115.74, 115.36, 113.94, 107.78, 101.74, 69.57, 68.34, 55.56, 30.43, 24.64; HRMS (ES+) for C24H22F2O5 (M + Na)+: calcd 451.1333; found, 451.1334.
4-(2-fluoro-4-(((4-fluoro-2′,4′-dimethyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14f): Colorless solid 0.24 g, yield 56% of two steps; m.p. 84~86 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.09 (dd, J = 8.6, 6.7 Hz, 2H), 7.07–7.04 (m, 2H), 7.04–6.99 (m, 3H), 6.97 (d, J = 7.7 Hz, 1H), 6.83 (td, J = 8.4, 2.4 Hz, 1H), 5.01 (s, 2H), 4.03 (t, J = 6.4 Hz, 2H), 2.37 (t, J = 7.3 Hz, 2H), 2.29 (s, 3H), 2.02 (s, 3H), 1.99–1.88 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.45, 163.60, 161.67, 156.80, 152.83, 146.38, 136.83, 136.34, 134.96, 132.15, 130.50, 130.04, 127.25, 126.54, 124.31, 115.63, 115.29, 107.42, 101.44, 69.34, 68.31, 30.41, 24.62, 21.14, 20.05; HRMS (ES+) for C25H24F2O4 (M + Na)+: calcd 449.1540; found, 449.1541.
4-(2-fluoro-4-(((4-fluoro-3′,4′-dimethyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14g): Colorless solid 0.23 g, yield 53% of two steps; m.p. 116~118 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.19 (s, 1H), 7.38–7.31 (m, 2H), 7.31–7.25 (m, 1H), 7.25–7.15 (m, 4H), 7.13 (dd, J = 11.3, 2.6 Hz, 1H), 6.90 (td, J = 8.3, 2.5 Hz, 1H), 5.10 (s, 2H), 4.10 (t, J = 6.4 Hz, 2H), 2.43 (t, J = 7.3 Hz, 2H), 2.28 (s, 6H), 2.04–1.95 (m, 2H). 13C NMR (125 MHz, DMSO-d6) δ 174.45, 163.53, 161.60, 156.61, 152.93, 150.99, 146.38, 136.02, 135.20, 131.71, 130.97, 130.09, 129.59, 126.98, 124.40, 115.80, 115.30, 107.75, 101.69, 69.58, 68.35, 30.40, 24.62, 19.77, 19.50; HRMS (ES+) for C25H24F2O4 (M + Na)+: calcd 449.1540; found, 449.1545.
4-(2-fluoro-4-(((4-fluoro-3′,5′-dimethyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14h): Colorless solid 0.26 g, yield 61% of two steps; m.p. 96~98 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.31 (dd, J = 8.5, 6.9 Hz, 1H), 7.23 (d, J = 13.2 Hz, 1H), 7.14 (d, J = 6.6 Hz, 2H), 7.12–7.03 (m, 3H), 6.94 (s, 1H), 6.85 (td, J = 8.4, 2.5 Hz, 1H), 5.05 (s, 2H), 4.05 (t, J = 6.4 Hz, 2H), 2.38 (t, J = 7.3 Hz, 2H), 2.28 (s, 6H), 1.99–1.90 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.45, 163.64, 161.71, 156.64, 152.97, 151.03, 146.35, 137.46, 137.27, 131.71, 130.15, 128.77, 127.62, 127.27, 124.22, 115.67, 115.30, 107.77, 101.74, 69.55, 68.38, 30.40, 24.61, 21.32; HRMS (ES+) for C25H24F2O4 (M + Na)+: calcd 449.1540; found, 449.1543.
4-(4-(((2′,4′-dichloro-4-fluoro-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14i): Colorless solid 0.29 g, yield 62% of two steps; m.p. 109~111 °C; 1H NMR (500 MHz, DMSO-d6) δ 12.16 (s, 1H), 7.69 (d, J = 2.2 Hz, 1H), 7.46 (dd, J = 8.3, 2.2 Hz, 1H), 7.36 (d, J = 8.2 Hz, 1H), 7.22 (t, J = 7.6 Hz, 1H), 7.18–7.00 (m, 4H), 6.89 (td, J = 8.5, 2.4 Hz, 1H), 5.05 (s, 2H), 4.04 (t, J = 6.5 Hz, 2H), 2.37 (t, J = 7.4 Hz, 2H), 2.06–1.83 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.46, 164.38, 162.43, 156.93, 152.81, 150.87, 146.43, 135.93, 134.53, 133.72, 133.39, 132.23, 129.77, 129.03, 127.63, 124.30, 115.67, 115.24, 107.67, 101.60, 69.51, 68.27, 30.40, 24.60; HRMS (ES+) for C23H18Cl2F2O4 (M + Na)+: calcd 489.0448; found, 489.0450.
4-(2-fluoro-4-(((4-fluoro-4′-methoxy-2′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)phenoxy)butanoic acid (14j): Colorless solid 0.26 g, yield 58% of two steps; m.p. 87~89 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.09 (t, J = 7.9 Hz, 2H), 7.06–6.99 (m, 4H), 6.86–6.79 (m, 2H), 6.76 (dd, J = 8.4, 2.7 Hz, 1H), 5.00 (s, 2H), 4.02 (t, J = 6.4 Hz, 2H), 3.75 (s, 3H), 2.36 (t, J = 7.3 Hz, 2H), 2.03 (s, 3H), 1.92 (t, J = 6.9 Hz, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.46, 163.56, 161.63, 158.93, 156.92, 152.85, 150.91, 146.36, 138.00, 132.35, 131.46, 130.18, 130.08, 127.04, 124.20, 115.34, 111.34, 107.39, 101.44, 69.32, 68.32, 55.42, 30.42, 24.62, 20.38; HRMS (ES+) for C25H24F2O5 (M + Na)+: calcd 465.1489; found, 465.1492.
4-(4-(((2′-chloro-4-fluoro-4′-methoxy-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14k): Colorless solid 0.25 g, yield 55% of two steps; m.p. 102~104 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.23 (d, J = 8.5 Hz, 1H), 7.17 (dd, J = 8.4, 6.9 Hz, 1H), 7.15–7.10 (m, 2H), 7.10–7.02 (m, 3H), 6.95 (dd, J = 8.5, 2.5 Hz, 1H), 6.85 (td, J = 8.4, 2.5 Hz, 1H), 5.05 (s, 2H), 4.04 (t, J = 6.4 Hz, 2H), 3.79 (s, 3H), 2.38 (t, J = 7.3 Hz, 2H), 2.05–1.85 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.47, 162.10, 159.77, 157.19, 152.89, 150.95, 146.38, 134.01, 133.01, 132.53, 128.99, 124.20, 115.54, 115.32, 114.80, 113.51, 107.39, 101.51, 69.42, 68.35, 56.07, 30.43, 24.65; HRMS (ES+) for C24H21ClF2O5 (M + Na)+: calcd 485.0943; found, 485.0948.
4-(4-(((2′,4-difluoro-4′-methoxy-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14l): Colorless solid 0.26 g, yield 59% of two steps; m.p. 116~118 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.27 (d, J = 8.6 Hz, 1H), 7.25–7.22 (m, 1H), 7.15–7.10 (m, 2H), 7.10–7.03 (m, 2H), 6.90–6.84 (m, 2H), 6.82 (dd, J = 8.5, 2.6 Hz, 1H), 5.05 (s, 2H), 4.04 (t, J = 6.4 Hz, 2H), 3.79 (s, 3H), 2.37 (t, J = 7.3 Hz, 2H), 1.99–1.88 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.53, 162.10, 160.62, 159.48, 157.15, 152.91, 150.98, 146.38, 132.66, 130.05, 124.14, 121.39, 117.37, 115.53, 115.35, 110.56, 107.59, 101.90, 101.54, 69.42, 68.36, 56.12, 30.48, 24.67; HRMS (ES+) for C24H21F3O5 (M + Na)+: calcd 469.1239; found, 469.1242.
4-(4-(((2′-chloro-4-fluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14m): Colorless solid 0.27 g, yield 60% of two steps; m.p. 105~107 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.34 (s, 1H), 7.21–7.14 (m, 3H), 7.14–7.02 (m, 4H), 6.85 (td, J = 8.4, 2.5 Hz, 1H), 5.03 (s, 2H), 4.03 (t, J = 6.3 Hz, 2H), 2.37 (t, J = 7.4 Hz, 2H), 2.33 (s, 3H), 1.97–1.84 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.50, 164.15, 162.21, 157.10, 152.90, 150.96, 146.45, 139.52, 133.96, 133.17, 132.36, 129.94, 128.15, 124.99, 115.68, 107.44, 101.51, 69.47, 68.33, 30.42, 24.67, 20.85; HRMS (ES+) for C24H21ClF2O4 (M + Na)+: calcd 469.0994; found, 469.0994.
4-(4-(((2′,4-difluoro-4′-methyl-[1,1′-biphenyl]-2-yl)oxy)methyl)-2-fluorophenoxy)butanoic acid (14n): Colorless solid 0.25 g, yield 57% of two steps; m.p. 92~94 °C; 1H NMR (500 MHz, DMSO-d6) δ 7.09 (dd, J = 8.6, 6.7 Hz, 2H), 7.07–7.04 (m, 2H), 7.04–6.99 (m, 3H), 6.97 (d, J = 7.7 Hz, 1H), 6.83 (td, J = 8.4, 2.4 Hz, 1H), 5.01 (s, 2H), 4.03 (t, J = 6.4 Hz, 2H), 2.37 (t, J = 7.3 Hz, 2H), 2.29 (s, 3H), 2.02 (s, 3H), 1.99–1.88 (m, 2H); 13C NMR (125 MHz, DMSO-d6) δ 174.47, 164.14, 162.20, 160.69, 158.74, 157.10, 152.86, 150.92, 146.38, 140.11, 132.52, 129.95, 125.28, 124.22, 122.32, 121.46, 116.13, 115.60, 115.23, 107.58, 101.49, 69.42, 68.30, 30.40, 24.61, 21.03; HRMS (ES+) for C24H21F3O4 (M + Na)+: calcd 453.1290; found, 453.1295.

3.2. Pharmacology Studies

3.2.1. In Vitro GPR120 Agonistic Activity and Selectivity Studies

A calcium flux assay was performed to assess the agonist activity of the test compounds against the hGPR120 (mGPR120) and hGPR40 (mGPR40) receptors. CHO cells stably transfected with hGPR120 were seeded in a 96-well plate at a density of 20,000 cells per well and incubated for 24 h at 37 °C in a 5% CO2 incubator. The medium in the wells was gently discarded, and the wells were washed with Hank’s balanced salt solution (HBSS; 100 μL per well). Cells were then incubated in HBSS containing Fluo-4 AM (2.5 μg/mL), 0.1% fatty acid-free bovine serum albumin, and probenecid (2.5 mM) for 60 min at 37 °C. The cells were then washed thrice in HBSS and allowed to equilibrate for 10 min before conducting the assay. Test compounds dissolved in dimethyl sulfoxide were diluted in HBSS at various concentrations and added to Fluo-4 AM-containing cells, and intracellular Ca2+ concentrations were measured using FlexStation3 Molecular Devices. The EC50 value of each compound was calculated using GraphPad Prism software (version 5.0; GraphPad Software, San Diego, CA, USA). The calcium influx assay of the target compounds in hGPR40-expressing CHO cells was similar to that of hGPR120.

3.2.2. Animals

Male C57BL/6 mice, eight-week aged, were purchased from Jinan Pengyue Experimental Animal Breeding Co., Ltd. (Jinan, China). The mice were housed in cages under a 12 h light/dark cycle from 7:00 to 19:00 at controlled temperatures (25–26 °C) and relative humidity (50 ± 10%) throughout the experimental period. All animals were allowed to eat and drink freely unless otherwise stated, and were allowed to acclimatize for 1 week before the experiment. All animal experimental protocols were performed following applicable institutional and governmental regulations concerning the ethical use of animals.

Pharmacokinetic Analysis of Compounds 10k and 14d in C57BL/6 Mice

Pharmacokinetic studies of compounds 10k and 14d were performed in male C57BL/6 mice, and TUG-891 was used as a positive control. Male C57BL/6 mice weighing 28–32 g were starved for 12 h and randomly divided into three groups (four mice per group). Compounds 10k, 14d, and TUG-891 were dissolved in 0.5% methylcellulose (0.5% MC) at a concentration of 1 mg/mL, and gavage was administered at a volume of 10 mL/kg. Blood samples were collected over a 24 h period post-dose into tubes containing EDTA-K2, and plasma was separated with centrifugation at 5.645 g for 10 min. Plasma was collected and precipitated using two volumes of acetonitrile containing an internal standard. This was followed by centrifugation at 15.680 g for 10 min after vortexing for 5 min. The supernatant was diluted with acetonitrile and 10 µL of the supernatant was analyzed by Waters LC-PDA-MS/MS to determine plasma drug levels. Pharmacokinetic parameters were determined using mean data from four mice at each time point. Statistical analysis of the data was performed using the DAS 2.1.1 statistical software program (BioVoice, Shanghai, China).

OGTT in Normal C57BL/6 Mice

Normal male C57BL/6 mice aged 9 weeks were used for the OGTT for compound 14d. C57BL/6 mice were weighed and randomly divided into six groups (eight mice per group) after 12 h of fasting. The test compounds were dissolved in 0.5% MC and vortexed before the study initiation. Mice in each group were gavaged with vehicle (0.5% MC aqueous solution, 10 mL/kg), TUG-891 (30 mg/kg, 10 mL/kg), or compound 14d (3, 10, 30, 100 mg/kg; 10 mL/kg), 30 min before oral glucose loading (3 g/kg, 10 mL/kg). The exact dose volume was calculated separately for each animal. Blood samples were collected via the tail tip 30 min before the compound dose, at t = 0 (immediately before glucose loading), and 15, 30, 60, and 120 min after glucose loading. Blood glucose levels were measured using blood glucose test strips (Sannuo GA-3 type; Changsha, China). Glucose values were entered into an Excel sheet and plotted using GraphPad Prism software.

Anti-Hyperglycemic Effects in DIO Mice

After 1 week of adaptation, male C57BL/6 mice were fed a high-fat diet (45% calories from fat, from Mediscience Ltd., Yangzhou, China) ad libitum for an additional 12 weeks to induce insulin resistance and were used as DIO mice. DIO mice were fasted overnight (12 h), weighed, and randomly divided into three groups (six mice per group). Thereafter, the DIO mice were orally administered a single dose of vehicle (0.5% MC, 10 mL/kg), TUG-891(20 mg/kg, 10 mL/kg), or 14d (20 mg/kg, 10 mL/kg), 30 min before oral glucose loading (2 g/kg). Blood samples were collected with retro-orbital sinus puncture, and glucose levels were measured in accordance with the OGTT in normal mice. Plasma insulin levels were measured using a mouse insulin RIA kit (Beijing North Institute of Biological Technology, Beijing, China).

3.3. Molecular Modeling Study

Homologous modeling was performed to model the structure of the GPR120 receptor using Accelrys Discovery Studio 2020 (DS2020), as the structure of GPR120 has not yet been obtained experimentally. The sequence of human GPR120 was obtained from the UniProtKB database (identifier: Q5NUL3), and NCBI BLAST was used to screen the template proteins. Neurotensin receptor21 (PDB code 4XES), β2-adrenoceptor (PDB code 3P0G), and turkey β1 adrenoceptor20 (PDB code 6IBL) were selected as templates because of their high homology with the GPR120 receptor. The DS2020 modeler was used according to the manufacturer’s instructions to construct a homology model of the GPR120 receptor after sequence alignment. The model with the lowest DOPE score was evaluated for energy minimization and reliability. The reliable model was used for molecular docking analysis. The CDOCKER molecular docking module in DS2020 was used for molecular docking research of compound 14d. The docking results were analyzed using the Discovery Studio software.

4. Conclusions

In summary, a series of aryloxyalkyl acid derivatives were designed, synthesized and evaluated for their biological activity. The chemical structures of these compounds were determined with 1H-NMR, 13C-NMR spectroscopy, and HRMS. Among these compounds, compound 14d was to found have excellent GPR120 agonistic activity and selectivity, and demonstrated excellent pharmacokinetic properties, with high oral exposure and acceptable half-life. The results of the in vivo hypoglycemic evaluation showed that compound 14d reduced the blood glucose levels in normal mice in a dose-dependent manner, and no hypoglycemic side effects were detected even at a dose of 100 mg/kg. Additionally, compound 14d significantly increased insulin secretion and played an anti-diabetic role in DIO mice. Molecular simulations showed that compound 14d bound well to the active site of GPR120 and formed hydrogen bonding interactions with ARG99. Collectively, these results revealed that compound 14d may be a promising lead compound deserving of further biological evaluation and structural modifications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27249018/s1, Figure S1: The pharmacokinetic profile traces of compound 14d and TUG-891; Figure S2: The dose-dependent curves of compound 14d and TUG-891 for hGPR120 and mGPR120; Tabel S1: EC50, Hill Slope, and Hill Slope (95%) of TUG-891 and 14d; Figures S3–S80: Copies of NMR and HRMS spectra of compounds 10a10l and 14a14n.

Author Contributions

X.W., G.J. and S.W. (Shiben Wang) conceived and wrote the paper; X.L., S.W. (Shiting Wei), M.W., Y.X., W.H., Z.G., R.L. and X.W. performed the experiments and analyzed the data. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (81803360 and 21675071), Major Project of Research and Development of Shandong Province (2019GSF108226), Guangyue Young Scholar Innovation Team of Liaocheng University (LCUGYTD2022-04) and the Doctoral Foundation of Liaocheng University (No. 318051745).

Institutional Review Board Statement

The protocol of the experiment was previously approved by the Scientific Research Ethic Welfare Committee of Liaocheng University (Approval No. SREWC-LCU2020012802).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of compounds 10a10l and 14a14n are available from the authors.

References

  1. Lapertosa, S.G.D.; Moura, A.F.D.; Decroux, C.; Duke, L.; Hammond, L.; Jacobs, E.; Kaundal, A.; Li, J.; Liu, J.; Ohlrogge, A.W.; et al. IDF Diabetes Atlas, 10th ed.; International Diabetes Federation: Brussells, Belgium, 2021; Available online: http://www.diabetesatlas.org (accessed on 7 September 2022).
  2. Joshi, P.; Joshi, S. Oral hypoglycaemic drugs and newer agents use in Type 2 diabetes mellitus. S. Afr. Fam. Pract. 2009, 51, 10–16. [Google Scholar] [CrossRef] [Green Version]
  3. Grundmann, M.; Bender, E.; Schamberger, J.; Eitner, F. Pharmacology of Free Fatty Acid Receptors and Their Allosteric Modulators. Int. J. Mol. Sci. 2021, 22, 1763. [Google Scholar] [CrossRef] [PubMed]
  4. Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.-Y.; Lannoy, V.; Decobecq, M.-E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional Characterization of Human Receptors for Short Chain Fatty Acids and Their Role in Polymorphonuclear Cell Activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Nilsson, N.E.; Kotarsky, K.; Owman, C.; Olde, B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem. Bioph. Res. Commun. 2003, 303, 1047–1052. [Google Scholar] [CrossRef]
  7. Briscoe, C.P.; Tadayyon, M.; Andrews, J.L.; Benson, W.G.; Chambers, J.K.; Eilert, M.M.; Ellis, C.; Elshourbagy, N.A.; Goetz, A.S.; Minnick, D.T. The orphan G protein-coupled receptor GPR40 is activated by medium and long chain fatty acids. J. Biol. Chem. 2003, 278, 11303–11311. [Google Scholar] [CrossRef] [Green Version]
  8. Burant, C.F. Activation of GPR40 as a Therapeutic Target for the Treatment of Type 2 Diabetes. Diabetes Care 2013, 36 (Suppl. S2), S175–S179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Hirasawa, A.; Tsumaya, K.; Awaji, T.; Katsuma, S.; Adachi, T.; Yamada, M.; Sugimoto, Y.; Miyazaki, S.; Tsujimoto, G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat. Med. 2005, 11, 90–94. [Google Scholar] [CrossRef]
  10. Milligan, G.; Alvarez-Curto, E.; Hudson, B.D.; Prihandoko, R.; Tobin, A.B. FFA4/GPR120: Pharmacology and Therapeutic Opportunities. Trends Pharmacol. Sci. 2017, 38, 809–821. [Google Scholar] [CrossRef] [Green Version]
  11. Miyauchi, S.; Hirasawa, A.; Iga, T.; Liu, N.; Itsubo, C.; Sadakane, K.; Hara, T.; Tsujimoto, G. Distribution and regulation of protein expression of the free fatty acid receptor GPR120. N-S. Arch. Pharmacol. 2009, 379, 427–434. [Google Scholar] [CrossRef] [PubMed]
  12. Talukdar, S.; Bae, E.J.; Imamura, T.; Morinaga, H.; Fan, W.; Li, P.; Lu, W.J.; Watkins, S.M.; Olefsky, J.M. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 2010, 142, 687–698. [Google Scholar]
  13. Meier, J.J.; Nauck, M.A. Glucagon-like peptide 1(GLP-1) in biology and pathology. Diabetes-Metab. Res. 2005, 21, 91–117. [Google Scholar] [CrossRef] [PubMed]
  14. Marathe, C.S.; Rayner, C.K.; Jones, K.L.; Horowitz, M. Glucagon-like peptides 1 and 2 in health and disease: A review. Peptides 2013, 44, 75–86. [Google Scholar] [CrossRef] [PubMed]
  15. Shimpukade, B.; Hudson, B.D.; Hovgaard, C.K.; Milligan, G.; Ulven, T. Discovery of a potent and selective GPR120 agonist. J. Med. Chem. 2012, 55, 4511–4515. [Google Scholar] [CrossRef] [PubMed]
  16. Sparks, S.M.; Chen, G.; Collins, J.L.; Danger, D.; Dock, S.T.; Jayawickreme, C.; Jenkinson, S.; Laudeman, C.; Leesnitzer, M.A.; Liang, X.; et al. Identification of diarylsulfonamides as agonists of the free fatty acid receptor 4 (FFA4/GPR120). Bioorgan. Med. Chem. Lett. 2014, 24, 3100–3103. [Google Scholar] [CrossRef] [PubMed]
  17. Azevedo, C.M.; Watterson, K.R.; Wargent, E.T.; Hansen, S.V.; Hudson, B.D.; Kepczynska, M.A.; Dunlop, J.; Shimpukade, B.; Christiansen, E.; Milligan, G.; et al. Non-Acidic Free Fatty Acid Receptor 4 Agonists with Antidiabetic Activity. J. Med. Chem. 2016, 59, 8868–8878. [Google Scholar] [CrossRef]
  18. Lombardo, M.; Bender, K.; London, C.; Kirkland, M.; Mane, J.; Pachanski, M.; Geissler, W.; Cummings, J.; Habulihaz, B.; Akiyama, T.E.; et al. Discovery of benzofuran propanoic acid GPR120 agonists: From uHTS hit to mechanism-based pharmacodynamic effects. Bioorgan. Med. Chem. Lett. 2016, 26, 5724–5728. [Google Scholar] [CrossRef] [PubMed]
  19. Adams, G.L.; Velazquez, F.; Jayne, C.; Shah, U.; Miao, S.; Ashley, E.R.; Madeira, M.; Akiyama, T.E.; Di Salvo, J.; Suzuki, T.; et al. Discovery of Chromane Propionic Acid Analogues as Selective Agonists of GPR120 with in Vivo Activity in Rodents. ACS Med. Chem. Lett. 2017, 8, 96–101. [Google Scholar] [CrossRef] [Green Version]
  20. Cox, J.M.; Chu, H.D.; Chelliah, M.V.; Debenham, J.S.; Eagen, K.; Lan, P.; Lombardo, M.; London, C.; Plotkin, M.A.; Shah, U.; et al. Design, Synthesis, and Evaluation of Novel and Selective G-protein Coupled Receptor 120 (GPR120) Spirocyclic Agonists. ACS Med. Chem. Lett. 2017, 8, 49–54. [Google Scholar] [CrossRef] [Green Version]
  21. Li, Z.; Xu, X.; Li, G.; Fu, X.; Liu, Y.; Feng, Y.; Wang, M.; Ouyang, Y.; Han, J. Improving metabolic stability with deuterium: The discovery of GPU-028, a potent free fatty acid receptor 4 agonists. Bioorgan. Med. Chem. 2017, 25, 6647–6652. [Google Scholar] [CrossRef]
  22. McCoull, W.; Bailey, A.; Barton, P.; Birch, A.M.; Brown, A.J.; Butler, H.S.; Boyd, S.; Butlin, R.J.; Chappell, B.; Clarkson, P.; et al. Indazole-6-phenylcyclopropylcarboxylic Acids as Selective GPR120 Agonists with In Vivo Efficacy. J. Med. Chem. 2017, 60, 3187–3197. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, X.; Cai, C.; Sui, Z.; Macielag, M.; Wang, Y.; Yan, W.; Suckow, A.; Hua, H.; Bell, A.; Haug, P.; et al. Discovery of an Isothiazole-Based Phenylpropanoic Acid GPR120 Agonist as a Development Candidate for Type 2 Diabetes. ACS Med. Chem. Lett. 2017, 8, 947–952. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, X.; Cai, C.; Winters, M.; Wells, M.; Wall, M.; Lanter, J.; Sui, Z.; Ma, J.; Novack, A.; Nashashibi, I.; et al. Design, synthesis and SAR of a novel series of heterocyclic phenylpropanoic acids as GPR120 agonists. Bioorgan. Med. Chem. Lett. 2017, 27, 3272–3278. [Google Scholar] [CrossRef] [PubMed]
  25. Ji, G.; Guo, Q.; Xue, Q.; Kong, R.; Wang, S.; Lei, K.; Liu, R.; Wang, X. Novel GPR120 Agonists with Improved Pharmacokinetic Profiles for the Treatment of Type 2 Diabetes. Molecules 2021, 26, 6907. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, X.; Ji, G.; Han, X.; Hao, H.; Liu, W.; Xue, Q.; Guo, Q.; Wang, S.; Lei, K.; Liu, Y. Thiazolidinedione derivatives as novel GPR120 agonists for the treatment of type 2 diabetes. RSC Adv. 2022, 12, 5732–5742. [Google Scholar] [CrossRef] [PubMed]
  27. Schilperoort, M.; van Dam, A.D.; Hoeke, G.; Shabalina, I.G.; Okolo, A.; Hanyaloglu, A.C.; Dib, L.H.; Mol, I.M.; Caengprasath, N.; Chan, Y.-W.; et al. The GPR120 agonist TUG-891 promotes metabolic health by stimulating mitochondrial respiration in brown fat. Embo Mol. Med. 2018, 10, e8047. [Google Scholar] [CrossRef] [PubMed]
  28. Oh, D.Y.; Walenta, E.; Akiyama, T.E.; Lagakos, W.S.; Lackey, D.; Pessentheiner, A.R.; Sasik, R.; Hah, N.; Chi, T.J.; Cox, J.M.; et al. A Gpr120-selective agonist improves insulin resistance and chronic inflammation in obese mice. Nat. Med. 2014, 20, 942–947. [Google Scholar] [CrossRef] [Green Version]
  29. Lee, Y.; Warne, T.; Nehmé, R.; Pandey, S.; Dwivedi-Agnihotri, H.; Chaturvedi, M.; Edwards, P.C.; García-Nafría, J.; Leslie, A.G.; Shukla, A.K. Molecular basis of β-arrestin coupling to formoterol-bound β 1-adrenoceptor. Nature 2020, 583, 862–866. [Google Scholar] [CrossRef]
  30. Krumm, B.E.; White, J.F.; Shah, P.; Grisshammer, R. Structural prerequisites for G-protein activation by the neurotensin receptor. Nat. Commun. 2015, 6, 7895. [Google Scholar] [CrossRef] [Green Version]
  31. Rasmussen, S.G.; Choi, H.-J.; Fung, J.J.; Pardon, E.; Casarosa, P.; Chae, P.S.; DeVree, B.T.; Rosenbaum, D.M.; Thian, F.S.; Kobilka, T.S. Structure of a nanobody-stabilized active state of the β 2 adrenoceptor. Nature 2011, 469, 175–180. [Google Scholar] [CrossRef] [Green Version]
  32. Suzuki, T.; Igari, S.-I.; Hirasawa, A.; Hata, M.; Ishiguro, M.; Fujieda, H.; Itoh, Y.; Hirano, T.; Nakagawa, H.; Ogura, M.; et al. Identification of G protein-coupled receptor 120-selective agonists derived from PPARγ agonists. J. Med. Chem 2008, 51, 7640–7644. [Google Scholar] [CrossRef] [PubMed]
  33. Sun, Q.; Hirasawa, A.; Hara, T.; Kimura, I.; Adachi, T.; Awaji, T.; Ishiguro, M.; Suzuki, T.; Miyata, N.; Tsujimoto, G. Structure-activity relationships of GPR120 agonists based on a docking simulation. Mol. Pharmacol. 2010, 78, 804–810. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representative GPR120 agonists.
Figure 1. Representative GPR120 agonists.
Molecules 27 09018 g001
Figure 2. The design of phenoxyalkanoic acid derivatives.
Figure 2. The design of phenoxyalkanoic acid derivatives.
Molecules 27 09018 g002
Scheme 1. Reagents and conditions: Synthesis of target compounds 10a10l. (a) K2CO3, DMF, rt., 12 h, 62–78%; (b) NaBH4, CH3OH, THF, 0 °C, 2 h; (c) PBr3, CH2Cl2, 0 °C, 2 h, 48–56% (two steps); (d) K2CO3, acetone, 50 °C, 12 h, 82%; (e) 4-Tolylboronic acid, Pd(PPh3)4, Na2CO3, toluene, ethanol, H2O, 80 °C, overnight, 75%; (f) H2, Pd-C, rt., 12 h, 77%; (g) K2CO3, DMF, rt., 12 h; (h) NaOH, H2O, CH3OH, THF, 2 h, 51–58% (two steps).
Scheme 1. Reagents and conditions: Synthesis of target compounds 10a10l. (a) K2CO3, DMF, rt., 12 h, 62–78%; (b) NaBH4, CH3OH, THF, 0 °C, 2 h; (c) PBr3, CH2Cl2, 0 °C, 2 h, 48–56% (two steps); (d) K2CO3, acetone, 50 °C, 12 h, 82%; (e) 4-Tolylboronic acid, Pd(PPh3)4, Na2CO3, toluene, ethanol, H2O, 80 °C, overnight, 75%; (f) H2, Pd-C, rt., 12 h, 77%; (g) K2CO3, DMF, rt., 12 h; (h) NaOH, H2O, CH3OH, THF, 2 h, 51–58% (two steps).
Molecules 27 09018 sch001
Scheme 2. Reagents and conditions: Synthesis of target compounds 14a14n. (a) Pd(PPh3)4, Na2CO3, toluene, ethanol, H2O, 80 °C, overnight, 70–76%; (b) H2, Pd-C, rt., 12 h, 66–78%; (c) K2CO3, DMF, rt., 12 h, 73–85%; (d) NaOH, H2O, CH3OH, THF, 2 h, 53–62% (two steps).
Scheme 2. Reagents and conditions: Synthesis of target compounds 14a14n. (a) Pd(PPh3)4, Na2CO3, toluene, ethanol, H2O, 80 °C, overnight, 70–76%; (b) H2, Pd-C, rt., 12 h, 66–78%; (c) K2CO3, DMF, rt., 12 h, 73–85%; (d) NaOH, H2O, CH3OH, THF, 2 h, 53–62% (two steps).
Molecules 27 09018 sch002
Figure 3. Effects of compound 14d and TUG-891 on plasma glucose levels during the OGTT in fasting C57BL/6 mice. (a) Time-dependent changes of blood glucose levels after oral administration of compound 14d, followed by the oral glucose load (3 g/kg). (b) AUC−30–120 min of plasma glucose levels. Values are expressed as the mean ± SD (n = 8). * p ≤ 0.05 compared with vehicle-treated C57BL/6 mice by Student’s t test; ** p ≤ 0.01 compared with vehicle-treated C57BL/6 mice by Student’s t test; *** p ≤ 0.001 compared with vehicle-treated C57BL/6 mice by Student’s t test; # p ≤ 0.05 compared with TUG-891-treated C57BL/6 mice by Student’s t test; ### p ≤ 0.001 compared with TUG-891-treated C57BL/6 mice by Student’s t test.
Figure 3. Effects of compound 14d and TUG-891 on plasma glucose levels during the OGTT in fasting C57BL/6 mice. (a) Time-dependent changes of blood glucose levels after oral administration of compound 14d, followed by the oral glucose load (3 g/kg). (b) AUC−30–120 min of plasma glucose levels. Values are expressed as the mean ± SD (n = 8). * p ≤ 0.05 compared with vehicle-treated C57BL/6 mice by Student’s t test; ** p ≤ 0.01 compared with vehicle-treated C57BL/6 mice by Student’s t test; *** p ≤ 0.001 compared with vehicle-treated C57BL/6 mice by Student’s t test; # p ≤ 0.05 compared with TUG-891-treated C57BL/6 mice by Student’s t test; ### p ≤ 0.001 compared with TUG-891-treated C57BL/6 mice by Student’s t test.
Molecules 27 09018 g003
Figure 4. Effects of compound 14d and TUG-891 on plasma glucose levels during the OGTT in fasting DIO mice. (a) The changes of blood glucose levels after oral administration of compound 14d, followed by the oral glucose load (2 g/kg); (b) AUC−30–120 min of plasma glucose levels. (c) Insulin levels at 30 min after the glucose load. Values are expressed as the mean ± SD (n = 6). * p ≤ 0.05 compared with vehicle-treated DIO mice by Student’s t test; ** p ≤ 0.01 compared with vehicle-treated DIO mice by Student’s t test; *** p ≤ 0.001 compared with vehicle-treated DIO mice by Student’s t test; # p ≤ 0.05 compared with TUG-891-treated DIO mice by Student’s t test; ns means no significant.
Figure 4. Effects of compound 14d and TUG-891 on plasma glucose levels during the OGTT in fasting DIO mice. (a) The changes of blood glucose levels after oral administration of compound 14d, followed by the oral glucose load (2 g/kg); (b) AUC−30–120 min of plasma glucose levels. (c) Insulin levels at 30 min after the glucose load. Values are expressed as the mean ± SD (n = 6). * p ≤ 0.05 compared with vehicle-treated DIO mice by Student’s t test; ** p ≤ 0.01 compared with vehicle-treated DIO mice by Student’s t test; *** p ≤ 0.001 compared with vehicle-treated DIO mice by Student’s t test; # p ≤ 0.05 compared with TUG-891-treated DIO mice by Student’s t test; ns means no significant.
Molecules 27 09018 g004
Figure 5. (a) Docking of compound 14d inside a homology GPR120 model. (b) Docking of TUG-891 inside a homology GPR120 model.
Figure 5. (a) Docking of compound 14d inside a homology GPR120 model. (b) Docking of TUG-891 inside a homology GPR120 model.
Molecules 27 09018 g005
Table 1. In vitro activities and selectivity of target compounds 10a10l.
Table 1. In vitro activities and selectivity of target compounds 10a10l.
Molecules 27 09018 i001
CompoundnR1hGPR120 a
(EC50, nM)
hGPR40 b
(EC50, µM)
Selectivity
TUG-891 68.172.31061.7
10a1H92.476.2824.7
10b2H218.3NTNT
10c3H128.6112.6875.6
10d4H103.2143.61391.5
10e1CH3138.5141.21019.5
10f2CH3267.8NTNT
10g3CH3380.4NTNT
10h4CH3205.6NTNT
10i1F77.287.21129.5
10j2F112.285.5762.0
10k3F57.674.31289.9
10l4F96.894.7978.3
NT: Not tested. a EC50 values for hGPR120 activity represent the mean of three determinations. b EC50 values for hGPR40 activity represent the mean of three determinations.
Table 2. In vitro activities and selectivity of target compounds 14a14n.
Table 2. In vitro activities and selectivity of target compounds 14a14n.
Molecules 27 09018 i002
CompoundR2hGPR120 a
(EC50, nM)
hGPR40 b
(EC50, µM)
Selectivity
TUG-891-68.172.31061.7
14a2-Me90.468.5757.7
14b3-Me82.593.21129.7
14c2-F37.564.81728.0
14d4-F168.584.6502.1
14e4-OMe260.2NTNT
14f2-Me, 4-OMe170.2102.8604.0
14g2-Cl, 4-OMe269.5NTNT
14h2-F, 4-OMe421.5NTNT
14i2,4-diMe158.695.1600.0
14j3,4-diMe150.382.3547.6
14k3,5-diMe125.672.4576.4
14l2,4-diCl136.590.8665.2
14m2-Cl, 4-Me187.378.6419.6
14n2-F, 4-Me312.7NTNT
NT: Not tested. a EC50 values for hGPR120 activity represent the mean of three determinations. b EC50 values for hGPR40 activity represent the mean of three determinations.
Table 3. In vitro activities and selectivity of compound 14d for mGPR120 and mGPR40.
Table 3. In vitro activities and selectivity of compound 14d for mGPR120 and mGPR40.
CompoundmGPR120 a
(EC50, nM)
mGPR40 b
(EC50, µM)
Selectivity
TUG-891156.15.937.8
14d83.212.7152.6
a EC50 values for mGPR120 activity represent the mean of three determinations. b EC50 values for mGPR40 activity represent the mean of three determinations.
Table 4. Pharmacokinetic profiles of compounds 10k and 14d in C57BL/6 mice a.
Table 4. Pharmacokinetic profiles of compounds 10k and 14d in C57BL/6 mice a.
CompoundDose
(mg/kg)
Cmax
(ng/mL)
Tmax
(min)
AUC
(ng.h/mL)
t1/2
(h)
TUG-89110.01892.3 ± 254.815.01410.7 ± 157.50.6 ± 0.2
10k10.02368.5 ± 335.4 *30.04251.6 ± 577.3 ***1.1 ± 0.3 *
14d10.02647.3 ± 287.5 **30.05895.8 ± 583.6 ***,#1.6 ± 0.4 **
a Values are expressed as the mean ± SD of four C57BL/6 mice. TUG-891 and compounds 10k and 14d were suspended in 0.5% MC aqueous solution. * p ≤ 0.05 compared with TUG-891-treated by Student’s t test; ** p ≤ 0.01 compared with TUG-891-treated by Student’s t test; *** p ≤ 0.001 compared with TUG-891-treated by Student’s t test; # p ≤ 0.05 compared with 10k-treated by Student’s t test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, X.; Li, X.; Wei, S.; Wang, M.; Xu, Y.; Hu, W.; Gao, Z.; Liu, R.; Wang, S.; Ji, G. Discovery of Novel and Selective G-Protein Coupled Receptor 120 (GPR120) Agonists for the Treatment of Type 2 Diabetes Mellitus. Molecules 2022, 27, 9018. https://doi.org/10.3390/molecules27249018

AMA Style

Wang X, Li X, Wei S, Wang M, Xu Y, Hu W, Gao Z, Liu R, Wang S, Ji G. Discovery of Novel and Selective G-Protein Coupled Receptor 120 (GPR120) Agonists for the Treatment of Type 2 Diabetes Mellitus. Molecules. 2022; 27(24):9018. https://doi.org/10.3390/molecules27249018

Chicago/Turabian Style

Wang, Xuekun, Xu Li, Shiting Wei, Min Wang, Yao Xu, Weidi Hu, Zhenzhen Gao, Renmin Liu, Shiben Wang, and Guoxia Ji. 2022. "Discovery of Novel and Selective G-Protein Coupled Receptor 120 (GPR120) Agonists for the Treatment of Type 2 Diabetes Mellitus" Molecules 27, no. 24: 9018. https://doi.org/10.3390/molecules27249018

Article Metrics

Back to TopTop