Next Article in Journal
Inositol Hexaphosphate Inhibits Proliferation and Induces Apoptosis of Colon Cancer Cells by Suppressing the AKT/mTOR Signaling Pathway
Previous Article in Journal
A Comparative Genomic and Transcriptomic Survey Provides Novel Insights into N-Acetylserotonin Methyltransferase (ASMT) in Fish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis and Biological Evaluation of N,N-Substituted Amine Derivatives as Cholesteryl Ester Transfer Protein Inhibitors

1
Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
2
Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53715, USA
*
Author to whom correspondence should be addressed.
Molecules 2017, 22(10), 1658; https://doi.org/10.3390/molecules22101658
Submission received: 20 August 2017 / Revised: 29 September 2017 / Accepted: 30 September 2017 / Published: 3 October 2017
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
N,N-Substituted amine derivatives were designed by utilizing a bioisosterism strategy. Consequently, twenty-two compounds were synthesized and evaluated for their inhibitory activity against CETP. Structure-activity relationship (SAR) studies indicate that hydrophilic groups at the 2-position of the tetrazole and 3,5-bistrifluoromethyl groups on the benzene ring provide important contributions to the potency. Among these compounds, compound 17 exhibited excellent CETP inhibitory activity (IC50 = 0.38 ± 0.08 μM) in vitro. Furthermore, compound 17 was selected for an in vitro metabolic stability study.

Graphical Abstract

1. Introduction

Cholesteryl ester transfer protein (CETP) is a hydrophobic glycoprotein secreted predominately by the liver. CETP facilitates the movement of cholesteryl esters (CEs) from high-density lipoprotein (HDL) to low-density lipoprotein (LDL) and very low-density lipoprotein (VLDL), in exchange for triglycerides (TGs) [1,2]. Epidemiological studies have provided compelling evidence to demonstrate an inverse association between HDL-C and cardiovascular events [3,4,5]. Therefore, CETP inhibitors can provide benefits to high risk coronary heart disease (CHD) patients by increasing HDL-C plasma levels [1,6,7,8].
Torcetrapib (1, Figure 1) was the first CETP inhibitor to undergo advanced clinical development. Although early studies have demonstrated that torcetrapib exhibited a significant lipid-regulating ability, subsequent trials indicated that this inhibitor possessed off-target effects affecting blood pressure and aldosterone levels, and failed to demonstrate any favourable impact on CHD patients [9,10,11,12,13]. Accordingly, torcetrapib’s phase III trial was halted in 2006. Dalcetrapib (2, Figure 1) was a modest inhibitor without the off-targets effect seen with torcetrapib, elevating HDL-C levels by up to 30% and with a minimal effect on LDL-C levels [14,15]. However, dal-OUTCOMES study was stopped due to lack of clinical benefit [16]. More recently, a Canadian study found that dalcetrapib exhibited potentially clinical effects on atherosclerotic patients with polymorphisms in the ADCY9 gene using a genome-wide approach [17]. Evacetrapib (3, Figure 1) possessed profound lipid-regulating effects, raising HDL-C levels by more than 129% and decreasing LDL-C by up to 36%. Evacetrapib did not exhibit torcetrapib-like side-effects, but its phase III ACCELERATE study was terminated due to clinical futility [18,19]. Anacetrapib (4, Figure 1) a potent CETP inhibitor currently in phase III, could raise HDL-C levels to 130% and decrease LDL-C levels to 40%. Researchers found that anacetrapib demonstrated potential clinical benefits and would not produce adverse clinical effects similar to those observed with torcetrapib in the DEFINE study [20,21]. TA-8995 (5, Figure 1), a novel CETP inhibitor, was well tolerated and had beneficial effects on lipids, raising HDL-C levels by up to 179% and decreasing LDL-C by up to 45% [22,23]. It remains to be seen whether these potent CETP inhibitors will proceed further in the future.
In a previous study, compound 6 (Figure 2) was identified to show weak micromolar activity (IC50 = 20.97 μM) [24]. Our initial approach was to build a ring to replace the carbamate part to investigate the effect of steric hindrance on activity. Considering that tetrazole is a bioisoster of the carboxylic acid and the carbamate part of compound 6 could be treated as a carboxylic acid methyl ester, we hold that 2-methyltetrazole is a bioisoster of carbamate. In this paper, our group investigated the cyclization of the carbamate using a bioisosterism strategy to afford a 2-methyltetrazole compound. Fortunately, the compound containing the tetrazole moiety (40, Figure 2, IC50 = 0.81 ± 0.03 μM) showed increased inhibitory activity. A series of novel N,N-substituted amine derivatives were then synthesized. Optimization efforts on this scaffold are discussed in this study.

2. Results and Discussion

2.1. Chemistry

Compounds 1222 were prepared according to the procedure shown in Scheme 1. The intermediate 10 was obtained in a manner similar to that described in the previously published paper [24]. The treatment of 10 with cyanogen bromide under basic conditions furnished 11 in good yield. Then, the -CN of the resulting intermediate was subjected to cycloaddition with sodium azide to produce 12. Compound 12 was subjected to substitution reactions with various substituted alkyl bromides to yield compounds 1315. Alternatively, treatment of 12 with 2-(methylsulfonyl) ethanol under Mitsunobu conditions afforded 16. Carboxylic acid derivatives 17 and 18 were prepared by hydrolysis of the ester groups of compounds 13 and 14, respectively. Subsequent reduction or aminolysis reaction of compounds 13 and 14 produced compounds 1921. Compound 11 reacted with hydroxylamine hydrochloride and subsequently, acetic anhydride, to obtained compound 22. Compounds 3237 were prepared according to the procedure in Scheme 2. The resulting 9 was reacted with NaBH4 and subsequently, SOCl2 to afford intermediate 23.
The starting materials 24 and 25 were subjected to reductive amination with 3,5-bis(trifluromethyl)benzaldehyde, which provided 26 and 27. Intermediates 28 and 29 were obtained by the nucleophilic substitution of 23 by 26 and 27 in the presence of NaH at room temperature. The key intermediates 30 and 31 were obtained by deprotection of 28 and 29 under the condition of TFA/DCM (v:v = 1:1). Intermediate 30 was reacted with methyl 2-bromoacetate, then the resultant ester intermediate was subjected to reduction conditions to yield compound 32 or hydrolysis conditions to furnish compound 33. The obtained intermediates 30 and 31 were treated with corresponding chloroformates to give compounds 3437. As illustrated in Scheme 3, compounds 4044 were synthesized from the corresponding starting benzaldehydes through reductive amination and substitution reactions.

2.2. In Vitro Activity Against Cholesteryl Ester Transfer Protein

The N,N-substituted amine derivatives and the reference compound anacetrapib (4) were screened for their in vitro activity against CETP by a BODIPY-CE fluorescence assay with the CETP RP Activity Assay Kit (Catalogue # RB-RPAK; Roar, New York, NY, USA). The results are shown in Table 1. As seen in the table, replacement of a carbamate (6, IC50 = 20.97 µM) with an N-2-methyltetrazole (40, IC50 = 0.81 ± 0.03 µM) caused a significant increase in the activity. Subsequently, we investigated the relationship between various substituents at the 2-position of the tetrazole (Part A) and the CETP inhibitory activity. The replacement of –CH3 by –H (12, IC50 = 2.84 ± 0.04 µM) was detrimental for activity. Introduction of carboxylic esters (e.g., compounds 13 and 14) at the 2-position of the tetrazole dramatically reduced the activity, however potency was recovered in the corresponding carboxylic acids (17 and 18) and straight chain saturated alcohols (19 and 20), where especially two atoms alkyl chains were better. We observed that amine and sulfone groups (15 and 16) were tolerated and an amide group (21) caused a weak decrease in the activity. Changing N-2-methyltetrazole (40) to 3-methyoxadiazole (22, IC50 = 0.72 ± 0.06 µM) showed no advantage. We speculate that carboxylic acids and saturated alcohols at the 2-position of tetrazole provide an important contribution to the potency.
Next, we investigated the relationship between an aliphatic heterocycle (Part A) and the CETP inhibitory activity. Unfortunately, replacement of N-2-substituted tetrazole with azetidine/ piperidine derivatives (compounds 3237) caused a dramatic decrease in the activity. These results indicate that a flexible fragment in Part A is unfavourable for inhibitory activity.
To further study the relationship between Part B and the CETP inhibitory activity, another five compounds 4044 were prepared and evaluated for their activity. Introduction of trifluoromethyl groups on the 3-position and the 5-position of benzene ring (40, IC50 = 0.81 ± 0.03 µM) were more beneficial for CETP inhibition. A single trifluoromethyl group on the meta-position of the benzene ring (41, IC50 = 10.05 ± 0.06 µM) caused a severe decrease in the activity. A trifluoromethyl group (43), fluorine atom (42), and trifluoromethoxy group (44) on the para-position of the benzene ring resulted in no CETP inhibition.
Half of the compounds showed excellent activities, and four of them exhibited submicromolar activities, but the activity of the most outstanding compound 17 (IC50 = 0.38 ± 0.08 μM) was still 10-fold weaker than the reference compound.

2.3. In Vitro Metabolic Stability Study

Based on the result of the in vitro CETP inhibitory assay, the potent inhibitor 17 was selected for the in vitro metabolic stability study. As shown in Table 2, compound 17 showed weak stability, with a clearance rate of 119.0 and 146.1 μL/min/mg in human and rat liver microsomes.
In addition, five cytochrome P450 (CYP) enzymes that commonly metabolize exogenous chemicals were used to test the direct inhibition of compound 17. Compound 17 possessed favourable metabolic properties, as the inhibition ratios for five CYPs were less than 20% even at the compound concentration of 10 μM.

3. Experimental

3.1. General Information

All chemicals were obtained from commercial sources and were used without purification unless otherwise specified. Solvents were distilled and dried using standard methods. TLC was performed on silica gel plates with F-254 indicator and visualized by UV-light. The purities of target compounds (all ≥95%). were detected by HPLC, performed on a Waters 1525–2489 system (Waters, Milford, MA, USA). The method conditions were as follows: 100% CH3OH or a mixture of solvents H2O (A) and MeOH (B) (VA:VB = 5:95) as eluent, flow rate at 1.0 mL/min. Peaks were detected at λ = 254 nm. NMR spectra were recorded on 400 MHz and 600 MHz instruments (Bruker, Karlsruhe, Germany) and the chemical shifts were reported in terms of parts per million with TMS as the internal reference. High-resolution accurate mass determinations (HRMS) for all final target compounds were obtained on a Bruker Micromass Time of Flight mass spectrometer equipped with electrospray ionisation (ESI). Column chromatography was performed with silica gel (200–300 mesh) purchased from Qingdao Haiyang Chemical Co. Ltd. (Qingdao, China)
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)cyanamide (11): Intermediate 10 (0.3 g, 0.6 mmol) was dissolved in THF (5 mL) and cyanogen bromide (0.2 g, 1.8 mmol) and N,N-diisopropylethylamine (0.4 mL, 2.4 mmol) were added. The reaction mixture was stirred at room temperature for 5 h and then poured into H2O (10 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 15:1) to give 11 (0.3 g, 92.8%) as a pale yellow oil. 1H-NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H), 7.90 (s, 2H), 7.05 (dd, J = 8.5, 2.2 Hz, 1H), 6.91–6.77 (m, 2H), 4.29 (s, 2H), 3.64 (s, 3H), 3.49–3.35 (m, 2H), 2.72 (dt, J = 13.7, 6.9 Hz, 1H), 2.40–2.28 (m, 1H), 2.11–1.99 (m, 1H), 1.90 (m, 2H), 1.40 (t, J = 6.4 Hz, 2H), 1.08 (dd, J = 6.9, 1.6 Hz, 6H), 0.98 (d, J = 2.8 Hz, 6H). HPLC: tR = 13.710 min, 99.27%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine (12): Intermediate 11 (0.1 g, 0.2 mmol) was dissolved in DMF (5 mL) and ammonium chloride (0.01 g, 0.8 mmol) and sodium azide (0.05 g, 0.2 mmol) were added. After being stirred at 100 °C for 5 h, the reaction mixture was cooled to room temperature, and H2O (20 mL) was added. The mixture was extracted with CH2Cl2 (20 mL × 3) and the combined organic layers were washed with water (20 mL × 3) and brine (20 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 2:1) to give 12 (0.1 g, 96.3%) as a white solid. Mp 150.6–152.6 °C. 1H-NMR (600 MHz, DMSO-d6) δ 14.88 (s, 1H), 7.94 (s, 1H), 7.66 (s, 2H), 6.99 (dd, J = 8.5, 2.2 Hz, 1H), 6.77 (d, J = 8.5 Hz, 1H), 6.71 (d, J = 2.2 Hz, 1H), 4.55 (s, 2H), 3.91 (m, 2H), 3.61 (s, 3H), 2.61 (dt, J = 13.8, 6.9 Hz, 1H), 2.36–2.27 (m, 1H), 2.06–2.03 (m, 1H), 1.77–1.69 (m, 2H), 1.38 (s, 2H), 1.36 (t, J = 6.4 Hz, 2H), 1.00 (d, J = 6.9 Hz, 6H), 0.91 (d, J = 10.3 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 151.95, 142.58, 139.79, 136.20, 132.01(×2), 129.76, 128.36, 127.92(×2), 127.27, 126.44, 123.95, 122.14, 121.79, 111.11(×2), 55.84(×2), 53.23, 39.75, 34.93, 33.16, 30.00, 28.75, 28.44, 27.24, 24.07, 23.99. HRMS calcd for C29H34F6N5O, [M + H]+, 582.2589; found 582.2668. HPLC: tR = 14.640 min, 96.47%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-carboxylic acid methyl ester (13): Compound 12 (0.1 g, 0.2 mmol) and triethylamine (0.1 mL, 0.8 mmol) were dissolved in acetonitrile (2 mL) followed by the addition of methyl 2-bromoacetate (0.03 mL, 0.4 mmol). After being stirred at 80 °C for 2 h, the reaction mixture was cooled to room temperature, and H2O (10 mL) was added. The aqueous layer was extracted with EtOAc (5 mL × 3) and the combined organic layers were washed with H2O (5 mL × 3) and brine (5 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 13 (0.09 g, 68.4%) as a colourless oil. 1H-NMR (400 MHz, CDCl3) δ 7.67 (s, 1H), 7.54 (s, 2H), 7.03 (dd, J = 8.4, 2.3 Hz, 1H), 6.76 (d, J = 2.3 Hz, 1H), 6.72 (d, J = 8.5 Hz, 1H), 5.18 (s, 2H), 4.58–4.38 (m, 2H), 4.19 (d, J = 14.5 Hz, 1H), 4.00 (d, J = 14.4 Hz, 1H), 3.76 (s, 3H), 3.68 (s, 3H), 2.76 (dt, J = 13.8, 6.9 Hz, 1H), 2.54–2.37 (m, 1H), 2.16–2.00 (m, 1H), 1.83 (s, 2H), 1.50–1.33 (m, 2H), 1.15 (d, J = 6.9 Hz, 6H), 0.94 (d, J = 11.9 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.64, 165.70, 154.12, 141.11, 140.87, 135.34, 131.28(×2), 130.52, 128.06(×2), 127.76(×2), 127.63, 125.66(×2), 110.64(×2), 55.24, 53.01, 52.91, 51.69, 49.38, 40.57, 35.42, 33.03, 29.10, 28.99, 28.03(×2), 23.99(×2). HRMS calcd for C32H38F6N5O3, [M + H]+, 654.2801; found 654.2877. HPLC: tR = 8.753 min, 95.8%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-butyric acid ethyl ester (14): Colourless oil; yield 71.3%; 1H-NMR (400 MHz, CDCl3) δ 7.66 (s, 1H), 7.54 (s, 2H), 7.03 (d, J = 7.6 Hz, 1H), 6.82–6.65 (m, 2H), 4.46 (d, J = 3.5 Hz, 4H), 4.18–4.11(m, 3H), 3.98 (d, J = 14.5 Hz, 1H), 3.67 (s, 3H), 2.76 (s, 1H), 2.46 (d, J = 19.3 Hz, 1H), 2.30 (d, J = 4.5 Hz, 2H), 2.23 (s, 2H), 2.12–2.03 (m, 1H), 1.82 (s, 2H), 1.42 (s, 2H), 1.25–1.24 (m, 3H), 1.14 (s, 6H), 0.94 (d, J = 11.1 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 172.11, 169.37, 154.14, 141.34, 140.85, 135.18, 131.24(×2), 130.57, 128.08, 127.77(×2), 125.63, 124.16, 122.35, 120.60, 110.62(×2), 60.54, 55.22, 51.77, 51.65, 49.36, 40.61, 35.43, 33.03, 30.60, 29.10, 29.00, 28.04, 28.02, 24.12, 23.99, 23.98, 14.04. HRMS calcd for C35H44F6N5O3, [M + H]+, 696.3270; found 656.3361. HPLC: tR = 7.653 min, 96.4%.
2-(2-Aminoethyl)-N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethyl-cyclohex-1-enyl)methyl)-2H-tetrazol-5-amine (15): Compound 12 (0.5 g, 0.9 mmol) and triethylamine (1.8 mL, 13.0 mmol) were dissolved in acetonitrile (10 mL), followed by the addition of tert-butyl 2-bromoethylcarbamate (0.6 mL, 2.6 mmol). After being stirred at 80 °C for 2 h, the reaction mixture was cooled to room temperature, and H2O (10 mL) was added. The aqueous layer was extracted with EtOAc (5 mL × 3) and the combined organic layers were washed with H2O (5 mL × 3) and brine (5 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was immediately dissolved in a trifluoroacetic acid–dichloromethane (1:1) solution (2 mL) and stirred at room temperature overnight. After concentration, the residue was dissolved in EtOAc (5 mL), washed with H2O (5 mL × 3) and brine (5 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 2:1) to give 15 (0.35 g, 62.2%) as a colourless oil. 1H-NMR (400 MHz, DMSO-d6) δ 7.93 (s, 1H), 7.66 (s, 2H), 7.00 (dd, J = 8.4, 2.2 Hz, 1H), 6.79–6.76 (m, 2H), 4.49 (s, 2H), 4.35 (t, J = 6.1 Hz, 2H), 4.02–3.89 (m, 2H), 3.62 (s, 3H), 2.94 (t, J = 6.2 Hz, 2H), 2.67 (dt, J = 13.8, 6.9 Hz, 1H), 2.54–2.37 (m, 1H), 2.16–2.00 (m, 1H), 1.76 (s, 2H), 1.35 (t, J = 6.3 Hz, 2H), 1.04 (d, J = 6.9 Hz, 6H), 0.89 (d, J = 10.2 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.41, 154.13, 141.31, 140.86, 135.29, 131.25(×2), 130.55, 128.07, 127.80, 127.70, 125.65, 124.14, 122.34, 120.63, 110.62(×2), 55.81, 55.22, 51.72, 49.39, 40.93, 40.64, 35.42, 33.03, 29.11, 29.01, 28.06, 28.03, 24.00, 23.98. HRMS calcd for C31H39F6N6O, [M + H]+, 625.3011; found 625.3070. HPLC: tR = 18.895 min, 96.6%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2-(2-(methylsulfonyl)ethyl)-2H-tetrazol-5-amine (16): 2-(methylsulfonyl)ethanol (0.04 mL, 0.4 mmol) was added to a solution of compound 12 (0.1 g, 0.2 mmol) in THF (2 mL) and cooled to 0 °C, followed by the addition of Ph3P (0.07 g, 0.3 mmol) and DIAD (0.05 mL, 0.3 mmol). After being stirred at room temperature for 4 h, the reaction mixture was poured into H2O (10 mL) and extracted with EtOAc (5 mL × 3), and the combined organic layers were washed with H2O (5 mL × 3) and brine (5 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 1:1) to give 16 (0.09 g, 65.3%) as a colourless oil. 1H-NMR (400 MHz, CDCl3) δ 7.68 (s, 1H), 7.50 (s, 2H), 7.03 (dd, J = 8.4, 2.2 Hz, 1H), 6.76 (d, J = 2.3 Hz, 1H), 6.71 (d, J = 8.5 Hz, 1H), 4.90 (t, J = 6.9 Hz, 2H), 4.48 (s, 2H), 4.17 (d, J = 15.4 Hz, 1H), 3.99 (d, J = 14.4 Hz, 1H), 3.70–3.60 (m, 5H), 2.82–2.71 (m, 4H), 2.46 (d, J = 18.1 Hz, 1H), 2.10 (d, J = 18.4 Hz, 1H), 1.79 (s, 2H), 1.43 (dd, J = 9.7, 6.4 Hz, 2H), 1.15 (d, J = 6.9 Hz, 6H), 0.94 (d, J = 11.3 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.58, 154.08,140.96, 135.68, 131.36(×2), 130.40, 128.01, 127.69, 127.33, 125.74(×2), 124.10, 122.29, 120.77, 110.66(×2), 55.25, 52.90, 51.66, 49.20, 46.20, 41.37, 40.66, 35.36, 33.03, 29.10, 29.02, 28.08, 28.03, 24.00, 23.98. HRMS calcd for C32H40F6N5O3S, [M + H]+, 688.2678; found 688.2754. HPLC: tR = 13.667 min, 95.6%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-acetic acid (17): Compound 13 (0.2 g, 0.3 mmol) was dissolved in MeOH (5 mL) and 1 mol/L NaOH (5 mL) was added and the mixture was stirred at room temperature for 2 h. After concentration, the residue was dissolved in H2O (10 mL). Then, 1 mol/L HCl (5 mL) was added to the mixture, the mixture was extracted with EtOAc (10 mL × 3), and the combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (DCM:MeOH = 5:1) to give 17 (0.17 g, 86.4%) as a colourless oil. 1H-NMR (600 MHz, DMSO-d6) δ 7.92 (s, 1H), 7.65 (s, 2H), 7.00 (dd, J = 8.5, 2.2 Hz, 1H), 6.77 (dd, J = 7.9, 5.4 Hz, 2H), 5.35 (s, 2H), 4.57–4.45 (m, 2H), 3.97 (t, J = 10.4 Hz, 2H), 3.63 (s, 3H), 2.67 (dt, J = 13.7, 6.9 Hz, 1H), 2.33 (d, J = 17.9 Hz, 1H), 2.02 (d, J = 18.0 Hz, 1H), 1.76 (s, 2H), 1.36 (t, J = 6.4 Hz, 2H), 1.04 (d, J = 6.9 Hz, 6H), 0.90 (d, J = 14.3 Hz, 6H). 13C-NMR (150 MHz, DMSO-d6) δ: 169.21, 167.87, 154.37, 142.28, 140.54, 134.58, 130.63(×2), 130.29, 127.81, 127.74, 127.62, 125.85, 124.50, 122.69, 120.98, 111.32(×2), 55.70, 53.85, 51.65, 49.55, 40.57, 35.36, 32.75, 29.24, 29.07, 28.46, 28.09, 24.27, 24.21. HRMS calcd for C31H36F6N5O3, [M + H]+, 640.2644; found 640.2709. HPLC: tR = 19.013 min, 97.3%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-butyric acid (18): Colourless oil; yield 81.3%; 1H-NMR (400 MHz, DMSO-d6) δ 12.18 (s, 1H), 7.90 (s, 1H), 7.65 (s, 2H), 6.99 (d, J = 8.5 Hz, 1H), 6.78 (d, J = 8.5 Hz, 2H), 4.50 (s, 2H), 4.44 (t, J = 6.8 Hz, 2H), 4.00–3.90 (m, 2H), 3.62 (s, 3H), 2.66 (dt, J = 13.7, 6.8 Hz, 1H), 2.32 (d, J = 17.7 Hz, 1H), 2.19–2.15(m, 2H), 2.07–1.98 (m, 3H), 1.75 (s, 2H), 1.35 (t, J = 5.9 Hz, 2H), 1.03 (d, J = 6.9 Hz, 6H), 0.88 (d, J = 7.5 Hz, 6H). 13C-NMR (150 MHz, DMSO-d6) δ: 173.76, 169.19, 154.39, 142.35, 140.54, 134.46, 130.61(×2), 127.88, 127.74, 127.72(×2), 125.84, 124.51, 122.70, 120.94, 111.33(×2), 55.69, 51.88, 51.70, 49.66, 40.63, 35.36, 32.76, 30.40, 29.25, 29.05, 28.40, 28.13, 24.44, 24.25, 24.21. HRMS calcd for C33H40F6N5O3, [M + H]+, 668.2957; found 668.3050. HPLC: tR = 18.767 min, 97.1%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-ethyl alcohol (19): Compound 13 (0.4 g, 0.6 mmol) was dissolved in THF (5 mL) and NaBH4 (0.05 g, 1.2 mmol) and 2 drops of MeOH were added and the mixture was stirred at 70 °C for 2 h and then cooled to room temperature. After concentration, the residue was dissolved in EtOAc (10 mL), washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 19 (0.3 g, 79.8%) as a colourless oil. 1H-NMR (600 MHz, DMSO-d6) δ 7.92 (s, 1H), 7.67 (s, 2H), 7.00 (dd, J = 8.5, 2.2 Hz, 1H), 6.78 (d, J = 8.5 Hz, 1H), 6.77 (d, J = 2.3 Hz, 1H), 4.95 (t, J = 5.6 Hz, 1H), 4.49 (s, 2H), 4.42 (t, J = 5.4 Hz, 2H), 3.97 (q, J = 14.4 Hz, 2H), 3.78 (dd, J = 10.8, 5.5 Hz, 2H), 3.62 (s, 3H), 2.67 (dt, J = 13.8, 6.9 Hz, 1H), 2.33 (d, J = 18.0 Hz, 1H), 2.02 (d, J = 18.0 Hz, 1H), 1.76 (s, 2H), 1.35 (t, J = 6.4 Hz, 2H), 1.04 (d, J = 6.9 Hz, 6H), 0.89 (d, J = 16.3 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.30, 154.11, 141.18, 140.89, 135.43, 131.32(×2), 130.51, 128.04, 127.72, 127.55, 125.67, 124.13, 122.32, 120.71, 110.63(×2), 60.30, 55.23, 54.95, 51.80, 49.41, 40.68, 35.40, 33.03, 29.12, 29.02, 28.04(×2), 23.99, 23.98. HRMS calcd for C31H38F6N5O2, [M + H]+, 626.2851; found 626.2920. HPLC: tR = 16.776 min, 95.8%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2H-tetrazol-5-amine-2-butanol (20): Colourless oil; yield 73.8%; 1H-NMR (400 MHz, DMSO-d6) δ 7.92 (s, 1H), 7.67 (s, 2H), 7.02 (dd, J = 8.5, 2.1 Hz, 1H), 6.83–6.76 (m, 2H), 4.52 (s, 2H), 4.48–4.39 (m, 3H), 4.03–3.92 (m, 2H), 3.64 (s, 3H), 3.37 (d, J = 5.4 Hz, 2H), 2.68 (dt, J = 13.7, 6.8 Hz, 1H), 2.40–2.29 (m, 1H), 2.06–2.00 (m, 1H), 1.89–1.80 (m, 2H), 1.77 (s, 2H), 1.40–1.28 (m, 4H), 1.05 (d, J = 6.9 Hz, 6H), 0.90 (d, J = 7.5 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.27, 154.14, 141.37, 140.85, 135.19, 131.22(×2), 130.58, 128.09, 127.77(×2), 125.63, 124.16, 122.36, 120.59, 110.62(×2), 61.78, 55.22, 52.52, 51.64, 49.35, 40.60, 35.43, 33.03, 29.12, 29.10, 29.00, 28.04(×2), 25.53, 23.99, 23.98. HRMS calcd for C33H42F6N5O2, [M + H]+, 654.3164; found 654.3257. HPLC: tR = 15.755 min, 96.4%.
2-(5-((3,5-Bis(trifluoromethyl)benzyl)((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)amino)-2H-tetrazol-2-yl)acetamide (21): Compound 13 (0.2 g, 0.3 mmol) was dissolved in a saturated NH3–EtOH solution (5 mL). After being stirred at room temperature for 12 h, the reaction mixture was concentrated in vacuo. The residue was dissolved in EtOAc (10 mL), washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 21 (0.12 g, 62.6%) as a colourless oil. 1H-NMR (400 MHz, DMSO-d6) δ 7.91 (s, 1H), 7.70 (s, 1H), 7.66 (s, 2H), 7.42 (s, 1H), 6.99 (dd, J = 8.5, 2.2 Hz, 1H), 6.78–6.75(m, 2H), 5.12 (s, 2H), 4.50 (s, 2H), 3.97 (d, J = 4.6 Hz, 2H), 3.62 (s, 3H), 2.66 (dt, J = 13.7, 6.8 Hz, 1H), 2.33 (d, J = 18.0 Hz, 1H), 2.01 (d, J = 17.7 Hz, 1H), 1.76 (s, 2H), 1.35 (t, J = 6.2 Hz, 2H), 1.04 (d, J = 6.9 Hz, 6H), 0.89 (d, J = 10.9 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.77, 154.08, 140.96, 140.88, 131.43(×2), 130.42, 127.99(×2), 127.73, 127.29, 125.77(×2), 124.07, 122.26, 120.84, 110.68(×2), 55.24, 54.83, 51.87, 49.43, 40.79, 35.35, 33.04, 29.13, 29.03, 28.10, 28.00, 24.01, 23.98. HRMS calcd for C31H37F6N6O2, [M + H]+, 639.2804; found 639.2909. HPLC: tR = 12.757 min, 98.2%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-5-methyl-1,2,4-oxadiazol-3-amine (22): Intermediate 11 (0.3 g, 0.6 mmol) was dissolved in EtOH (5 mL) and triethylamine (0.08 mL, 0.6 mmol) and hydroxylamine hydrochloride (0.04 g, 0.6 mmol) were added. After being stirred at 80 °C for 2 h, the reaction mixture was cooled to room temperature and concentrated in vacuo. Then, the residue was added pyridine (5 mL) and acetic anhydride (0.07 mL, 0.7 mmol). After being stirred at 80 °C for 12 h, the reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc (10 mL), washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 22 (0.15 g, 41.9%) as a colourless oil. 1H-NMR (600 MHz, CDCl3) δ 7.66 (s, 1H), 7.50 (s, 2H), 7.00 (dd, J = 8.4, 2.1 Hz, 1H), 6.72 (d, J = 2.0 Hz, 1H), 6.67 (d, J = 8.4 Hz, 1H), 4.43 (q, J = 16.2 Hz, 2H), 4.06 (d, J = 14.5 Hz, 1H), 3.90 (d, J = 14.5 Hz, 1H), 3.65 (s, 3H), 2.73 (dt, J = 13.8, 6.9 Hz, 1H), 2.47–2.43 (m, 4H), 2.08 (d, J = 18.2 Hz, 1H), 1.81 (s, 2H), 1.42 (dq, J = 13.0, 6.5 Hz, 2H), 1.13 (d, J = 6.9 Hz, 6H), 0.95 (d, J = 13.1 Hz, 6H). 13C- NMR (150 MHz, CDCl3) δ: 175.39, 170.36, 154.07, 140.92, 140.85, 135.59, 131.31(×2), 130.37, 128.00, 127.54, 127.35, 125.66, 124.13, 122.32, 120.70, 110.62(×2), 55.22, 51.08, 48.61, 40.40, 35.41, 33.01, 29.04, 29.01, 28.10, 27.97, 23.96(×2), 12.63. HRMS calcd for C31H36F6N3O2, [M + H]+, 596.2633; found 596.2716. HPLC: tR = 9.953 min, 95.8%.
2-(2-(Chloromethyl)-4,4-dimethylcyclohex-1-enyl)-4-isopropyl-1-methoxybenzene (23): Intermediate 9 (114.5 mg, 0.4 mmol) was dissolved in ethanol (5 mL). Sodium borohydride (19.0 mg, 0.5 mmol) was added to the mixture. After being stirred at room temperature for 30 min, the reaction mixture was poured into H2O (20 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. SOCl2 (0.1 mL, 1.4 mmol) was added to a solution of the above residue in DMF (2 mL) cooled to 0 °C. After being stirred at room temperature for 1 h, the reaction mixture was poured into H2O (10 mL) and was extracted with EtOAc (5 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 10:1) to give 23 (92.5 mg, 75.3% in two steps), which was used immediately for the next step because of its instability. HPLC: tR = 7.706 min, 96.8%.
tert-Butyl 4-(3,5-bis(trifluoromethyl)benzylamino)piperidine-1-carboxylate (26): tert-butyl 4-amino-piperidine-1-carboxylate (1.0 g, 5.0 mmol) and 3,5-bis(trifluoromethyl) benzaldehyde (1.0 g, 4.1 mmol) were dissolved in MeOH (15 mL). After stirring for 3 h at room temperature, NaBH4 (0.3 g, 7.0 mmol) was added to the mixture. The reaction mixture was stirred at room temperature for 30 min and then poured into a saturated sodium bicarbonate solution (20 mL). The mixture was extracted with CH2Cl2 (20 mL × 3) and the combined organic layers were washed with water (20 mL × 3) and brine (20 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 26 (1.4 g, 80.0%) as a pale yellow oil. 1H-NMR (600 MHz, CDCl3) δ 7.83 (s, 2H), 7.76 (s, 1H), 4.04 (s, 2H), 3.96 (s, 2H), 2.82 (s, 2H), 2.71–2.63 (m, 1H), 1.88 (d, J = 11.2 Hz, 2H), 1.46 (s, 9H), 1.35–1.26 (m, 2H). HPLC: tR = 13.657 min, 95.7%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)- methyl)piperidine-4-amine-1-carboxylic acid tert-butyl ester (28): NaH (30.0 mg, 0.7 mmol, 60% in oil) was added to a solution of intermediate 26 (213.1 mg, 0.5 mmol) in DMF (5 mL) cooled to 0 °C. After stirring at 0 °C for 30 min, a solution of intermediate 23 (153.4 mg, 0.5 mmol) in DMF (5 mL) was added. The reaction mixture was allowed to warm to room temperature and stirred for 30 min, and then was poured onto crushed ice. The mixture was diluted with EtOAc (15 mL) and the layers were separated. The aqueous layer was extracted with EtOAc (5 mL × 3), and the combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 20:1) to give 28 (235.9 mg, 67.7%) as a colourless oil. 1H-NMR (400 MHz, CDCl3) δ 7.83 (s, 2H), 7.73 (s, 1H), 7.08 (dd, J = 8.4, 2.2 Hz, 1H), 6.79 (d, J = 8.4 Hz, 1H), 6.74 (d, J = 2.3 Hz, 1H), 4.11 (s, 2H), 3.70 (s, 3H), 3.50 (q, J = 15.0 Hz, 2H), 2.90–2.78 (m, 2H), 2.74–2.48 (m, 4H), 2.44–2.31 (m, 1H), 2.09–2.00 (m, 1H), 1.99–1.83 (m, 2H), 1.52 (d, J = 12.0 Hz, 1H), 1.45–1.34 (m, 12H), 1.34–1.25 (m, 2H), 1.22 (dd, J = 6.9, 1.1 Hz, 6H), 0.96 (s, 6H). HPLC: tR = 7.023 min, 96.6%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)piperidin-4-amine (30): Intermediate 28 (0.2 g, 0.3 mmol) was dissolved in a trifluoroacetic acid–dichloromethane (1:1) solution (20 mL) and stirred at room temperature overnight. After concentration, the residue was dissolved in EtOAc (10 mL), washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 2:1) to give 30 (172.1 mg, 96.1%) as a colourless oil. 1H-NMR (400 MHz, DMSO-d6) δ 8.81 (s, 1H), 8.10–7.88 (m, 3H), 7.11 (dd, J = 8.4, 2.1 Hz, 1H), 6.91 (d, J = 8.5 Hz, 1H), 6.83 (d, J = 2.0 Hz, 1H), 3.66 (s, 3H), 3.57 (d, J = 2.4 Hz, 2H), 3.29 (d, J = 11.8 Hz, 2H), 2.89–2.68 (m, 6H), 2.26 (d, J = 17.6 Hz, 1H), 2.01 (s, 1H), 1.87 (d, J = 7.9 Hz, 2H), 1.67–1.48 (m, 4H), 1.35 (t, J = 6.2 Hz, 2H), 1.17 (d, J = 6.9 Hz, 6H), 0.92 (d, J = 10.5 Hz, 6H). HPLC: tR = 13.003 min, 95.9%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)piperidin-4-amine-1-ethyl alcohol (32): Intermediate 30 (0.7 g, 1.2 mmol) was dissolved in acetonitrile (10 mL) and methyl 2-bromoacetate (0.4 g, 2.4 mmol) and triethylamine (0.6 mL, 4.7 mmol) were added. The reaction mixture was stirred at room temperature for 16 h and then poured into H2O (10 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was dissolved in THF (10 mL), and sodium borohydride (0.1 g, 2.6 mmol) and 2 drops of MeOH were added. The mixture was stirred at 70 °C for 2 h and then cooled to room temperature. After concentration, the residue was dissolved in EtOAc (10 mL), washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 1:1) to give 32 (0.4 g, 52.0%) as a colourless oil. 1H-NMR (400 MHz, DMSO-d6) δ 8.01 (s, 2H), 7.93 (s, 1H), 7.08 (dd, J = 8.4, 2.0 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.74 (d, J = 2.0 Hz, 1H), 4.39 (s, 1H), 3.64 (s, 3H), 3.55 (d, J = 18.4 Hz, 2H), 3.44 (d, J = 5.9 Hz, 2H), 2.84 (d, J = 13.3 Hz, 2H), 2.78 (dd, J = 13.7, 6.8 Hz, 1H), 2.69 (s, 2H), 2.43–2.37 (m, 3H), 2.26 (d, J = 17.8 Hz, 1H), 1.96–1.77 (m, 4H), 1.44–1.42 (m, 7H), 1.16 (d, J = 6.9 Hz, 6H), 0.91 (d, J = 7.1 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 154.49, 144.61, 140.59, 132.83, 131.67, 131.25(×2), 129.57, 128.19, 128.00, 125.01, 124.38, 122.57, 120.40, 110.56(×2), 59.29, 57.73, 55.43, 55.13, 53.47, 53.39, 52.55, 52.25, 40.99, 35.60, 33.21(×2), 29.30, 28.93, 28.28, 27.72. 27.25, 24.23, 24.12. HRMS calcd for C35H47F6N2O2, [M + H]+, 641.3463; found 641.3525. HPLC: tR = 16.772 min, 97.9%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)piperidin-4-amine-1-acetic acid (33): Intermediate 30 (0.5 g, 0.9 mmol) was dissolved in acetonitrile (10 mL), and methyl 2-bromoacetate (0.3 g, 1.8 mmol) and triethylamine (0.4 mL, 3.4 mmol) were added. The reaction mixture was stirred at room temperature for 16 h and then poured into H2O (10 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was dissolved in MeOH (5 mL) and 1 mol/L NaOH (5 mL) was added and the mixture was stirred at room temperature for 2 h. After concentration, the residue was dissolved in H2O (10 mL). Then, 1 mol/L HCl (5 mL) was added to the mixture, the mixture was extracted with EtOAc (10 mL × 3), and the combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (DCM:MeOH = 5:1) to give 33 (0.4 g, 67.8%) as a colourless oil. 1H-NMR (400 MHz, DMSO-d6) δ 8.02 (s, 2H), 7.95 (s, 1H), 7.09 (dd, J = 8.5, 2.1 Hz, 1H), 6.89 (d, J = 8.5 Hz, 1H), 6.77 (d, J = 2.1 Hz, 1H), 3.65 (s, 3H), 3.63–3.51 (m, 2H), 3.20 (s, 2H), 3.14 (d, J = 10.4 Hz, 2H), 2.79 (dt, J = 13.7, 6.9 Hz, 1H), 2.70 (s, 2H), 2.62–2.51 (m, 3H), 2.26 (d, J = 17.7 Hz, 1H), 1.96 (s, 1H), 1.91–1.75 (m, 2H), 1.70–1.51 (m, 2H), 1.40 (s, 2H), 1.35 (t, J = 6.3 Hz, 2H), 1.16 (d, J = 6.8 Hz, 6H), 0.91 (d, J = 8.7 Hz, 6H). 13C-NMR (150 MHz, DMSO-d6) δ: 168.60, 154.62(×2), 145.76, 140.43, 132.53, 131.21, 130.22(×2), 129.42, 128.93, 127.74, 125.54, 124.78, 122.97, 111.48(×2), 59.03, 55.81, 54.09, 52.70, 52.43, 52.37, 51.58, 41.02, 35.50, 32.94, 29.40, 29.00, 28.53, 27.95, 24.55(×2), 24.44(×2). HRMS calcd for C35H45F6N2O3, [M + H]+, 655.3256; found 655.3345. HPLC: tR = 19.973 min, 98.1%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)piperidin-4-amine-1- methanoic acid tetrahydro-2H-pyran-4-ol ester (34): Intermediate 30 (0.2 g, 0.3 mmol) was dissolved in CH2Cl2 (5 mL), and tetrahydro-2H-pyran-4-yl carbonochloridate (0.1 g, 0.5 mmol) and triethylamine (0.2 mL, 1.4 mmol) were added. The reaction mixture was stirred at room temperature for 30 min and then poured into H2O (10 mL) and extracted with CH2Cl2 (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 34 (0.1 g, 45.9%) as a colourless oil. 1H-NMR (600 MHz, CDCl3) δ 7.82 (s, 2H), 7.73 (s, 1H), 7.08 (dd, J = 8.3, 2.0 Hz, 1H), 6.79 (d, J = 8.4 Hz, 1H), 6.73 (d, J = 2.0 Hz, 1H), 4.86–4.80 (m, 1H), 4.13 (d, J = 7.1 Hz, 2H), 3.90–3.85 (m, 2H), 3.71 (s, 3H), 3.56–3.53 (m, 3H), 3.49–3.41 (m, 1H), 2.84–2.81 (m, 2H), 2.75–2.53 (m, 4H), 2.37 (d, J = 17.7 Hz, 1H), 2.02 (s, 1H), 1.93–1.90 (m, 3H), 1.87 (s, 1H), 1.65–1.63 (m, 3H), 1.55 (d, J = 12.5 Hz, 1H), 1.46–1.37 (m, 3H), 1.35–1.29 (m, 1H), 1.22 (dd, J = 6.9, 2.4 Hz, 6H), 0.96 (s, 6H). 13C-NMR (150 MHz, CDCl3) δ: 154.47(×2), 144.35, 140.64, 131.57, 131.32, 131.10, 129.35, 128.18(×2), 125.09(×2), 124.35, 122.54, 120.51, 110.61(×2), 69.63, 65.30(×2), 55.44(×2), 55.35, 52.46, 52.23, 43.82, 43.66, 41.07, 35.57, 33.19, 32.08(×2), 29.24, 28.94, 28.33, 27.64, 24.24, 24.11(×2). HRMS calcd for C39H51F6N2O4, [M + H]+, 725.3675; found 725.3751. HPLC: tR = 15.553 min, 96.0%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((5′-isopropyl-2′-methoxy-4,4-dimethyl-3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-yl)methyl)azetidin-3-amine-1-methanoic acid ethyl ester (35): Intermediate 31 (0.2 g, 0.3 mmol) was dissolved in CH2Cl2 (5 mL), and ethyl chloroformate (0.1 g, 0.7 mmol) and triethylamine (0.2 mL, 1.4 mmol) were added. The reaction mixture was stirred at room temperature for 30 min and then poured into H2O (10 mL) and extracted with CH2Cl2 (10 mL × 3). The combined organic layers were washed with H2O (10 mL × 3) and brine (10 mL × 3), dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel (petroleum ether:EtOAc = 4:1) to give 35 (0.2 g, 89.2%) as a colourless oil. 1H-NMR (400 MHz, CDCl3) δ 7.78 (s, 2H), 7.76 (s, 1H), 7.09 (dd, J = 8.4, 2.1 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.74 (d, J = 2.1 Hz, 1H), 4.07 (q, J = 7.1 Hz, 2H), 3.88–3.81 (m, 1H), 3.80–3.72 (m, 3H), 3.71 (s, 3H), 3.68–3.56 (m, 2H), 3.55–3.47 (m, 1H), 2.90–2.77 (m, 2H), 2.77–2.65 (m, 1H), 2.43–2.29 (m, 1H), 2.10–2.04 (m, 1H), 2.01–1.82 (m, 2H), 1.45–1.35 (m, 2H), 1.24–1.19 (m, 9H), 0.96 (s, 6H). 13C-NMR (150 MHz, CDCl3) δ: 156.60(×2), 154.30(×2), 140.75, 131.29(×2), 131.14, 128.37, 127.94(×2), 125.39, 124.21, 122.41, 120.87, 110.55(×2), 60.92, 55.32, 54.44, 53.43, 41.59, 35.49, 33.14(×2), 29.38, 28.97, 28.22, 27.80, 24.18, 24.12(×2), 14.60(×2). HRMS calcd for C34H43F6N2O3, [M + H]+, 641.3100; found 641.3180. HPLC: tR = 11.763 min, 96.7%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((5′-isopropyl-2′-methoxy-4,4-dimethyl-3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-yl)methyl)azetidin-3-amine-1-methanoic acid isopropyl ester (36): Colourless oil; yield 69.3%; 1H-NMR (400 MHz, CDCl3) δ 7.77 (s, 2H), 7.76 (s, 1H), 7.09 (dd, J = 8.4, 2.2 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.74 (d, J = 2.2 Hz, 1H), 4.84 (dt, J = 12.5, 6.2 Hz, 1H), 3.83 (t, J = 8.4 Hz, 1H), 3.77 (d, J = 7.9 Hz, 1H), 3.73 (d, J = 5.4 Hz, 2H), 3.70 (s, 3H), 3.67–3.56 (m, 2H), 3.55–3.47 (m, 1H), 2.89–2.77 (m, 2H), 2.76–2.64 (m, 1H), 2.40–2.28 (m, 1H), 2.09–2.04 (m, 1H), 2.00–1.83 (m, 2H), 1.44–1.35 (m, 2H), 1.22 (d, J = 6.9 Hz, 6H), 1.19 (d, J = 6.2 Hz, 6H), 0.96 (d, J = 2.8 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 156.38(×2), 154.32(×2), 140.72, 131.48, 131.26, 131.19, 128.34, 127.93(×2), 125.35, 124.23, 122.43, 120.83, 110.53(×2), 68.17, 55.31(×2), 54.44, 53.43, 51.10, 41.56, 35.50, 33.14, 29.38, 28.96(×2), 28.23, 27.79, 24.19, 24.13, 22.11(×2). HRMS calcd for C35H45F6N2O3, [M + H]+, 655.3256; found 655.3344. HPLC: tR = 10.757 min, 96.8%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((5′-isopropyl-2′-methoxy-4,4-dimethyl-3,4,5,6-tetrahydro-[1,1′-biphenyl]-2-yl)methyl)azetidin-3-amine-1-methanoic acid tetrahydro-2H-pyran-4-ol ester (37): Colourless oil; yield 73.6%; 1H NMR (600 MHz, CDCl3) δ 7.78 (s, 2H), 7.76 (s, 1H), 7.09 (dd, J = 8.4, 2.1 Hz, 1H), 6.80 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 2.2 Hz, 1H), 4.79 (tt, J = 8.4, 4.0 Hz, 1H), 3.89–3.83 (m, 3H), 3.82–3.78 (m, 1H), 3.75 (s, 2H), 3.71 (s, 3H), 3.62 (s, 2H), 3.55–3.49 (m, 3H), 2.90–2.77 (m, 2H), 2.73 (s, 1H), 2.39–2.30 (m, 1H), 2.09–2.02 (m, 1H), 2.00–1.94 (m, 1H), 1.92–1.85 (m, 3H), 1.64–1.59 (m, 2H), 1.45–1.34 (m, 2H), 1.22 (d, J = 6.9 Hz, 6H), 0.97 (d, J = 4.2 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 155.77(×2), 154.30, 140.76, 131.53, 131.31, 131.14, 128.36, 127.90(×3), 125.40, 124.20, 122.40, 120.92, 110.57(×2), 69.44, 65.26(×2), 55.33(×2), 54.49, 53.46, 41.59, 35.48, 33.14(×2), 32.07(×2), 29.40, 28.98(×2), 28.23, 27.82, 24.20, 24.13. HRMS calcd for C37H47F6N2O4, [M + H]+, 697.3362; found 697.3459. HPLC: tR = 16.003 min, 95.8%.
N-(3,5-Bis(trifluoromethyl)benzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)-methyl)-2-methyl-2H-tetrazol-5-amine (40): The title compound was obtained in a manner similar to that described for the preparation of intermediate 28. Colourless oil; yield 70.5%; 1H-NMR (600 MHz, DMSO-d6) δ 7.92 (s, 1H), 7.64 (s, 2H), 6.98 (dd, J = 8.4, 2.2 Hz, 1H), 6.77–6.73 (m, 2H), 4.50 (s, 2H), 4.10 (s, 3H), 3.99–3.93 (m, 2H), 3.60 (s, 3H), 2.65 (dt, J = 13.7, 6.9 Hz, 1H), 2.32 (d, J = 18.0 Hz, 1H), 2.01 (d, J = 18.0 Hz, 1H), 1.79–1.72 (m, 2H), 1.35 (t, J = 6.4 Hz, 2H), 1.02 (d, J = 6.9 Hz, 6H), 0.89 (d, J = 13.2 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.46, 154.12, 141.34, 140.83, 135.26, 131.26(×2), 130.54, 128.05, 127.70, 127.65, 125.61, 124.16, 122.35, 120.60, 110.59(×2), 55.21, 51.59, 49.22, 40.56, 39.20, 35.44, 33.02, 29.09, 29.02, 28.08, 27.99, 23.98(×2). HRMS calcd for C30H36F6N5O, [M + H]+, 596.2746; found 596.2847. HPLC: tR = 8.776 min, 96.1%.
N-((2-(5-Isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)methyl)-2-methyl-N-(3-(trifluoromethyl)-benzyl)-2H-tetrazol-5-amine (41): Colourless oil; yield 78.4%; 1H-NMR (600 MHz, DMSO-d6) δ 7.52 (d, J = 7.7 Hz, 1H), 7.41 (t, J = 7.7 Hz, 1H), 7.34 (s, 1H), 7.22 (d, J = 7.8 Hz, 1H), 7.02 (dd, J = 8.5, 2.3 Hz, 1H), 6.80 (d, J = 8.5 Hz, 1H), 6.77 (d, J = 2.3 Hz, 1H), 4.41 (s, 2H), 4.10 (s, 3H), 3.92 (q, J = 14.7 Hz, 2H), 3.63 (s, 3H), 2.68 (dt, J = 13.8, 6.9 Hz, 1H), 2.32 (d, J = 18.0 Hz, 1H), 2.04 (d, J = 18.1 Hz, 1H), 1.76 (s, 2H), 1.38 (t, J = 6.5 Hz, 2H), 1.05 (dd, J = 6.9, 0.8 Hz, 6H), 0.91 (d, J = 5.1 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.66, 154.26, 140.64, 139.40, 134.35, 130.80, 130.73, 128.32, 128.03, 125.36, 124.31, 124.29, 123.43, 123.40, 110.45(×2), 55.17, 51.16, 49.31, 40.42, 39.15, 35.54, 33.05, 29.19, 29.03, 28.20, 27.98, 24.09, 24.01. HRMS calcd for C29H37F3N5O, [M + H]+, 528.2872; found 528.2960. HPLC: tR = 8.335 min, 96.2%.
N-(4-Fluorobenzyl)-N-((2-(5-isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)methyl)-2-methyl-2H-tetrazol-5-amine (42): Colourless oil; yield 80.1%; 1H-NMR (600 MHz, DMSO-d6) δ 7.07–7.05 (m, 1H), 6.97–6.93 (m, 4H), 6.86–6.85 (m, 1H), 6.80 (d, J = 2.3 Hz, 1H), 4.30 (q, J = 15.8 Hz, 2H), 4.10 (s, 3H), 3.85 (dd, J = 44.8, 14.8 Hz, 2H), 3.66 (s, 3H), 2.72 (dt, J = 13.8, 6.9 Hz, 1H), 2.32 (d, J = 18.0 Hz, 1H), 2.08 (d, J = 18.0 Hz, 1H), 1.76 (s, 2H), 1.40 (t, J = 6.5 Hz, 2H), 1.09 (dd, J = 6.9, 0.6 Hz, 6H), 0.93 (s, 6H). 13C- NMR (150 MHz, CDCl3) δ: 169.67, 154.42, 140.66, 133.58, 130.93, 129.47, 129.41, 128.30, 128.14, 127.90, 125.25, 114.72, 114.58, 110.44(×2), 55.23, 50.53, 48.79, 40.35, 39.12, 35.64, 33.12, 29.28, 29.04, 28.25, 28.01, 24.20, 24.06. HRMS calcd for C28H37FN5O, [M + H]+, 478.2904; found 478.2999. HPLC: tR = 7.750 min, 96.7%.
N-((2-(5-Isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)methyl)-2-methyl-N-(4-(trifluoromethyl)-benzyl)-2H-tetrazol-5-amine (43): Colourless oil; yield 67.5%; 1H-NMR (600 MHz, CDCl3) δ 7.37 (d, J = 8.1 Hz, 2H), 7.10 (d, J = 8.0 Hz, 2H), 7.02 (dd, J = 8.4, 2.3 Hz, 1H), 6.76 (d, J = 2.3 Hz, 1H), 6.70 (d, J = 8.4 Hz, 1H), 4.45 (s, 2H), 4.11 (s, 3H), 4.02 (s, 2H), 3.65 (s, 3H), 2.73 (dt, J = 13.8, 6.9 Hz, 1H), 2.44 (d, J = 18.2 Hz, 1H), 2.12 (d, J = 18.1 Hz, 1H), 1.84 (d, J = 1.6 Hz, 2H), 1.48–1.42 (m, 2H), 1.14 (dd, J = 6.9, 1.8 Hz, 6H), 0.96 (s, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.62, 154.32, 142.35, 140.67, 134.24, 130.74, 128.03, 127.98, 127.70(×3), 125.30, 124.87, 124.85, 110.43(×2), 55.20, 50.82, 49.03, 40.41, 39.16, 35.60, 33.06, 29.22, 29.05, 28.30, 27.93, 24.15, 23.98. HRMS calcd for C29H37F3N5O, [M + H]+, 528.2872; found 528.2953. HPLC: tR = 8.985 min, 96.7%.
N-((2-(5-Isopropyl-2-methoxyphenyl)-5,5-dimethylcyclohex-1-enyl)methyl)-2-methyl-N-(4-(trifluoro-methoxy)benzyl)-2H-tetrazol-5-amine (44): Colourless oil; yield 66.3%; 1H-NMR (600 MHz, DMSO-d6) δ 7.14 (d, J = 8.1 Hz, 2H), 7.06–7.01 (m, 3H), 6.82 (d, J = 8.5 Hz, 1H), 6.78 (d, J = 2.2 Hz, 1H), 4.35 (s, 2H), 4.11 (s, 3H), 3.88 (m, 2H), 3.66 (s, 3H), 2.70 (dt, J = 13.8, 6.9 Hz, 1H), 2.33 (d, J = 18.4 Hz, 1H), 2.06 (d, J = 18.0 Hz, 1H), 1.76 (s, 2H), 1.39 (t, J = 6.4 Hz, 2H), 1.06 (dd, J = 6.9, 1.0 Hz, 6H), 0.92 (d, J = 1.6 Hz, 6H). 13C-NMR (150 MHz, CDCl3) δ: 169.66, 154.37, 140.66, 136.94, 133.96, 130.81, 129.00(×3), 128.15, 128.11, 125.29, 120.46(×2), 110.43(×2), 55.19, 50.66, 48.76, 40.39, 39.13, 35.61, 33.10, 29.21, 29.03, 28.25, 27.97, 24.14, 24.01. HRMS calcd for C29H37F3N5O2, [M + H]+, 544.2821; found 544.2898. HPLC: tR = 8.302 min, 97.3%.

3.2. In Vitro Test for CETP Inhibitory Activity

All tested compounds were dissolved in 100% DMSO, making sure the compound was dissolved totally. The solution was vibrated hard on an oscillator for more than 30 s and then stored in a nitrogen cabinet. The stock solutions (10 mM) were diluted with DMSO for an 8-point titration (1:5 serial dilutions) in a 96-well dilution plate. The activity was estimated in accordance with the instruction for the CETP inhibitor screening kit and recombinant CETP. Compounds were tested at eight concentrations, and the fluorescence intensity were measured using a fluorometer (ExEm = 465/535 nm). The IC50 was determined from a curve fit of the data with each concentration tested three times.

3.3. Cytochrome P450 Inhibition Assay

Five specific probe substrates (CYP3A4, 2 μM midazolam; CYP2D6, 5 μM dextromethorphan; CYP2C9, 5 μM diclofenac; CYP1A2, 10 μM phenacetin; CYP2C19, 30 μM S-mephenytoin) were used to evaluated cytochrome P450 inhibition in human liver microsomes (0.25 mg/mL). A mixture of 20 μL specific probe substrate solution and 20 μL buffer solution (100 mM K3PO4, 33 mM MgCl2, pH = 7.4) was added compound (2 μL) and 158 μL human liver microsomes solution (0.25 mg/mL). The mixture were incubated at 37 °C for 10 min and another 10 min underwent after 20 μL NADPH solution added. Consecutively, 400 µL cold stop solution (200 ng/mL tolbutamide and 200 ng/mL labetalol in acetonitrile) was added to terminated the reaction. After the reactions were terminated, The plates were centrifuged (4000 rpm) at room temperature for 20 min, and the supernatants were analysed by LC/MS/MS.

3.4. Metabolic Stability Study

Ten μL (100 μM/L) of compound and 80 μL liver microsomes were mixture and incubated at 37 °C for 10 min, and then 10 μL NADPH regenerating system was added. Samples were obtained at 0 min, 5 min, 10 min, 20 min, 30 min and 60 min respectively, and 300 μL stop solution (cold in 4 °C, including 100 ng/mL tolbutamide and 100 ng/mL labetalol) was added to terminate the reaction. After oscillating for 10 min, the plates were centrifuged (4000 rpm) at room temperature for 20 min, and the supernatants were used for analyzation.

4. Conclusions

A series of novel N,N-substituted amine derivatives were designed by utilizing bioisosterism. New compounds were synthesized and evaluated for their inhibitory activity against CETP by a BODIPY-CE fluorescence assay. Compound 17 was identified as a promising CETP inhibitor with good inhibitory activity (IC50 = 0.38 ± 0.08 μM), and compound 17 demonstrated weak human/rat liver microsomes stability and low CYP inhibition.

Supplementary Materials

Supplementary File 1

Acknowledgments

We gratefully acknowledge the financial support from the National Natural Science Foundation of China (Grant 81373324), and Program for Innovative Research Team of the Ministry of Education, and Program for Liaoning Innovative Research Team in University.

Author Contributions

Xinran Wang and Lijuan Hao designed and carried out the experimental and wrote the paper; Xuanqi Xu, Wei Li and Chunchi Liu assisted in experiment; Dongmei Zhao and Maosheng Cheng supervised the whole experiment and provided technical guidance. All authors have read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Le Goff, W.; Guerin, M.; Chapman, M.J. Pharmacological modulation of cholesteryl ester transfer protein, a new therapeutic target in atherogenic dyslipidemia. Pharmacol. Ther. 2004, 101, 17–38. [Google Scholar] [CrossRef] [PubMed]
  2. Barter, P.J. Hugh Sinclair Lecture: The regulation and remodelling of HDL by plasma factors. Atheroscler. Suppl. 2002, 3, 39–47. [Google Scholar] [CrossRef]
  3. Curb, J.D.; Abbott, R.D.; Rodriguez, B.L.; Masaki, K.; Chen, R.; Sharp, D.S.; Tall, A.R. A prospective study of HDL-C and cholesteryl ester transfer protein gene mutations and the risk of coronary heart disease in the elderly. J. Lipid Res. 2004, 45, 948–953. [Google Scholar] [CrossRef] [PubMed]
  4. Barter, P.; Gotto, A.M.; LaRosa, J.C.; Maroni, J.; Szarek, M.; Grundy, S.M.; Kastelein, J.J.P.; Bittner, V.; Fruchart, J.C. HDL cholesterol, very low levels of LDL cholesterol, and cardiovascular events. N. Engl. J. Med. 2007, 357, 1301–1310. [Google Scholar] [CrossRef] [PubMed]
  5. Di Angelantonio, E.; Sarwar, N.; Perry, P.; Kaptoge, S.; Ray, K.K.; Thompson, A.; Wood, A.M.; Lewington, S.; Sattar, N.; Packard, C.J.; et al. Major Lipids, Apolipoproteins, and Risk of Vascular Disease. JAMA 2009, 302, 1993–2000. [Google Scholar] [PubMed]
  6. De Grooth, G.J.; Klerkx, A.H.E.M.; Stroes, E.S.G.; Stalenhoef, A.F.H.; Kastelein, J.J.P.; Kuivenhoven, J.A. A review of CETP and its relation to atherosclerosis. J. Lipid Res. 2004, 45, 1967–1974. [Google Scholar] [CrossRef] [PubMed]
  7. Chapman, M.J.; Le Goff, W.; Guerin, M.; Kontush, A. Cholesteryl ester transfer protein: at the heart of the action of lipid-modulating therapy with statins, fibrates, niacin, and cholesteryl ester transfer protein inhibitors. Eur. Heart J. 2010, 31, 149–164. [Google Scholar] [CrossRef] [PubMed]
  8. Mabuchi, H.; Nohara, A.; Inazu, A. Cholesteryl ester transfer protein (CETP) deficiency and CETP Inhibitors. Mol. Cells 2014, 37, 777–784. [Google Scholar] [CrossRef] [PubMed]
  9. Brousseau, M.E.; Schaefer, E.J.; Wolfe, M.L.; Bloedon, L.T.; Digenio, A.G.; Clark, R.W.; Mancuso, J.P.; Rader, D.J. Effects of an inhibitor of cholesteryl ester transfer protein on HDL cholesterol. N. Engl. J. Med. 2004, 350, 1505–1515. [Google Scholar] [CrossRef] [PubMed]
  10. Barter, P.J.; Caulfield, M.; Eriksson, M.; Grundy, S.M.; Kastelein, J.J.P.; Komajda, M.; Lopez-Sendon, J.; Mosca, L.; Tardif, J.; Waters, D.D.; et al. Effects of torcetrapib in patients at high risk for coronary events. N. Engl. J. Med. 2007, 357, 2109–2122. [Google Scholar] [CrossRef] [PubMed]
  11. Nissen, S.E.; Tardif, J.; Nicholls, S.J.; Revkin, J.H.; Shear, C.L.; Duggan, W.T.; Ruzyllo, W.; Bachinsky, W.B.; Lasala, G.P.; Tuzcu, E.M. Effect of torcetrapib on the progression of coronary atherosclerosis. N. Engl. J. Med. 2007, 356, 1304–1316. [Google Scholar] [CrossRef] [PubMed]
  12. Bots, M.L.; Visseren, F.L.; Evans, G.W.; Riley, W.A.; Revkin, J.H.; Tegeler, C.H.; Shear, C.L.; Duggan, W.T.; Vicari, R.M.; Grobbee, D.E.; et al. Torcetrapib and carotid intima-media thickness in mixed dyslipidaemia (RADIANCE 2 study): a randomised, double-blind trial. Lancet 2007, 370, 153–160. [Google Scholar] [CrossRef]
  13. Kastelein, J.J.P.; van Leuven, S.I.; Burgess, L.; Evans, G.W.; Kuivenhoven, J.A.; Barter, P.J.; Revkin, J.H.; Grobbee, D.E.; Riley, W.A.; Shear, C.L.; et al. Effect of torcetrapib on carotid atherosclerosis in familial hypercholesterolemia. N. Engl. J. Med. 2007, 356, 1620–1630. [Google Scholar] [CrossRef] [PubMed]
  14. Luscher, T.F.; Taddei, S.; Kaski, J.C.; Jukema, J.W.; Kallend, D.; Munzel, T.; Kastelein, J.J.P.; Deanfield, J.E.; Dal-VESSEL Investigators. Vascular effects and safety of dalcetrapib in patients with or at risk of coronary heart disease: the dal-VESSEL randomized clinical trial. Eur. Heart J. 2012, 33, 857–865. [Google Scholar] [CrossRef] [PubMed]
  15. Fayad, Z.A.; Mani, V.; Woodward, M.; Kallend, D.; Abt, M.; Burgess, T.; Fuster, V.; Ballantyne, C.M.; Stein, E.A.; Tardif, J.C.; et al. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): A randomised clinical trial. Lancet 2011, 378, 1547–1559. [Google Scholar] [CrossRef]
  16. Schwartz, G.G.; Olsson, A.G.; Abt, M.; Ballantyne, C.M.; Barter, P.J.; Brumm, J.; Chaitman, B.R.; Holme, I.M.; Kallend, D.; Leiter, L.A.; et al. Effects of Dalcetrapib in Patients with a Recent Acute Coronary Syndrome. N. Engl. J. Med. 2012, 367, 2089–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Tardif, J.C.; Rheaume, E.; Perreault, L.P.L.; Gregoire, J.C.; Zada, Y.F.; Asselin, G.; Provost, S.; Barhdadi, A.; Rhainds, D.; L’Allier, P.L.; et al. Pharmacogenomic Determinants of the Cardiovascular Effects of Dalcetrapib. Circ. Cardiovasc. Genet. 2015, 8, 372–382. [Google Scholar] [CrossRef] [PubMed]
  18. Nicholls, S.J.; Brewer, H.B.; Kastelein, J.J.P.; Krueger, K.A.; Wang, M.D.; Shao, M.Y.; Hu, B.; McErlean, E.; Nissen, S.E. Effects of the CETP Inhibitor Evacetrapib Administered as Monotherapy or in Combination With Statins on HDL and LDL Cholesterol A Randomized Controlled Trial. JAMA 2011, 306, 2099–2109. [Google Scholar] [CrossRef] [PubMed]
  19. Nicholls, S.J.; Lincoff, A.M.; Barter, P.J.; Brewer, H.B.; Fox, K.A.A.; Gibson, C.M.; Grainger, C.; Menon, V.; Montalescot, G.; Rader, D.; et al. Assessment of the clinical effects of cholesteryl ester transfer protein inhibition with evacetrapib in patients at high-risk for vascular outcomes: Rationale and design of the ACCELERATE trial. Am. Heart J. 2015, 170, 1061–1069. [Google Scholar] [CrossRef] [PubMed]
  20. Cannon, C.P.; Shah, S.; Dansky, H.M.; Davidson, M.; Brinton, E.A.; Gotto, A.M.; Stepanavage, M.; Liu, S.X.; Gibbons, P.; Ashraf, T.B.; et al. Safety of Anacetrapib in Patients with or at High Risk for Coronary Heart Disease. N. Engl. J. Med. 2010, 363, 2406–2415. [Google Scholar] [CrossRef] [PubMed]
  21. Brinton, E.A.; Kher, U.; Shah, S.; Cannon, C.P.; Davidson, M.; Gotto, A.M.; Ashraf, T.B.; Sisk, C.M.; Dansky, H.; Mitchel, Y.; et al. Effects of anacetrapib on plasma lipids in specific patient subgroups in the DEFINE (Determining the Efficacy and Tolerability of CETP INhibition with AnacEtrapib) trial. J. Clin. Lipidol. 2015, 9, 65–71. [Google Scholar] [CrossRef] [PubMed]
  22. Ford, J.; Lawson, M.; Fowler, D.; Maruyama, N.; Mito, S.; Tomiyasu, K.; Kinoshita, S.; Suzuki, C.; Kawaguchi, A.; Round, P.; et al. Tolerability, pharmacokinetics and pharmacodynamics of TA-8995, a selective cholesteryl ester transfer protein (CETP) inhibitor, in healthy subjects. Br. J. Clin. Pharmacol. 2014, 78, 498–508. [Google Scholar] [CrossRef] [PubMed]
  23. Hovingh, G.K.; Kastelein, J.J.P.; van Deventer, S.J.H.; Round, P.; Ford, J.; Saleheen, D.; Rader, D.J.; Brewer, H.B.; Barter, P.J. Cholesterol ester transfer protein inhibition by TA-8995 in patients with mild dyslipidaemia (TULIP): A randomised, double-blind, placebo-controlled phase 2 trial. Lancet 2015, 386, 452–460. [Google Scholar] [CrossRef]
  24. Liu, C.C.; Luo, C.Q.; Hao, L.J.; Wu, Q.; Xie, H.L.; Zhao, S.Z.; Hao, C.Z.; Zhao, D.M.; Cheng, M.S. Design, synthesis and biological evaluation of novel cholesteryl ester transfer protein inhibitors bearing a cycloalkene scaffold. Eur. J. Med. Chem. 2016, 123, 419–430. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds 1222, 3237, 4044 are available from the authors.
Figure 1. Representative CETP inhibitors.
Figure 1. Representative CETP inhibitors.
Molecules 22 01658 g001
Figure 2. Design of new structures based on compound 6.
Figure 2. Design of new structures based on compound 6.
Molecules 22 01658 g002
Scheme 1. Synthesis of target compounds 1222. Reagents and conditions: (a) DMF, PBr3, CHCl3, rt; (b) 5-isopropyl-2-methoxyphenylboronic acid, Pd(OAc)2, K2CO3, acetylacetone, EtOH, 80 °C; (c) 3,5-bis(trifluromethyl)benzyl amine, NaBH(OAc)3, 1,2-dichloroethane, rt; (d) CNBr, DIEA, THF, rt; (e) NaN3, NH4Cl, DMF, 100 °C; (f) R1-X, Et3N, acetonitrile, reflux; or (i) Boc-R1-X, Et3N, acetonitrile, reflux, (ii) TFA, DCM, rt; or R1-OH, PPh3, DIAD, rt; (g) NaBH4/MeOH, THF, reflux; or NH3-EtOH, rt; (h) (i) 1 M aq. NaOH, (ii) 1 M aq. HCl; (l) NH2OH·HCl, Ac2O, Et3N, EtOH, reflux.
Scheme 1. Synthesis of target compounds 1222. Reagents and conditions: (a) DMF, PBr3, CHCl3, rt; (b) 5-isopropyl-2-methoxyphenylboronic acid, Pd(OAc)2, K2CO3, acetylacetone, EtOH, 80 °C; (c) 3,5-bis(trifluromethyl)benzyl amine, NaBH(OAc)3, 1,2-dichloroethane, rt; (d) CNBr, DIEA, THF, rt; (e) NaN3, NH4Cl, DMF, 100 °C; (f) R1-X, Et3N, acetonitrile, reflux; or (i) Boc-R1-X, Et3N, acetonitrile, reflux, (ii) TFA, DCM, rt; or R1-OH, PPh3, DIAD, rt; (g) NaBH4/MeOH, THF, reflux; or NH3-EtOH, rt; (h) (i) 1 M aq. NaOH, (ii) 1 M aq. HCl; (l) NH2OH·HCl, Ac2O, Et3N, EtOH, reflux.
Molecules 22 01658 sch001
Scheme 2. Synthesis of target compounds 3237. Reagents and conditions: (a) (i) NaBH4, MeOH, rt, (ii) SOCl2, DMF, rt; (b) 3,5-bis(trifluromethyl)benzaldehyde, NaBH4, MeOH, rt; (c) NaH, DMF, 0 °C; (d) TFA/CH2Cl2 (1:1), rt; (e) (i) methyl 2-bromoacetate, TEA, DMF, rt, (ii) NaBH4/MeOH, THF, reflux; or 1 M aq. NaOH, 1 M aq. HCl; (f) TEA, DCM, rt.
Scheme 2. Synthesis of target compounds 3237. Reagents and conditions: (a) (i) NaBH4, MeOH, rt, (ii) SOCl2, DMF, rt; (b) 3,5-bis(trifluromethyl)benzaldehyde, NaBH4, MeOH, rt; (c) NaH, DMF, 0 °C; (d) TFA/CH2Cl2 (1:1), rt; (e) (i) methyl 2-bromoacetate, TEA, DMF, rt, (ii) NaBH4/MeOH, THF, reflux; or 1 M aq. NaOH, 1 M aq. HCl; (f) TEA, DCM, rt.
Molecules 22 01658 sch002
Scheme 3. Synthesis of target compounds 4044. Reagents and conditions: (a) 2-methyl-2H-tetrazol-5-amine, NaBH4, MeOH, rt; (b) NaH, DMF, 0 °C.
Scheme 3. Synthesis of target compounds 4044. Reagents and conditions: (a) 2-methyl-2H-tetrazol-5-amine, NaBH4, MeOH, rt; (b) NaH, DMF, 0 °C.
Molecules 22 01658 sch003
Table 1. Structures and activities of compounds 1222, 3237, 4044.
Table 1. Structures and activities of compounds 1222, 3237, 4044.
Molecules 22 01658 i028
NO.Part AR (Part B)IC50 (µM)NO.Part AR (Part B)IC50 (µM)
12 Molecules 22 01658 i001 Molecules 22 01658 i0232.84 ± 0.0433 Molecules 22 01658 i013 Molecules 22 01658 i023>50 b
13 Molecules 22 01658 i002 Molecules 22 01658 i023>50 b34 Molecules 22 01658 i014 Molecules 22 01658 i023>50 b
14 Molecules 22 01658 i003 Molecules 22 01658 i023>50 b35 Molecules 22 01658 i015 Molecules 22 01658 i023>50 b
15 Molecules 22 01658 i004 Molecules 22 01658 i0231.92 ± 0.1036 Molecules 22 01658 i016 Molecules 22 01658 i023>50 b
16 Molecules 22 01658 i005 Molecules 22 01658 i0231.20 ± 0.0137 Molecules 22 01658 i017 Molecules 22 01658 i023>50 b
17 Molecules 22 01658 i006 Molecules 22 01658 i0230.38 ± 0.0840 Molecules 22 01658 i018 Molecules 22 01658 i0230.81 ± 0.03
18 Molecules 22 01658 i007 Molecules 22 01658 i0231.60 ± 0.0241 Molecules 22 01658 i019 Molecules 22 01658 i02410.05 ± 0.06
19 Molecules 22 01658 i008 Molecules 22 01658 i0230.73 ± 0.0942 Molecules 22 01658 i020 Molecules 22 01658 i025>50 b
20 Molecules 22 01658 i009 Molecules 22 01658 i0235.76 ± 0.0343 Molecules 22 01658 i021 Molecules 22 01658 i026>50 b
21 Molecules 22 01658 i010 Molecules 22 01658 i02320.06 ± 0.1244 Molecules 22 01658 i022 Molecules 22 01658 i027>50 b
22 Molecules 22 01658 i011 Molecules 22 01658 i0230.72 ± 0.06
32 Molecules 22 01658 i012 Molecules 22 01658 i023>50 bAnacetrapib a 0.04 ± 0.01
a Used as a positive control. b Considered with no CETP inhibition activity.
Table 2. In vitro DMPK profile.
Table 2. In vitro DMPK profile.
CompoundHLM Stability a (μL/min/mg)RLM Stability b (μL/min/mg)CYPs Direct Inhibition Mean (10 μM)
3A42D62C91A22C19
17119.0146.117.7%No inhibition14.3%No inhibition3.1%
a Human liver microsomal intrinsic clearance. b Rat liver microsomal intrinsic clearance.

Share and Cite

MDPI and ACS Style

Wang, X.; Hao, L.; Xu, X.; Li, W.; Liu, C.; Zhao, D.; Cheng, M. Design, Synthesis and Biological Evaluation of N,N-Substituted Amine Derivatives as Cholesteryl Ester Transfer Protein Inhibitors. Molecules 2017, 22, 1658. https://doi.org/10.3390/molecules22101658

AMA Style

Wang X, Hao L, Xu X, Li W, Liu C, Zhao D, Cheng M. Design, Synthesis and Biological Evaluation of N,N-Substituted Amine Derivatives as Cholesteryl Ester Transfer Protein Inhibitors. Molecules. 2017; 22(10):1658. https://doi.org/10.3390/molecules22101658

Chicago/Turabian Style

Wang, Xinran, Lijuan Hao, Xuanqi Xu, Wei Li, Chunchi Liu, Dongmei Zhao, and Maosheng Cheng. 2017. "Design, Synthesis and Biological Evaluation of N,N-Substituted Amine Derivatives as Cholesteryl Ester Transfer Protein Inhibitors" Molecules 22, no. 10: 1658. https://doi.org/10.3390/molecules22101658

Article Metrics

Back to TopTop