Archives

  • 2018-07
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • casin australia Free fatty acid receptor FFAR also known as

    2022-08-03

    Free fatty casin australia receptor 1 (FFAR1, also known as GPR40) belongs to the family of G protein-coupled receptors (GPCRs) and was de-orphanized in 2003 as a receptor for medium-to long-chain free fatty acids (FFAs) [9,10]. Mainly expressed in beta cells of pancreatic islets, GPR40 can be activated by the endogenous FFAs, which subsequently elicit amplified glucose-dependent insulin secretion without high risk of hypoglycemia [9,[11], [12], [13], [14], [15]]. GPR40 is also expressed in the incretin-producing enteroendocrine L cells of the intestinal tract and its activation results in secretion of incretins such as glucagon like peptide 1 (GLP-1) [16,17]. Therefore, the ability of GPR40 to regulate glucose homeostasis by two mechanisms, glucose-stimulated insulin secretion and incretin secretion, makes it an excellent target for T2DM drug development [18,19]. Reports described a range of synthetic GPR40 agonists with desirable safety and efficacy profiles, compared with existing antidiabetic therapies such as insulin and sulfonylureas [[20], [21], [22], [23], [24], [25], [26], [27], [28]]. Among them, several small molecules were tested in the clinical trial such as TAK-875 [20,29], AMG 837 [21,30], LY2922083 and LY2881835 [23,31] (Fig. 1). The most advanced compound TAK-875 (Fasiglifam, 1) from Takeda, while providing proof of concept, was withdrawn from phase III trial due to liver toxicity [32]. The exact reason for its hepatotoxicity is currently unclear, but some reports indicated that it may be linked with high distribution of TAK-875 and TAK-875 conjugations (TAK-875 acyl glucuronide, TAK-875 glucuronide and TAK-875 taurine conjugate) in liver, as well as their ability to alter the bile acid homeostasis [[33], [34], [35]]. Moreover, GPR40 is highly expressed in the brain and can be activated by polyunsaturated FFAs [9]. The exact function of GPR40 in the brain remains unclear, but it might have correlation with antinociception, adult neurogenesis and neurovascular degeneration [[36], [37], [38], [39], [40], [41]]. Therefore, to balance the blood glucose levels, it would be more preferable to directly target the GPR40 in the pancreatic beta cells and enteroendocrine L cells rather than to elicit the undesired effects in the central nervous system (CNS) or liver. In general, properties such as total polar surface area (tPSA), LogP, molecular weight, hydrogen bond, pKa and rigidity of the molecule are closely linked to blood-brain barrier (BBB) penetration [42]. Most reported GPR40 agonists possess relatively high lipophilicity [[43], [44], [45]]. Amgen described an analogue of AMG 837 (LogP = 6.8, tPSA = 47) with significant CNS exposure (total brain-to-plasma drug distribution ratio, B/P = 0.6). Incorporation of polar groups, as in AMG-3189 (B/P = 0.04) and AMG-4668 (B/P = 0.02) (Fig. 1), substantially decreased BBB penetration [46,47]. LY2881835 (2), which finished Phase I clinical trial in 2011, possessed high in vitro potency and selectivity, as well as significant insulin and GLP-1 secretion in both in vitro and in vivo assays [23,31]. However, 2 (LogP = 6.5, tPSA = 50), like AMG-837, had a certain amount of CNS exposure (B/P = 0.14). In addition, 2 displayed high clearance in vitro (t1/2 = 18 min in human liver microsomes, t1/2 = 2 min in mouse liver microsomes) and short half-life in vivo (t1/2 = 1.3 h). We sought to modify the pharmacokinetic (PK) and physicochemical properties of 2 b y introducing polar atoms, blocking the metabolically susceptible position and cleaving the indene ring. After a systematic structure-activity relationship (SAR) study and further optimization, we identified an orally active compound 15k, which exhibited improved metabolic stability and high plasma exposure, along with lowered CNS exposure and liver to plasma distribution ratio (L/P).
    Chemistry The general synthetic approach for amide analogues is summarized in Scheme 1. The carboxylic acids of 7a-d were protected by tert-butyl group, yielding intermediates 8a-d, and subsequent bromination of 8a-d gave intermediates 9a-d. A nucleophilic substitution reaction between 9a-b and methyl 3-(4-hydroxyphenyl)hex-4-ynoic acid (16), (S)-methyl 3-(4-hydroxyphenyl)hex-4-ynoate (17), methyl 3-cyclopropyl-3-(4-hydroxyphenyl)propanoate (18), methyl 2-(6-hydroxy-2,3-dihydrobenzofuran-3-yl)acetate (19) or methyl 3-(4-hydroxyphenyl)propanoate (20) in the presence of K2CO3 produced intermediates 10a-g. Deprotection of the tert-butyl group with CF3COOH yielded acids 11a-g. 11a-g were then condensed with corresponding amines using HATU, and the resultant amide 12a-v, 14f-i and 15l-v were hydrolyzed to afforded 13a-v, 14a-d and 15a-k.