Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 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
  • Direct Mouse Genotyping Kit PBI is an agonist of GPR and act

    2022-01-14

    PBI-4050 is an agonist of GPR40 and acts as an antagonist or inverse agonist of GPR84. It cannot be excluded that other targets besides GPR40 and GPR84 could be implicated in the mechanism of action of PBI-4050 and could be explored in future studies. However, the present study, and in particular the receptor KO models, strongly supports GPR40 and GPR84 as major mediators in pathologic fibrotic pathways and as the targets of the antifibrotic effects of PBI-4050. Our data show that PBI-4050 significantly attenuated fibrosis in a variety of injury contexts, as evidenced with the antifibrotic activity observed in kidney, liver, lung, heart, pancreas, and skin fibrosis models. Given our findings with both Gpr40 and Gpr84 KO mice, both receptors appear to be involved in the fibrotic pathways. Considering their expression along the nephron, within the glomerulus, and in numerous bone marrow–derived cell types, it is likely that GPR40 and GPR84 modulate profibrotic, inflammatory, and epithelial-mesenchymal transition processes. Therefore, GPR40 may partially protect against development of fibrosis, whereas GPR84 may induce the promotion and stimulation of fibrosis, as observed with the significant increase in fibrosis in Gpr40−/− mice and reduction of fibrosis in Gpr84−/− mice. The dual modulator PBI-4050 reinforces the involvement of both GPR40 and GPR84 in multiple models of fibrosis. In the context of the adenine-induced CKD model, treatment of Gpr40 and Gpr84 KO mice with PBI-4050 suggests its antifibrosis effects to be mainly mediated by GPR40 activation, whereas inhibition of GPR84 mostly accounts for reduction of cystic lesions. The relative role of each receptor may vary depending on the type of insult, pathology, and organ. Future work will aim to elucidate the precise intracellular signaling pathways used by both GPR40 and GPR84 to regulate fibrotic events in the pathogenesis of disease. The inhibition of fibrotic and inflammatory markers by PBI-4050, found in human proximal tubule epithelial cells, podocytes, and primary fibroblasts (Figure 3), suggests that the attenuation of fibrosis Direct Mouse Genotyping Kit that we have shown in various rodent fibrosis models could translate to human disease. Moreover, significant clinical activity observed in recent phase 2 clinical trials in type 2 diabetes subjects with metabolic syndrome and in idiopathic pulmonary fibrosis patients confirm translation of pharmacologic activity of PBI-4050 in humans. Of interest, PBI-4050 was well tolerated and demonstrated a good safety profile in these two early-phase clinical trials.
    Type 2 Direct Mouse Genotyping Kit (T2DM) is a disease characterized by defects in insulin secretion from pancreatic β-cells and/or insulin resistance in target tissues of insulin., , Insulin secretagogues, such as sulfonylureas and glinides are commonly used to stimulate insulin secretion in diabetic patients. However, these drugs promote insulin secretion independent of blood glucose levels, thereby leading to the risk of hypoglycemia., GPR40 (also known as FFAR1) is a novel G protein-coupled receptor (GPCR) that is expressed in pancreatic β-cells and responds to free-fatty acid (FFA) concentrations. Activation of GPR40 potentiates glucose-stimulated insulin secretion and lowers plasma glucose concentrations in multiple animal models of insulin resistance and obesity., , Because GPR40 mediated insulin secretion is glucose-dependent, it is believed that pharmacologic activation of the receptor should not induce hypoglycemia in either fed or fasted states. Consequently, a GPR40 agonist has potential to be a safe and effective alternative to currently available therapies for T2DM., , Therapeutic efficacy has been demonstrated in several clinical trials targeting the GPR40 pathway, for example, LY-2881835 (entered Phase I clinical trial), JTT-851 (entered Phase II clinical trial, structure not disclosed) and TAK-875 (entered Phase III clinical trial).We previously described the development of the GPR40 agonist AMG 837 from a series of beta-substituted propionic acids, which were identified in a high-throughput screen. In multiple animal models, AMG 837 enhances glucose-stimulated insulin secretion and lowers plasma glucose levels. Because GPR40 activity is glucose-dependent, AMG 837 did not induce hypoglycemia in any of the models tested. Encouraged by these results, we initiated a Phase I clinical trial of AMG 837.While the clinical evaluation of AMG 837 was ongoing, we turned our attention to the development of a structurally distinct GPR40 agonist. Although AMG 837 is a carboxylic acid, its physicochemical properties, including low polar surface area (tPSA 47, ), suggest a reasonable possibility of achieving CNS exposure. Additional support comes from a structurally close analog of AMG 837 (with 4′-chloro-2′-ethoxy-(1,1′-biphenyl)-4-yl replacing 4′-(trifluoromethyl)-(1,1′-biphenyl)-3-yl of AMG 837) which showed a brain to plasma ratio of 0.6 3h after an oral dose of 5mg/kg in rats. Given that the efficacy of GPR40 agonists is derived peripherally and that they are likely to be dosed chronically, we sought molecules with minimal brain penetration. In general, increasing the polar surface area (PSA) of a molecule tends to decrease its blood–brain barrier permeability. In keeping with this principle, we focused on increasing the PSA of AMG 837, while maintaining its potency and metabolic stability. The approach taken was to introduce polar groups to the tail group and/or the head group of the AMG 837 class of GPR40 agonists.