Combinations comprising benzodioxoles as GLP-1R agonists for the treatment of NASH/NAFLD and related diseases

文档序号:473798 发布日期:2021-12-31 浏览:9次 中文

阅读说明:本技术 用于治疗nash/nafld及相关疾病的包含作为glp-1r激动剂的苯并二氧杂环戊烯的组合 (Combinations comprising benzodioxoles as GLP-1R agonists for the treatment of NASH/NAFLD and related diseases ) 是由 G·E·阿斯普内斯 S·W·巴格利 J·M·柯托 M·道林 D·J·埃德蒙斯 D·费尔南多 于 2020-05-15 设计创作,主要内容包括:本发明部分地提供包含(1)GLP-1R激动剂及(2)ACC抑制剂或DGAT2抑制剂、或KHK抑制剂或FXR激动剂的新颖组合。本发明进一步提供例如使用本申请中所述的新颖组合治疗下列疾病和障碍的新颖方法:例如,脂肪肝、非酒精性脂肪肝疾病、非酒精性脂肪性肝炎、伴有肝纤维化的非酒精性脂肪性肝炎、伴有肝硬化的非酒精性脂肪性肝炎以及伴有肝硬化与伴有肝细胞癌或伴有代谢相关疾病的非酒精性脂肪性肝炎、肥胖症和2型糖尿病。(The present invention provides, in part, novel combinations comprising (1) a GLP-1R agonist and (2) an ACC inhibitor or a DGAT2 inhibitor, or a KHK inhibitor or an FXR agonist. The invention further provides novel methods of treating the following diseases and disorders, for example using the novel combinations described in this application: for example, fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes.)

1. A combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

Or a pharmaceutically acceptable salt thereof.

2. A method of treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

3. A method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprises administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

4. The method of claim 2, wherein the disease or condition is fatty liver.

5. The method of claim 2, wherein the disease or condition is nonalcoholic fatty liver disease.

6. The method of claim 2, wherein the disease or condition is non-alcoholic steatohepatitis.

7. The method of claim 2, wherein the disease or disorder is non-alcoholic steatohepatitis with liver fibrosis.

8. The method of claim 2, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis.

9. The method of claim 2, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

10. The method of claim 2, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

11. A combination according to claim 1 or a method according to claims 2 to 10 wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

12. A combination or method according to claim 11 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

13. The combination or method of claim 12, wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

14. The combination of claim 1 or the method of claims 2 to 10, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

15. The combination or method of claim 14, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

16. The combination or method of claim 15, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

17. The combination or method of any one of claims 1 to 16, wherein the 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or pharmaceutically acceptable salt thereof is the 2-amino-2- (hydroxymethyl) propan-1, 3-dio-late salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid.

18. The combination or method of claim 17, wherein the 2-amino-2- (hydroxymethyl) propane-1, 3-dioi salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is in crystalline form.

19. The combination or method of claim 18, wherein the ratio of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and 2-amino-2- (hydroxymethyl) propane-1, 3-diol in the crystalline form is 1: 1.

20. The combination or method of claim 18 or 19, wherein the crystalline form is an anhydrous crystalline form.

21. The combination or process of claim 20, wherein the anhydrous crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.6, 10.7, and 11.32 Θ, ± 0.2 ° 2 Θ.

22. The combination or process of claim 20 or 21, wherein the anhydrous crystalline form has a crystal size comprised between 1511, 1561 and 1615cm-1,±2cm-1Raman spectrum of the peak shift of (a).

23. The combination or method of any one of claims 20 to 22, wherein the anhydrous crystalline form has chemical shifts comprised between 22.9, 146.2 and 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

24. The combination or method of claim 23, wherein the anhydrous crystalline form has analytical parameters selected from the group consisting of: included at 1511 and 1615cm-1,±2cm-1And a Raman spectrum of peak shift (D) and a spectrum of peaks at 22.9, 146.2 or 161.9ppm, +/-0.2 ppmAt least one chemical shift13C ssNMR spectrum.

25. The combination or process of claim 18 or 19, wherein the crystalline form is a trihydrate crystal.

26. The combination or process of claim 25, wherein the trihydrate crystals have a PXRD pattern comprising peaks at diffraction angles of 8.4, 9.0, and 10.52 Θ, ± 0.2 ° 2 Θ.

27. The combination or process of claim 25 or 26, wherein the trihydrate crystals have a crystal size comprised between 1507, 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (a).

28. The combination or process of any one of claims 25 to 27, wherein the trihydrate crystals have chemical shifts comprised between 19.2, 149.5 and 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

29. The combination or method of claim 28, wherein the trihydrate crystals have analytical parameters selected from the group consisting of:

PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta,

contained in 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (A), and

comprising at least one chemical shift in the range of 19.2, 149.5 or 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

30. The combination or method of claim 28, wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta and PXRD patterns containing peaks at 1507, 1557 or 1610cm -1,±2cm-1At least one peak shift of the raman spectrum.

31. The combination or method of claim 28, wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns comprising peaks at diffraction angles of 8.4 and 9.02 theta, + -0.2 DEG 2 theta and PXRD patterns comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm, + -0.2 ppm13C ssNMR spectrum.

32. The method of any one of claims 2 to 31, wherein the GLP-1R antagonist and the 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or salt thereof are administered simultaneously.

33. The method of claim 32, wherein the GLP-1R antagonist and the 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or salt thereof are present in the same pharmaceutical composition, or wherein the GLP-1R antagonist and the 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or salt thereof are each present in different pharmaceutical compositions, and wherein each of said compositions independently further comprises a pharmaceutically acceptable excipient.

34. The method of any one of claims 2 to 31, wherein said GLP-1R antagonist and said 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or salt thereof are administered sequentially and in any order, wherein said GLP-1R antagonist and said 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or salt thereof are each present in different pharmaceutical compositions, and wherein each of said compositions independently further comprises a pharmaceutically acceptable excipient.

35. The combination of claim 1 and any one of claims 11 to 31, wherein the composition additionally comprises at least one other agent.

36. The method of any one of claims 2 to 34, wherein the method further comprises administering to said patient at least one additional agent.

37. The combination of claim 35 or method of claim 36, wherein the at least one other agent is selected from the group consisting of: acetyl-CoA carboxylase- (ACC) inhibitors, diacylglycerol O-acyltransferase 1(DGAT-1) inhibitors, monoacylglycerol O-acyltransferase inhibitors, Phosphodiesterase (PDE) -10 inhibitors, AMPK activators, sulfonylureas, meglitinides, alpha-amylase inhibitors, alpha-glucosidase hydrolase inhibitors, alpha-glucosidase inhibitors, PPAR γ agonists, PPAR α/γ agonists, biguanides, glucagon-like peptide 1(GLP-1) modulators, liraglutide, albiglutide, exenatide, albiglutide, lixivide, dolaglutide, somagluteline, protein tyrosine phosphatase-1B (PTP-1B) inhibitors, SIRT-1 activators, dipeptidyl peptidase IV (DPP-IV) inhibitors, DPP-1 inhibitors, and methods of using the same, Insulin secretagogues, fatty acid oxidation inhibitors, A2 antagonists, c-jun amino terminal kinase (JNK) inhibitors, glucokinase activators (GKa), insulin mimetics, glycogen phosphorylase inhibitors, VPAC2 receptor agonists, SGLT2 inhibitors, glucagon receptor modulators, GPR119 modulators, FGF21 derivatives or analogs, TGR5 receptor modulators, a GPBAR1 receptor modulator, a GPR40 agonist, a GPR120 modulator, a high affinity nicotinic acid receptor (HM74A) activator, an SGLT1 inhibitor, a carnitine palmitoyl transferase inhibitor or modulator, a fructose 1, 6-bisphosphatase inhibitor, an aldose reductase inhibitor, a mineralocorticoid receptor inhibitor, a TORC2 inhibitor, a CCR2 and/or CCR5 inhibitor, a PKC isoform (e.g., PKC α, PKC β, PKC γ) inhibitor, a fatty acid synthase inhibitor, a serine palmitoyl transferase inhibitor; a GPR81 modulator, a GPR39 modulator, a GPR43 modulator, a GPR41 modulator, a GPR105 modulator, a kv1.3 modulator, a retinol binding protein 4 modulator, a glucocorticoid receptor modulator, a somatostatin receptor modulator; PDHK2 or PDHK4 inhibitors or modulators, MAP4K4 inhibitors, modulators of the IL1 family including IL1 β, HMG-CoA reductase inhibitors, squalene synthetase inhibitors, fibrates, bile acid sequestrants, ACAT inhibitors, MTP inhibitors, lipoxygenase inhibitors, cholesterol absorption inhibitors, PCSK9 modulators, cholesteryl ester transfer protein inhibitors, and rxra modulators.

38. The combination of claim 35 or method of claim 36, wherein the at least one other agent is selected from the group consisting of: cysteamine or a pharmaceutically acceptable salt thereof, cystamine or a pharmaceutically acceptable salt thereof, an antioxidant compound, lecithin, a vitamin B complex, a bile salt formulation, a cannabinoid-1 (CB1) receptor antagonist, a cannabinoid-1 (CB1) receptor inverse agonist, a peroxisome proliferator activated receptor activity modulator, benzothiazepineOr a benzothiepine compound, an RNA antisense construct that inhibits the protein tyrosine phosphatase PTPRU, a heteroatom-linked substituted piperidine and derivatives thereof, an azacyclopentane derivative capable of inhibiting stearoyl-coenzyme α δ -9 desaturase, an acyl amide compound having the secretagogue activity or inducer activity of adiponectin, a quaternary ammonium compound, glatiramer acetate, a pentraxin protein, an HMG-CoA reductase inhibitor, N-acetyl cysteine, an isoflavone compound, a macrolide antibiotic, a galectin inhibitor, an antibody, or any combination of the above.

39. A combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

40. A method of treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

41. A method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprises administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

42. The method of claim 40, wherein the disease or condition is fatty liver.

43. The method of claim 40, wherein said disease or condition is nonalcoholic fatty liver disease.

44. The method of claim 40, wherein said disease or condition is non-alcoholic steatohepatitis.

45. The method of claim 40, wherein the disease or disorder is non-alcoholic steatohepatitis with liver fibrosis.

46. The method of claim 40, wherein said disease or condition is nonalcoholic steatohepatitis with cirrhosis.

47. The method of claim 40, wherein said disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

48. The method of claim 40, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

49. The combination of claim 39 or the method of any one of claims 40 to 48, wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

50. A combination or method according to claim 49 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

51. The combination or method of claim 50, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

52. The combination of claim 39 or the method of any one of claims 40 to 48, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

53. The combination or method of claim 52, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

54. The combination or method of claim 53, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

55. The combination or method of any one of claims 39 to 54 wherein the [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or salt thereof is a crystalline form of [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid.

56. A combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

57. A method of treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

58. A method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or alleviating a medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

Or a pharmaceutically acceptable salt thereof.

59. The method of claim 58, wherein said disease or condition is fatty liver.

60. The method of claim 58, wherein said disease or condition is nonalcoholic fatty liver disease.

61. The method of claim 58, wherein said disease or condition is non-alcoholic steatohepatitis.

62. The method of claim 58, wherein said disease or disorder is non-alcoholic steatohepatitis with liver fibrosis.

63. The method of claim 58, wherein said disease or condition is nonalcoholic steatohepatitis with cirrhosis.

64. The method of claim 58, wherein said disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

65. The combination or method of claim 58, wherein the disease or condition is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

66. The combination of claim 56 or a method of 57 to 65, wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

67. The combination or method of claim 66 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

68. The combination or method of claim 67, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

69. The combination of claim 56 or the method of 57 to 65, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

70. The combination or method of claim 69, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

71. The combination or method of claim 70, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

72. The combination or method of any one of claims 56 to 71, wherein the (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or salt thereof, is a crystalline form of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide.

73. A combination comprising (1) a GLP-1R agonist and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

74. A method of treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

The GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

75. A method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or alleviating the medical consequences of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

76. The method of claim 74, wherein the disease or condition is fatty liver.

77. The method of claim 74, wherein said disease or condition is nonalcoholic fatty liver disease.

78. The method of claim 74, wherein said disease or condition is non-alcoholic steatohepatitis.

79. The method of claim 74, wherein said disease or condition is non-alcoholic steatohepatitis with liver fibrosis.

80. The method of claim 74, wherein said disease or condition is nonalcoholic steatohepatitis with cirrhosis.

81. The method of claim 74, wherein said disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

82. The method of claim 74, wherein said disease or condition is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

83. The combination of claim 73 or the method of claims 74 to 82 wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

84. The combination or method of claim 83 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

85. The combination or method of claim 84, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

86. The combination of claim 73 or the method of claims 74 to 82, wherein said GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

87. The combination or method of claim 86, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

88. The combination or method of claim 87, wherein the pharmaceutically acceptable salt is a tris (hydroxymethyl) aminomethane salt.

89. The combination or method of any one of claims 56 to 71, wherein the 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or salt thereof is 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid.

Background

Non-alcoholic steatohepatitis (NASH) is a clinical and histological subset of non-alcoholic fatty liver disease (NAFLD, defined as the presence of > 5% hepatic steatosis) that is associated with increased mortality from all causes, cirrhosis and end-stage liver disease, increased cardiovascular mortality, and increased incidence of both liver-related and non-liver related cancers (Sanyal et al, Hepatology 2015; 61(4): 1392-. NAFLD is the most common cause of chronic liver disease in the western world. It is a hepatic manifestation of metabolic syndrome and is a series of liver disorders including steatosis, NASH, fibrosis, cirrhosis, end-stage liver disease and eventually hepatocellular carcinoma. NAFLD and NASH are considered major fatty liver diseases because they account for the greatest proportion of individuals with elevated liver lipids. The severity of NAFLD/NASH is based on the presence of lipids, inflammatory cell infiltration, hepatocyte vacuolization, and the degree of fibrosis. Although not all individuals with steatosis progress to NASH, this is true for a significant number of individuals. Currently, treatment options are limited to lifestyle changes, including exercise and diet (EASL-EASD-EASO Clinical Practice Guidelines, J.hepatol.2016; 64(6): 1388) 1402).

It has been hypothesized that changes in lipid metabolism contribute to the molecular pathogenesis of NAFLD and NASH. Steatosis is a necessary but not sufficient element of the pathogenesis of NASH (Day c. and James o., hepatology.1998; 27(6): 1463-6). In line with this, several studies have demonstrated that the severity of steatosis predicts the risk of concomitant steatohepatitis and the risk of developing cirrhosis (Sorensen et al, Lancet. 1984; 2(8397): 241-4; Walless I and Lentz J, Hepatology 1990; 12(5): 1106-10; Reeves H. et al, J.hepatol. 1996; 25(5): 677-83). Hepatic steatosis is the result of TG production/uptake in the liver and elimination/elimination imbalances (Cohen JC et al, science.2011; 332(6037): 1519-. It is hypothesized that reducing steatosis (the metabolic driver supporting the development of NAFLD/NASH) leads to improvements in subsequent hepatitis and fibrosis.

Savage et al, demonstrated that both ACC 1and ACC2 are involved in regulating fat oxidation in hepatocytes, while ACC1 (the major isoform in rat liver) is the only regulator of fatty acid synthesis. Furthermore, in their model, a combined reduction of both isoforms is required to significantly reduce hepatic malonyl-CoA levels, increase fed state fat oxidation, reduce lipid accumulation and improve insulin action in vivo. Therefore, inhibitors of hepatic ACC 1and ACC2 may be useful in the treatment of NAFLD and hepatic insulin resistance. See Savage, D.B. et al, "reverse of di-induced pathological and pathological insulin resistance by antisense oligonucleotide inhibitors of acyl-CoA carboxylases 1and 2" J.Clin. invest.2006; 116(3):817-24. See also Oh, W, et al, "Glucose and fat metabolism in adsorption tissue of acyl-CoA carboxylase 2 knock out mice," PNAS,102(5) 1384-.

acetyl-CoA carboxylase (ACC) plays a key role in regulating lipid metabolism. ACC catalyzes an essential and rate-limiting step in the de novo lipid production (DNL) process (Saggerson D, Annu. Rev. Nutr.2008; 28: 253-72). Furthermore, ACC also regulates the mitochondrial beta oxidation of fatty acids by the ectopic regulation of the enzyme carnitine palmitoyl transferase 1(CPT1) (Saggerson, 2008; Waite M. and Wakil SJ. J.biol.chem.1962; 237: 2750-2757). Emerging data also indicate that DNL inhibition by ACC inhibition can directly reduce inflammation by limiting the formation of inflammatory interleukin-17 (IL-17) by T cells secreting helper T cell 17 lineage (Th17 cells) and favoring the development of anti-inflammatory FoxP3(+) regulatory T (treg) cells (Berod L et al, nat. med. 2014; 20(11): 1327-33). Recently, ACC has also been shown to inhibit in vitro human primary hepatic stellate cell activation and to reduce liver fibrosis in a rat model (Ross et al, Abstract PS-132 Journal of Hepatology 2019 vol. page 70 e 86).

Inhibition of ACC activity is hypothesized to favor patients with NASH by at least three independent mechanisms. As summarized above, humans with NAFLD show a significant elevation of hepatic DNL and it is hypothesized that normalizing this increased flux by pharmacological hepatic ACC inhibition reduces steatosis. Consistent with this, ACC inhibitors have been shown to inhibit DNL. In addition, the effect of ACC inhibition on increased fatty acid oxidation may also contribute to decreased liver fat content. See Griffith DA et al, j.med.chem.2014; 57(24) 10512 and 10526; kim CW et al, Cell metab.2017; 26,394-406; stiede k. et al, hepatology.2017; 66(2) 324, 334; lawitz EJ et al, Clin Gastroenterol Heastol.2018 (https:// doi.org/10.1016/j.cgh.2018.04.042). In addition, DNL inhibition in T cell secreting IL-17 is expected to inhibit liver inflammation by limiting the formation of inflammatory Th17 cells (Berod et al, 2014), a pathway that may be important in the pathogenesis of NASH (Rau M et al, J.Immunol.2016; 196(1):97-105), and favoring the development of anti-inflammatory Treg cells. Furthermore, ACC inhibition reduces astrocyte activation and fibrosis (Ross et al, 2019).

4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is a selective ACC inhibitor and was prepared as the free acid in example 9 of U.S. patent No. 8,859,577, which is a U.S. national phase of international application No. PCT/IB2011/054119, the disclosure of which is hereby incorporated by reference in its entirety for all purposes. Crystalline forms of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, including the anhydrous mono tris (hydroxymethyl) aminomethane (tris) form (form 1) and the trihydrate of the mono tris (hydroxymethyl) aminomethane salt (form 2), are described in international PCT application No. PCT/IB2018/058966, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.

Triglycerides or Triacylglycerols (TG) represent the major energy storage form in mammals. TG is formed by sequential esterification of glycerol with three fatty acids of different chain length and saturation (Coleman, R.A. and Mashek, D.G. 2011.chem.Rev.111: 6359-6386). TG synthesized in the intestine or liver are encapsulated in chylomicrons or Very Low Density Lipoproteins (VLDL), respectively, and exported to peripheral tissues where they are hydrolyzed by lipoprotein lipase (LPL) into their constituent fatty acids and glycerol. The resulting unesterified fatty acids (NEFA) can be metabolized for further energy production or re-esterified and stored.

Under normal physiological conditions, energy-intensive TGs remain sequestered in various fat depots until their release is required, at which time they are hydrolyzed to glycerol and free fatty acids, which are then released into the bloodstream. This process is tightly regulated by the opposing effects of insulin and hormones (such as catecholamines), which promote the deposition and mobilization of TG stores under a variety of physiological conditions. In a postprandial setting, insulin acts to inhibit lipolysis, thereby limiting energy release in the form of NEFA and ensuring that dietary lipids are properly stored in the fat depot. However, in patients with type 2 diabetes, the ability of insulin to inhibit lipolysis is impaired and NEFA flux from adipocytes is inappropriately elevated. This in turn results in an increase in lipid delivery to tissues such as muscle and liver. Without energy demand, TG and other lipid metabolites such as Diacylglycerol (DAG) can accumulate and cause loss of insulin sensitivity (2010. Nat Med 16: 400-. Insulin resistance in muscle is characterized by reduced glucose uptake and glycogen storage, while loss of insulin signaling in the liver leads to deregulated glucose output and overproduction of TG-rich VLDL (marker of type 2 diabetes) (Choi, s.h. and Ginsberg, h.n. 2011.Trends Endocrinol. Metab.22: 353-. Elevated secretion of TG-rich VLDL (also known as VLDL1 particles) is considered to stimulate the production of small, dense low-density lipoproteins (sdLDL), a small fraction of pro-atherosclerotic LDL, which is associated with an elevated risk of coronary heart disease (St-Pierre, a.c. et al, 2005. arteroscler.Thromb.Vasc.biol.25: 553-.

In mammals, two diacylglycerol acyltransferase (DGAT) enzymes (DGAT1 and DGAT2) have been characterized. Although these enzymes catalyze the same enzyme reaction, their respective amino acid sequences are not related and they occupy different gene families. Anchored mice disrupted by the gene encoding DGAT1 are resistant to diet-induced obesity and have elevated energy expenditure and activity (Smith, s.j. et al, 2000.Nat Genet 25: 87-90). Dgat 1-/-mice display a deregulated release of chylomicrons after absorption and accumulation of lipids in intestinal cells (Buhman, K.K. et al, 2002.J.biol.chem.277: 25474-25479). The metabolically favourable phenotype observed in these mice suggests a drive by an intestinal loss of DGAT1 expression (Lee, B. et al, 2010.J. lipid Res.51: 1770-1780). Importantly, despite lactation defects in female Dgat 1-/-mice, these animals retain the ability to synthesize TG, suggesting the presence of additional DGAT enzymes. This observation and the second DGAT isolated from the fungus Mortierella pusilla (Mortierella rampinana) led to the identification and characterization of DGAT2 (Yen, C.L. et al, 2008. lipid Res.49: 2283-.

DGAT2 is highly expressed in liver and fat and, unlike DGAT1, exhibits acute substrate specificity for DAG, (Yen, c.l., 2008). Deletion of the DGAT2 gene in rodents results in defective intrauterine growth, severe lipemia, impaired skin barrier function and early postpartum death (Stone, S.J. et al, 2004.J.biol. chem.279: 11767-11776). Due to the loss of fatality caused by DGAT2, most of our understanding of the physiological role of DGAT2 stems from studies with antisense oligonucleotides (ASOs) in rodent models of metabolic disease. In this setting, inhibition of hepatic DGAT2 resulted in an improvement in plasma lipoprotein profile (lowering total cholesterol and TG) and a reduction in hepatic lipid burden, which was accompanied by improved insulin sensitivity and systemic glucose control (Liu, y. et al, 2008.biochim. biophysis. acta 1781: 97-104; Choi, c.s. et al, 2007.j.biol. chem.282: 22678-. Although the molecular mechanisms underlying the observations are not fully elucidated, it is clear that inhibition of DGAT2 leads to down-regulation of the expression of multiple genes encoding proteins involved in adipogenesis, including sterol regulatory module-binding protein 1c (SREBP1c) and stearoyl CoA desaturase 1(SCD1) (Choi, 2007; Yu, 2005). In parallel, induction of the oxidative pathway was demonstrated by increased gene expression, such as carnitine palmitoyl transferase 1(CPT1) (Choi, 2007). The net result of these changes is a reduction in hepatic DAG and TG lipid levels, which in turn leads to improved intrahepatic insulin responses. In addition, DGAT2 inhibition inhibits hepatic VLDL TG secretion and a decrease in circulating cholesterol levels. Finally, plasma apolipoprotein B (APOB) levels are suppressed, possibly due to a reduced TG supply for lipidation of newly synthesized APOB protein (Liu, 2008; Yu, 2005). The beneficial effects of DGAT2 inhibition on both glycemic control and plasma cholesterol profiles suggest that this target may be of value in the treatment of metabolic diseases (Choi, 2007). In addition, the observation that inhibition of DGAT2 activity results in reduced liver lipid accumulation suggests that inhibitors of this enzyme may have utility in the treatment of NASH.

(S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide [ including its solid crystalline forms (form 1 and form 2) ] is the DGAT2 inhibitor described in example 1 of U.S. Pat. No. 10,071,992, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.

Ketohexokinase (KHK) is the major enzyme in fructose metabolism and catalyzes the conversion of fructose to fructose-1-phosphate (F1P). KHK is expressed as two alternative mRNA splice variants (expressed as KHKa and KHKc) resulting from alternative splicing of the third exon. KHKc has a much greater affinity and capacity for fructose phosphorylation than KHKa as demonstrated by a much lower Km (Ishimoto, Lanaspa et al, PNAS 109, 4320-. Although KHKa is ubiquitously expressed, the expression of KHKc is highest in the liver, kidney and intestine (the major site of fructose metabolism in vivo) (Diggle CP et al, (2009) J Histochem Cytochem 57: 763-. In addition, an loss of function mutation in humans has been reported and is called primary diabetes mellitus (OMIM #229800), without any adverse effects, except that the sugar appears in the urine after ingestion of fructose.

A more serious condition involving fructose metabolism is hereditary fructose intolerance (HFI, OMIM #229600), which is caused by a defect in the enzyme aldolase B (GENE: ALDOB), an enzyme responsible for breaking down F1P and immediately downstream of the KHK step in the pathway (Bouteldja N. et al, J.Inhert.Metab.Dis.2010 Apr; 33(2): 105-12; Tolan, DR Hum Mutat.1995; 6(3): 210-8; http:// www.omim.org/entry/229600). This is a rare disease, of which 1 is estimated to be affected in 20,000 people, and mutations that result in F1P accumulation, ATP depletion, and increased uric acid, the combination causing hypoglycemia, hyperuricemia, and lactic acidosis, as well as other metabolic disorders. HFI impairs the body's ability to metabolize dietary fructose, resulting in acute symptoms such as vomiting, severe hypoglycemia, diarrhea, and abdominal distress, resulting in long-term growth deficits, liver and kidney damage, and possible death (Ali M et al j. med. gene.1998 May:35(5): 353-65). Patients usually suffer during the first years before diagnosis and the only treatment is to avoid fructose in the diet. This macronutrient is present in most food items and poses a challenge to this approach. In addition to physical symptoms, many patients experience emotional and social isolation as a result of their unusual diet, and continually strive to comply with strict dietary restrictions (HFI-INFO discussion board, http:// hfiinfo. proboards. com.2015 12 months and 14 days visit). Even when patients develop asymptomatic, some patients develop NAFLD and kidney disease that highlights the shortfall of self-imposed dietary restrictions as the only treatment option and a highly unmet medical need for this condition.

In hyperglycemic conditions, endogenous fructose production occurs through the polyol pathway, through which glucose is converted to fructose, with sorbitol as an intermediate. The activity of this pathway increases with hyperglycemia. In the study, the authors demonstrated that KHK knockout mice are protected from glucose-induced weight gain, insulin resistance and hepatic steatosis, suggesting that endogenously produced fructose may promote insulin resistance and hepatic steatosis in hyperglycemic conditions (Lanaspa, m.a. et al, Nature comm.4,2434, 2013). Therefore, inhibition of KHK is expected to be beneficial in many diseases involving one or both changes in endogenous or ingested fructose.

[ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid (including its free acid crystalline form) is a ketohexokinase inhibitor and is illustrated in example 4 of U.S. patent No. 9,809,579, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.

Currently, various pharmacological approaches are available for the treatment of hyperglycemia and subsequent type 2 Diabetes, also known as T2DM (Hampp, c. et al, Use of anti-diabetic Drugs in the u.s.,2003-2012, Diabetes Care 2014,37, 1367-1374). These can be divided into six main categories, each functioning through a different main mechanism: (A) insulin secretagogues, including sulfonylureas (e.g., glipizide, glimepiride, glyburide), glibenclamide, meglitinide), meglitinides (e.g., nateglinide, repaglinide), dipeptidyl peptidase IV (DPP-IV) inhibitors (e.g., sitagliptin, vildagliptin, alogliptin, dulagliptin, liragliptin, linagliptin, saxagliptin, and glucagon-1 receptor (GLP-1R) agonists (e.g., liraglutide, albiglutide), exenatide, lixigliptin, dulagliptin, and palmatin beta secretion by pancreatic cells) are enhanced. Sulfonylureas and meglitinides have limited efficacy and tolerability, cause weight gain and often induce hypoglycemia. DPP-IV inhibitors have limited efficacy. Commercially available GLP-1R agonists are peptides administered by subcutaneous injection. Liraglutide is additionally approved for the treatment of obesity. (B) Biguanides (e.g., metformin (metformin)) are considered to act primarily to reduce hepatic glucose production. Biguanides often cause gastrointestinal discomfort and lactic acidosis, further limiting their use. (C) Alpha glucosidase inhibitors, such as acarbose, reduce glucose absorption in the intestine. These agents often cause gastrointestinal discomfort. (D) Thiazolidinediones (e.g. pioglitazone (pioglitazone), rosiglitazone (rosiglitazone)) act at specific receptors (peroxisome proliferator-activated receptor γ) in liver, muscle and adipose tissue. They regulate lipid metabolism and subsequently enhance the response of the tissue to the action of insulin. Frequent use of these drugs may lead to weight gain and may induce edema and anemia. (E) Insulin alone or in combination with the above agents is used in more severe cases, and frequent use may also lead to weight gain and carry the risk of hypoglycemia. (F) Sodium-glucose associated transporter cotransporter 2(SGLT2) inhibitors (e.g., dapagliflozin (dapagliflozin), engagliflozin (empagliflozin), canagliflozin (canagliflozin), egagliflozin (ertugliflozin)) inhibit glucose reabsorption in the kidney and thereby lower glucose levels in the blood. This emerging class of drugs may be associated with ketoacidosis and urinary tract infections.

However, except GLP-1R agonists and SGLT2 inhibitors, the efficacy of drugs for T2DM is limited and the most important problem, beta cell function decline and associated obesity, is not addressed.

Obesity is a chronic disease that is very prevalent in modern society and is associated with a number of medical problems, including hypertension, hypercholesterolemia, and coronary heart disease. It is further highly correlated with T2DM and insulin resistance, the latter often accompanied by hyperinsulinemia or hyperglycemia, or both. In addition, T2DM is associated with a two to four fold increased risk of coronary artery disease. Currently, the only treatment that eliminates obesity with high efficacy is weight loss surgery, but this treatment is expensive and at high risk. Pharmacological interventions are generally less effective and associated with side effects. Thus, there is a clear need for more effective pharmacological interventions with fewer side effects and convenient administration.

Although T2DM is most commonly associated with hyperglycemia and insulin resistance, other diseases associated with T2DM include hepatic insulin resistance, glucose intolerance, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, obesity, dyslipidemia, hypertension, hyperinsulinemia, and nonalcoholic fatty liver disease (NAFLD).

NAFLD is a hepatic manifestation of metabolic syndrome and is a range of liver disorders including steatosis, nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis and eventually hepatocellular carcinoma. NAFLD and NASH are considered major fatty liver diseases because they account for the greatest proportion of individuals with elevated liver lipids. The severity of NAFLD/NASH is based on the presence of lipids, inflammatory cell infiltration, hepatocyte vacuolization, and the degree of fibrosis. Although not all individuals with steatosis progress to NASH, this is true of a considerable number of people.

GLP-1 is a 30 amino acid long incretin hormone that is secreted in the intestine by L cells in response to food intake. GLP-1 has been shown to stimulate insulin secretion, decrease glucagon secretion, inhibit gastric emptying, decrease appetite, and stimulate beta cell proliferation in a physiological and glucose-dependent manner. In non-clinical trials, GLP-1 promotes sustained beta cell capacity by stimulating transcription of genes important for glucose-dependent insulin secretion and by promoting beta cell neogenesis (Meier et al, Biodrugs.2003; 17(2): 93-102).

In healthy individuals, GLP-1 plays an important role in regulating postprandial blood glucose levels by stimulating glucose-dependent insulin secretion from the pancreas, leading to increased peripheral glucose absorption. GLP-1 also inhibits glucagon secretion, resulting in reduced hepatic glucose output. Additionally, GLP-1 delays gastric emptying and slows small bowel motility to delay food absorption. In humans with T2DM, the normal postprandial rise in GLP-1 was not seen or reduced (Vilsball T et al diabetes 2001.50; 609-.

Holst (physiol.rev.2007,87,1409) and Meier (nat.rev.endocrinol.2012,8,728) describe that GLP-1 receptor agonists (such as GLP-1, liraglutide and exendin (exendin) -4) have 3 major pharmacological activities that improve glycemic control in patients (such as those with T2 DM) by lowering fasting and postprandial glucose (FPG and PPG): (i) increased glucose-dependent insulin secretion (improved first and second phases), (ii) glucagon inhibitory activity under hyperglycemic conditions, (iii) delayed gastric emptying rate, resulting in a diet-derived glucose absorption block.

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically salt thereof [ such as its 2-amino-2- (hydroxymethyl) propan-1, 3-diolate, also known as its tris (hydroxymethyl) aminomethane salt (tris salt) ] is a GLP-1R agonist described in U.S. Pat. No. 10,208,019 (see example 4A-01 of that patent), the disclosure of which is hereby incorporated by reference in its entirety for all purposes.

Additional GLP-1R agonists useful for the present invention are described in U.S. provisional patent application No. 62/684,696, filed on 6/13/2018 and U.S. provisional patent application No. 62/846,944, filed on 5/13/2019, the disclosures of each of which are hereby incorporated by reference in their entirety for all purposes.

Farnesoid X Receptor (FXR) is a member of the nuclear hormone receptor superfamily and is mainly expressed in the liver, kidney and intestine (see, e.g., Seol et al, (1995) mol. Endocrinol.9:72-85 and Forman et al, (1995) Cell 81: 687-693). It functions as a heterodimer with Retinoid X Receptor (RXR) and binds to a responsive component in the promoter of the target gene to regulate gene transcription. The FXR-RXR heterodimer binds with highest affinity to the inverted repeat-1 (IR-1) reaction module, where hexamers binding to the shared receptor are separated by one nucleotide. FXR is part of a interrelated process, since FXR is activated as bile acid (the end product of cholesterol metabolism) (see, e.g., Makishima et al, (1999) Science 284: 1362-1365; Parks et al, (1999) Science 284: 1365-1368; Wang et al, (1999) mol. cell.3:543-553), which is useful for inhibiting cholesterol catabolism. See also Urizar et al, (2000) J.biol.chem.275: 39313-39317.

FXR is a key regulator of cholesterol homeostasis, triglyceride synthesis, and lipid production. (Expert Opinion patents (2010),20(8): 1047-. In addition to the treatment of dyslipidemia, various indications for FXR have been described, including the treatment of liver disease, diabetes, vitamin D related diseases, drug induced side effects and hepatitis. (Crawley, as before). Despite the advances made in the development of novel FXR agonists, there remains considerable room for improvement.

The FXR agonist tropifexol (Tropifexor), or a pharmaceutically acceptable salt thereof, is described in example 1-1B of U.S. patent No. 9,150,568, the disclosure of which is hereby incorporated by reference in its entirety for all purposes. The chemical name of tropisethoxy is 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid.

In view of the above, a combination comprising a GLP-1R agonist and an ACC1 and/or ACC2 inhibitor for the treatment of a disease or disorder (including NAFLD and NASH); a combination of a GLP-1R agonist and a DGAT2 inhibitor; a combination of a GLP-1R agonist and a KHK inhibitor; and/or combinations of a GLP-1R agonist and an FXR agonist (e.g., oral agents) remain in need. The specific combinations described in this application satisfy the existing needs.

Summary of The Invention

The present invention relates to a combination and a method for treating fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with liver cirrhosis, and non-alcoholic steatohepatitis with liver cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases. In part, the present invention provides novel combinations of a GLP-1R agonist comprised in a pharmaceutical composition and an ACC inhibitor comprised in the same or separate compositions. The invention also provides novel combinations of a GLP-1R agonist included in a pharmaceutical composition and a DGAT2 inhibitor included in the same or separate compositions. The invention also provides novel combinations comprising a GLP-1R agonist in a pharmaceutical composition and a KHK inhibitor in the same or separate compositions. The invention also provides novel combinations of a GLP-1R agonist and an FXR agonist in the same or separate compositions comprised in a pharmaceutical composition. The invention further provides novel methods of treating fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, e.g., using the novel combinations described herein.

In one embodiment, the present invention provides a combination comprising a therapeutically effective amount of a composition comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1 to 103 or a pharmaceutically acceptable salt thereof) and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof in the same or separate compositions.

One embodiment of the present invention (embodiment a) provides a combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof.

One embodiment of the present invention (embodiment B) provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

the GLP-1R agonist is selected from:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

Or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading score system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprises administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof.

One embodiment of the invention (embodiment C) provides a method of reducing the severity of non-alcoholic fatty liver disease or non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidin ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1 to 103 or a pharmaceutically acceptable salt thereof) in a pharmaceutical composition and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, in the same or separate compositions.

One embodiment of the present invention (embodiment D) provides a combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof.

One embodiment of the present invention (embodiment E) provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

The GLP-1R agonist is selected from:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading score system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof A salt is accepted.

One embodiment of the invention (embodiment F) provides a method of reducing the severity of non-alcoholic fatty liver disease or non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a combination comprising a therapeutically effective amount of (1) a GLP-1R agonist (e.g., one of the compounds of examples 1 to 103 or a pharmaceutically acceptable salt thereof) in a pharmaceutical composition and (2) in the same or separate compositions (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide or a pharmaceutically acceptable salt thereof.

One embodiment of the present invention (embodiment G) provides a combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1 to 103 or a pharmaceutically acceptable salt thereof) and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide or a pharmaceutically acceptable salt thereof.

One embodiment of the invention (embodiment H) provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein:

said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

The GLP-1R agonist is selected from:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading score system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprises administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide or a pharmaceutically acceptable salt thereof.

One embodiment of the invention (embodiment I) provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprises administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2) (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a combination comprising a therapeutically effective amount of (1) a GLP-1R agonist (e.g., one of the compounds of examples 1 to 103 or a pharmaceutically acceptable salt thereof) in a pharmaceutical composition and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof, in the same or separate compositions.

One embodiment of the invention (embodiment J) provides a combination comprising (1) a GLP-1R agonist and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2) (2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

One embodiment of the invention (embodiment K) provides a method for treating a disease or disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof, wherein:

Said disease or condition is selected from fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, and non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases, obesity, and type 2 diabetes; and

the GLP-1R agonist is selected from:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

In one embodiment, the invention provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading score system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist (e.g., one of the compounds of examples 1-103 or a pharmaceutically acceptable salt thereof) and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4- Fluoro-1, 3-benzothiazole-6-carboxylic acid or its pharmaceutically acceptable salts.

One embodiment of the invention (embodiment L) provides a method of reducing the severity of non-alcoholic fatty liver disease or a non-alcoholic steatohepatitis grading scoring system by at least one point, reducing the serum marker level of non-alcoholic steatohepatitis activity, reducing non-alcoholic steatohepatitis disease activity, or reducing the medical outcome of non-alcoholic steatohepatitis in a patient, the method comprising administering to the patient a therapeutically effective amount of a combination comprising (1) a GLP-1R agonist and (2)2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid, or a pharmaceutically acceptable salt thereof, wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2; and

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid,

or a pharmaceutically acceptable salt thereof.

Any combination of the invention described herein may optionally further comprise at least one additional agent.

Any of the methods of use/treatment of the invention described herein may optionally further comprise administering at least one additional agent.

In some embodiments, the at least one additional agent (in the combinations of the invention, including both the pharmaceutical compositions of the invention and the methods of use of the invention) is selected from the group consisting of: acetyl-CoA carboxylase- (ACC) inhibitors, diacylglycerol O-acyltransferase 1(DGAT-1) inhibitors, monoacylglycerol O-acyltransferase inhibitors, Phosphodiesterase (PDE) -10 inhibitors, AMPK activators, sulfonylureas, meglitinides, alpha-amylase inhibitors, alpha-glucosidase hydrolase inhibitors, alpha-glucosidase inhibitors, PPAR γ agonists, PPAR α/γ agonists, biguanides, glucagon-like peptide 1(GLP-1) modulators, liraglutide, albiglutide, exenatide, albiglutide, lixivide, dolaglutide, somagluteline, protein tyrosine phosphatase-1B (PTP-1B) inhibitors, SIRT-1 activators, dipeptidyl peptidase IV (DPP-IV) inhibitors, dipeptidyl glycerol O-acyltransferase inhibitors, dihydroglyceryl glycerol O-acyltransferase inhibitors, Phosphodiesterase (PDE) -10 inhibitors, AMPK activators, dihydrogliobulingitide, dihydrogliptin-1B, and dipeptidyl peptidase IV (DPP-IV) inhibitors, Insulin secretagogues, fatty acid oxidation inhibitors, A2 antagonists, c-jun amino terminal kinase (JNK) inhibitors, glucokinase activators (GKa), insulin mimetics, glycogen phosphorylase inhibitors, VPAC2 receptor agonists, SGLT2 inhibitors, glucagon receptor modulators, GPR119 modulators, FGF21 derivatives or analogs, TGR5 receptor modulators, GPBAR1 receptor modulators, GPR40 agonists, GPR120 modulators, high affinity nicotinic acid receptor (HM74A) activators, SGLT1 inhibitors, carnitine palmitoyl transferase inhibitors or modulators, fructose 1, 6-bisphosphatase inhibitors, aldose reductase inhibitors, mineralocorticoid receptor inhibitors, TORC2 inhibitors, 2 and/or 5 inhibitors, PKC isoforms (e.g., CCR α, PKC β, γ PKC synthetase inhibitors, lipodystrophy inhibitors, serine palmitoyl transferase inhibitors, insulin mimetics, glycogen phosphorylase inhibitors, VPR 2 receptor agonists, SGLT2 inhibitors, glucagon receptor modulators, GPR1 modulators, and the like, Modulators of GPR81, modulators of GPR39, modulators of GPR43, modulators of GPR41, modulators of GPR105, modulators of kv1.3, modulators of retinol binding protein 4, modulators of the glucocorticoid receptor, modulators of the somatostatin receptor, inhibitors or modulators of PDHK2 or PDHK4, MAP4K4 inhibitors, modulators of the IL1 family including IL1 β, HMG-CoA reductase inhibitors, squalene synthetase inhibitors, fibrates, bile acid sequestrants, ACAT inhibitors, MTP inhibitors, lipoxygenase inhibitors, cholesterol absorption inhibitors, PCSK9 modulators, cholesteryl ester transfer protein inhibitors, and RXR α modulators.

In some embodiments, the at least one additional agent (in the combinations of the invention, including both the pharmaceutical compositions of the invention and the methods of use of the invention) is selected from the group consisting of: cysteamine or a pharmaceutically acceptable salt thereof, cystamine or a pharmaceutically acceptable salt thereof, an antioxidant compound, lecithin, a vitamin B complex, a bile salt formulation, a cannabinoid-1 (CB1) receptor antagonist, a cannabinoid-1 (CB1) receptor inverse agonist, a peroxisome proliferator activated receptor activity modulator, benzothiazepineOr benzothiepines, RNA antisense constructs that inhibit protein tyrosine phosphatase PTPRU, heteroatom-linked substituted piperidines and derivatives thereof, azacyclopentane derivatives capable of inhibiting stearoyl-coenzyme α δ -9 desaturase, acyl amide compounds having adiponectin secretagogue activity or inducer activity, quaternary ammonium compounds, Glatiramer (Glatiramer) acetate, pentraxin proteins, HMG-CoA reductase inhibitors, N-acetyl cysteine, isoflavone compounds, macrolide antibiotics, galectin inhibitors, antibodies, or any combination of the foregoing.

The present invention also includes:

any one of the combinations of the invention for use as a medicament;

any one of the combinations of the present invention for use in the treatment of fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, or non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases;

use of any one of the combinations of the invention for the treatment of fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, or non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases; and

use of any one of the combinations of the invention for the manufacture of a medicament for the treatment of fatty liver, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis with liver fibrosis, non-alcoholic steatohepatitis with cirrhosis, or non-alcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma or with metabolic-related diseases.

It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.

Brief description of the drawings

FIG. 1 is a graph showing the characteristic x-ray powder diffraction pattern (vertical axis: intensity (CPS); horizontal axis: 2 θ (degrees)) of form 1 of the compound of example DGAT2 i.

FIG. 2 is a graph showing the characteristic x-ray powder diffraction pattern (vertical axis: intensity (CPS); horizontal axis: 2 θ (degrees)) for form 2 of the compound of example DGAT2 i.

Figure 3 shows an illustrative PXRD pattern of form 1 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (the compound of example ACCi) performed on a Bruker AXS D4 energy diffractor equipped with a Cu radiation source.

Figure 4 shows an exemplary raman spectrum of form 1 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (example ACCi compound) collected using a Nicolet NXR FT raman accessory attached to an FT-IR bench.

FIG. 5 shows a sample at 500MHz set at Bruker-BioSpin Avance III (1H frequency) NMR Spectroscopy on a 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4' -piperidine) on a Bruker-BioSpin CPMAS Probe ]Illustrative of form 1 of (E) -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (example ACCI Compound)13C ssNMR pattern.

Figure 6 shows an illustrative PXRD pattern of form 2 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (the compound of example ACCi) performed on a Bruker AXS D4 energy diffractor equipped with a Cu radiation source.

Figure 7 shows an illustrative raman spectrum of form 2 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (example ACCi compound) collected using a Nicolet NXR FT-raman accessory attached to an FT-IR bench.

FIG. 8 shows the signal at 500MHz set at Bruker-BioSpin Avance III (1H frequency) NMR Spectroscopy on a 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4' -piperidine) on a Bruker-BioSpin CPMAS Probe]Illustrative of form 2 of (E) -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (example ACCI Compound)13C ssNMR pattern.

Figure 9 shows an illustrative single crystal structure of form 2 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (the compound of example ACCi).

Figure 10 summarizes the effect of orally administering 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidin ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on plasma triglyceride levels in Sprague Dawley rats fed on a western diet measured as a monotherapy and in combination in the fed state.

Figure 11 summarizes the effect of orally administering 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on plasma triglyceride levels in Sprague Dawley rats fed on a western diet measured as a monotherapy and in combination in the fasted state.

Figure 12 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on SREBP-1 nuclear localization as monotherapy and in combination in rats fed on a western diet.

Figure 13 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic adipogenic gene expression, particularly acetyl-CoA carboxylase (ACC1), as monotherapy and in combination administration in rats fed on a western diet.

Figure 14 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic lipogenic gene expression, particularly Fatty Acid Synthase (FASN), as monotherapy and in combination administration in rats fed on a western diet.

Figure 15 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic adipogenic gene expression, particularly sterol-CoA desaturase (SCD1), as monotherapy and in combination in rats fed on a western diet.

Figure 16 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic adipogenesis gene expression, particularly sterol regulatory module-binding protein 1c (SREBP-1c), as monotherapy and in combination administration in rats fed on a western diet.

Figure 17 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on liver adipogenesis gene expression, particularly proprotein convertase subtilisin/kexin type 9 (PCSK9), as monotherapy and in combination administration in rats fed on a western diet.

Figure 18 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic triglyceride levels as monotherapy and in combination oral administration in Sprague Dawley rats fed on a western diet.

Figure 19 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic elasticity (a marker of liver inflammation and fibrosis) as monotherapy and in combination oral administration in male Wistar Hann rats fed a choline deficient and high fat diet (CDAHFD).

Figure 20 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic alpha smooth actin (alpha SMA) immunohistochemistry (a marker of myofibroblast activation and fibrogenesis) as monotherapy and in combination with oral administration in male Wistar Hann rats fed CDAHFD.

Figure 21 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic stellar red staining as monotherapy and in combination oral administration in male Wistar Hann rats fed CDAHFD.

Figure 22 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic alpha smooth actin (alpha SMA) gene expression as monotherapy and in combination oral administration in male Wistar Hann rats fed CDAHFD.

Figure 23 summarizes the effect of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidin ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid and (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide on hepatic collagen 1a1 gene expression as monotherapy and in combination administered orally in male Wistar Hann rats fed CDAHFD.

Fig. 24 shows a powder X-ray diffraction pattern observed for the anhydrous (anhydrate) crystalline form (form 1) of the tris (hydroxymethyl) aminomethane salt of compound example 7.

Fig. 25 shows a powder X-ray diffraction pattern observed for the anhydrous (anhydrate) crystalline form (form a) of tris (hydroxymethyl) aminomethane salt of compound example 10.

Detailed description of the invention

The present invention may be understood more readily by reference to the following detailed description of exemplary embodiments of the invention and the examples included therein.

It is to be understood that the invention is not limited to the particular synthetic method of construction, which may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. In this specification and in the claims which follow, reference will be made to a number of terms which shall be defined to have the following meanings:

the use of "a" or "an" in this specification can mean one or more. As used in the claims of this application, the words "a" or "an" when used in conjunction with the word "comprising" may mean one or more than one. "another," as used herein, may mean at least a second or more.

The term "about" refers to relative terms indicating an approximation of plus or minus 10%, in one embodiment plus or minus 5%, and in another embodiment plus or minus 2% of the nominal value to which it refers. This approximate level is appropriate in the field of the invention unless a narrower range is specifically stated to be required for the value.

When used herein, a "compound" includes any pharmaceutically acceptable derivative or modification, including conformational isomers (e.g., cis and trans isomers) and all optical isomers (e.g., enantiomers and diastereomers), racemic, diastereomeric and other mixtures of such isomers, and solvates, hydrates, isomorphous (isomorphs), polymorphs, tautomers, esters, salt forms and prodrugs. The expression "prodrug" refers to a compound that is a drug precursor that releases drug in vivo via some chemical or physiological process after administration (e.g., a prodrug is converted to the desired drug form upon reaching physiological pH or by enzymatic action). Exemplary prodrugs release the corresponding free acid upon cleavage, and such hydrolyzable ester-forming residues of the compounds of the present invention include, but are not limited to, residues having a carboxyl moiety wherein the free hydrogen is replaced by: (C)1-C4) Alkyl, (C)2-C7) Alkanoyloxymethyl, 1- (alkanoyloxy) ethyl having 4 to 9 carbon atoms, 1-methyl-1- (alkanoyloxy) -ethyl having 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having 3 to 6 carbon atoms, 4 to 6 carbon atoms 1- (alkoxycarbonyloxy) ethyl having 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy) ethyl having 5 to 8 carbon atoms, N- (alkoxycarbonyl) aminomethyl having 3 to 9 carbon atoms, 1- (N- (alkoxycarbonyl) amino) ethyl having 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactone (crotonolactonyl), gamma-butyrolactone-4-yl, di-N, N- (C)1-C2) Alkylamino radical (C)2-C3) Alkyl (such as beta-dimethylaminoethyl), carbamoyl- (C)1-C2) Alkyl, N-di (C)1-C2) Alkylcarbamoyl- (C)1-C2) Alkyl and piperidinyl-, pyrrolidinyl-or N-morpholinyl (C)2-C3) An alkyl group.

Arrow as used in this applicationOr wavy linesRepresents the point of attachment of a substituent to another group.

By "patient" is meant a warm-blooded animal, such as, for example, guinea pig, mouse, rat, gerbil, cat, rabbit, dog, cow, goat, sheep, horse, monkey, chimpanzee, and human. A "mammal" is a patient.

By "pharmaceutically acceptable" is meant that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith.

As used herein, the following terms have the general meaning used for administering pharmaceutical agents: QD means once per day and BID means twice per day.

As used herein, the expressions "reaction-inert solvent" and "inert solvent" refer to a solvent or mixture thereof that does not interact with starting materials, reagents, intermediates or products in a manner that adversely affects the yield of the desired product.

As used herein, the term "selective" or "selected" refers to a compound that has a greater effect in a first assay than the same compound in a second assay. For example, in a "gut-selective" compound, a first assay is used for the half-life of the compound in the gut and a second assay is used for the half-life of the compound in the liver.

By "therapeutically effective amount" is meant the amount of all pharmacologically active agents in the combination therapy described herein for the treatment of a particular disease, condition, or disorder described herein.

The terms "treating", "treatment" or "treatment" as used in the present application include prophylactic, that is, prophylactic; palliative treatment, i.e., reducing, moderating, or slowing the progression of the disease (or disorder) or any tissue damage associated with the disease (or disorder) in the patient; and reversal, in which the patient's disease (or disorder) is not only alleviated, but any tissue damage associated with the disease (or disorder) is in a better state than when treatment was initiated. This latter may occur, for example, but not limited to, any one or more of the following: evidence of NASH regression and/or improvement in fibrosis score based on liver biopsy; a lower incidence of progression to cirrhosis, hepatocellular carcinoma, and/or other liver-related outcomes; a reduction or improvement in serum-or image-based marker levels of non-alcoholic steatohepatitis activity; reduction or improvement in nonalcoholic steatohepatitis disease activity; or a reduction in the medical consequences of non-alcoholic steatohepatitis.

It appears that administration of ACC inhibitors may have a positive effect on reducing hepatic TG and may have other benefits for the treatment of NASH. It is reported that an increase in circulating TG levels is a mechanistic consequence of hepatic ACC inhibition (Kim et al, 2017), although doses of ACC inhibitors that only partially inhibit DNL may not cause an increase in circulating TG (Bergman et al, (2018) j.of Hepatology, Volume 68, S582). It has been found that administration of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, optionally administered as a pharmaceutically acceptable salt, has the potential to result in elevated circulating TG (typically measured from plasma) in western diet-fed Sprague Dawley rats, as observed in human individuals.

The compounds of the present invention may contain asymmetric or chiral centers and thus exist in different stereoisomeric forms. Unless otherwise indicated, all stereoisomeric forms of the compounds of the present invention, as well as mixtures thereof (including racemic mixtures), are intended to form part of the present invention. In addition, the present invention encompasses all geometric and positional isomers. For example, if the compounds of the present invention introduce double bonds or fused rings, both cis and trans forms as well as mixtures are included within the scope of the present invention.

The chiral compounds of the invention (and chiral precursors thereof) can be obtained in enantiomerically enriched form on a resin using chromatography, typically High Pressure Liquid Chromatography (HPLC) or Supercritical Fluid Chromatography (SFC), with an asymmetric stationary phase and a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing 0 to 50% isopropanol, typically 2% to 20%, and 0 to 5% alkylamine, typically 0.1% Diethylamine (DEA) or isopropylamine. Concentration of the eluent provides an enriched mixture.

Mixtures of diastereomers may be separated into their individual diastereomers on the basis of their physicochemical differences by methods well known to those skilled in the art, such as chromatography and/or fractional crystallization. Enantiomers can be separated by: the enantiomeric mixture is converted into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., a chiral auxiliary, such as a chiral alcohol or a Mosher acid chloride), the diastereomers are separated and the monomeric diastereomers are converted (e.g., hydrolyzed) into the corresponding pure enantiomers. Enantiomers can also be separated using a chiral HPLC column. Alternatively, a particular stereoisomer may be synthesized by: asymmetric synthesis using optically active starting materials, using optically active reagents, substrates, catalysts or solvents, or the conversion of one stereoisomer to another by asymmetric transformations.

Where a compound of the invention has two or more stereocenters and absolute or relative stereochemistry is given in the name, the designations R and S refer to each stereocenter, respectively, as numbered in ascending order (1, 2, 3, etc.) according to the conventional IUPAC numbering scheme for each molecule. Where a compound of the invention has one or more stereogenic centers and stereochemistry is not given in the name or structure, it is to be understood that the name or structure is intended to encompass all forms of the compound, including the racemate form.

It is also possible for the intermediates and compounds of the invention to exist in different tautomeric forms and all such forms are included within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via proton migration, such as keto-enol and imine-enamine isomerizations.

Valence tautomers include interconversion with recombination of some of the bonding electrons.

Included within the scope of the claimed compounds are all stereoisomers, geometric isomers and tautomeric forms of the compounds of the invention, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts in which the counter ion is optically active, such as D-lactate or L-lysine; or racemates, for example DL-tartrate or DL-arginine.

The present invention includes all pharmaceutically acceptable isotopically-labeled compounds of the present invention in which one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.

Examples of isotopes suitable for inclusion in compounds of the invention include the following isotopes: hydrogen (such as2H and3H) carbon (such as11C、13C and14C) chlorine (such as36Cl), fluorine (such as18F) Iodine (such as123I、124I and125I) nitrogen (such as13N and15n), oxygen (such as15O、17O and18o), phosphorus (such as32P) and sulfur (such as35S)。

Certain isotopically-labeled compounds of the present invention (e.g., those into which a radioisotope is introduced) are useful in drug and/or substrate tissue distribution studies. Radioisotope tritium (i.e., tritium3H) And carbon-14 (i.e.14C) Are particularly useful for this purpose in view of their ease of introduction and ready availability for detection.

With heavier isotopes such as deuterium (i.e. deuterium2H) Substitution may result in particular therapeutic advantages due to greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements, and may therefore be preferred in some circumstances.

With positron-emitting isotopes (such as 11C、18F、15O and13n) substitution can be used in Positron Emission Tomography (PET) studies to examine substrate receptor occupancy.

Isotopically-labeled compounds of the present invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and preparations using appropriate isotopically-labeled reagents in place of the unlabeled reagents previously used.

The compounds of the invention may be isolated and used as such, or, where possible, in the form of their pharmaceutically acceptable salts. The term "salt" refers to both inorganic and organic salts of the compounds of the present invention. The salts may be prepared in situ during the final isolation and purification of the compounds or by separately treating the compounds with a suitable organic or inorganic acid or base and isolating the salt thus formed. The acids used to prepare the pharmaceutically acceptable acid addition salts of the foregoing base compounds of the present invention are those which form non-toxic acid addition salts (i.e., salts containing pharmacologically acceptable anions such as hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, naphthenate, methanesulfonate, glucoheptonate, lactobionate, laurylsulfonate, hexafluorophosphate, benzenesulfonate, toluenesulfonate, formate, trifluoroacetate, oxalate, benzenesulfonate, palmitate, pamoate, malonate, stearate, citrate, tartrate, hydrochloride, fumarate, tartrate, hydrochloride, or hydrochloride, Laurate, malate, borate, p-toluenesulfonate, and pamoate (i.e., 1' -methylene-bis- (2-hydroxy-3-naphthoate)).

The invention also relates to base addition salts of the compounds of the invention. Chemical bases that can be used as reagents to prepare pharmaceutically acceptable base salts of those compounds of the invention that are acidic in nature are those that form non-toxic base salts with such compounds. Such non-toxic base salts include, but are not limited to, those derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., lithium, potassium, and sodium) and alkaline earth metal cations (e.g., calcium and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine (meglumine), tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and lower alkanolammonium, and other base salts of pharmaceutically acceptable organic amines. See, e.g., Berge et al, J.pharm.Sci.66,1-19 (1977).

A particular compound of the invention may exist in more than one crystal form (commonly referred to as a "polymorph"). Polymorphs can be prepared by crystallization under various conditions, e.g., recrystallization using different solvents or different solvent mixtures; crystallization at different temperatures; and/or various cooling patterns that change from very fast to very slow cooling during crystallization. Polymorphs may also be obtained by heating or melting the compounds of the present invention followed by gradual or rapid cooling. The presence of polymorphs can be determined by solid probe NMR spectroscopy, IR spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques.

In a further embodiment, the composition further comprises at least one additional agent selected from the group consisting of: anti-inflammatory agents, antidiabetics, anti-fibrotic agents, anti-steatosis agents and cholesterol/lipid modulating agents.

In another embodiment, a method for treating a condition selected from the group consisting of: hyperlipidemia, type I diabetes, type II diabetes, idiopathic type I diabetes (type Ib), latent autoimmune diabetes in adults (LADA), early-onset type 2 diabetes (EOD), juvenile-onset atypical diabetes (YOAD), adult-onset diabetes in young adults (MODY), diabetes associated with malnutrition, gestational diabetes, coronary heart disease, ischemic stroke, vascular restenosis following angioplasty, peripheral vascular disease, intermittent claudication, myocardial infarction (e.g., necrosis and apoptosis), dyslipidemia, postprandial dyslipidemia, conditions of glucose Intolerance (IGT), conditions of fasting glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, Cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transient cerebral ischemia, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, impaired glucose metabolism, glucose intolerance conditions, impaired fasting glucose conditions, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcers and ulcerative colitis, endothelial dysfunction and vascular compliance impairment, hyperlipoproteinemia B, Alzheimer's disease, schizophrenia, cognitive impairment, inflammatory bowel disease, ulcerative colitis, Crohn's disease and irritable bowel syndrome, Nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD).

In a further embodiment, a method for treating a metabolic or metabolic-related disease, condition, or disorder comprises the step of administering to a patient in need of such treatment a combination comprising at least two separate pharmaceutical compositions:

(i) a first composition comprising (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, present in a therapeutically effective amount in admixture with at least one pharmaceutically acceptable excipient;

(ii) a second composition comprising 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof present in a therapeutically effective amount in admixture with at least one pharmaceutically acceptable excipient; and optionally

(iii) A third composition comprising at least one additional agent selected from the group consisting of: anti-inflammatory agents, antidiabetics, anti-fibrotic agents, anti-steatosis agents and cholesterol/lipid modulating agents and antidiabetics, and at least one pharmaceutically acceptable excipient.

In yet a further embodiment, the method of the invention is carried out when the first composition, the second composition and the third composition are administered simultaneously.

In yet another embodiment, the method of the present invention is carried out when the first composition, the second composition and the third composition are administered separately or sequentially and in any order.

In one embodiment, when three compositions are administered, the first composition is administered simultaneously with the second composition, while the third composition is administered sequentially. In another embodiment, the three separate compositions are administered sequentially and in any order.

The compounds of the invention can be synthesized by synthetic routes, which include methods analogous to those well known in the chemical arts, particularly in accordance with the teachings contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.), or are readily prepared using methods well known to those skilled in the art (e.g., prepared in the methods generally described in Reagents for Organic Synthesis, v.1-19, Wiley, New York (1967. 1999 ed.) or Beilsteins Handbuch der organischen Chemie,4, autofl. ed. Springer-Verlag, Berlin, including the appendix (also available via the Beilstein on-line database)). The preparation of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is provided in example 1 of US 2018-. Preparation of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid example 9 of US 8,859,577, which is hereby incorporated by reference in its entirety for all purposes.

GLP-1R agonist/ACCI combinations

Embodiments A, B and C relate to a combination of a GLP-1R agonist (including GLP-1R agonists described herein) and the ACC inhibitor 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid or a pharmaceutically acceptable salt thereof.

Embodiment a1 is a further embodiment of embodiment a wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment a2 is a further embodiment of embodiment a or a1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment A3 is a further embodiment of embodiment a2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment A4 is a further embodiment of embodiment A wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

Or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment A5 is a further embodiment of embodiment A4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment a6 is a further embodiment of embodiment a5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment a7 is a further embodiment of any one of embodiments a1 to a5, wherein 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, is 2-amino-2- (hydroxymethyl) propan-1 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, 3-dialkoxide.

Embodiment A8 is a further embodiment of embodiment a7 wherein the 2-amino-2- (hydroxymethyl) propane-1, 3-diol salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is in crystalline form.

Embodiment a9 is a further embodiment of embodiment A8 wherein the ratio of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid to 2-amino-2- (hydroxymethyl) propane-1, 3-diol in the crystalline form is 1: 1.

Embodiment a10 is a further embodiment of embodiment A8 or a9 wherein the crystalline form is an anhydrous crystalline form.

Embodiment a11 is a further embodiment of embodiment a10 wherein the crystalline anhydrous form has a PXRD pattern comprising peaks at diffraction angles of 9.6, 10.7, and 11.32 theta, ± 0.2 ° 2 theta.

Embodiment a12 is a further embodiment of embodiment a10 or a11 wherein the anhydrous crystalline form has a crystal size comprised between 1511, 1561 and 1615cm-1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment a13 is a further embodiment of any one of embodiments a10 to a12, wherein the anhydrous crystalline form has chemical shifts comprised at 22.9, 146.2, and 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment a14 is a further embodiment of embodiment a13, wherein the crystalline anhydrate form has analytical parameters selected from the group consisting of: included at 1511 and 1615cm-1,±2cm-1And at least one chemical shift comprised at 22.9, 146.2 or 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment a15 is a further embodiment of embodiment A8 or a9 wherein the crystalline form is a trihydrate crystal.

Embodiment a16 is a further embodiment of embodiment a15 wherein the trihydrate crystals have a PXRD pattern comprising peaks at diffraction angles of 8.4, 9.0, and 10.52 theta, ± 0.2 ° 2 theta.

Embodiment a17 is a further embodiment of embodiment a15 or a16 wherein the trihydrate crystals have a crystalline form comprised between 1507, 1557 and 1610cm -1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment a18 is a further embodiment of any one of embodiments a15 to a17, wherein the trihydrate crystals have chemical shifts comprised between 19.2, 149.5 and 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment a19 is a further embodiment of embodiment a18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of:

PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta,

contained in 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (A), and

comprising at least one chemical shift in the range of 19.2, 149.5 or 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment a20 is a further embodiment of embodiment a18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta and PXRD patterns containing peaks at 1507, 1557 or 1610cm-1,±2cm-1At least one peak shift of the raman spectrum.

Embodiment a21 is a further embodiment of embodiment a18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns comprising peaks at diffraction angles of 8.4 and 9.02 theta, + -0.2 DEG 2 theta and PXRD patterns comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm, + -0.2 ppm 13C ssNMR spectrum.

Embodiment a22 is a further embodiment of any one of embodiment a1 to a12, the composition further comprising at least one additional agent.

Embodiment a23 is a further embodiment of embodiment a22 wherein the at least one other agent is selected from the group consisting of: acetyl-CoA carboxylase- (ACC) inhibitors, diacylglycerol O-acyltransferase 1(DGAT-1) inhibitors, monoacylglycerol O-acyltransferase inhibitors, Phosphodiesterase (PDE) -10 inhibitors, AMPK activators, sulfonylureas, meglitinides, alpha-amylase inhibitors, alpha-glucosidase hydrolase inhibitors, alpha-glucosidase inhibitors, PPAR γ agonists, PPAR α/γ agonists, biguanides, glucagon-like peptide 1(GLP-1) modulators, liraglutide, albiglutide, exenatide, albiglutide, lixivide, dolaglutide, somagluteline, protein tyrosine phosphatase-1B (PTP-1B) inhibitors, SIRT-1 activators, dipeptidyl peptidase IV (DPP-IV) inhibitors, DPP-1 inhibitors, and methods of using the same, Insulin secretagogues, fatty acid oxidation inhibitors, A2 antagonists, c-jun amino terminal kinase (JNK) inhibitors, glucokinase activators (GKa), insulin mimetics, glycogen phosphorylase inhibitors, VPAC2 receptor agonists, SGLT2 inhibitors, glucagon receptor modulators, GPR119 modulators, FGF21 derivatives or analogs, TGR5 receptor modulators, GPBAR1 receptor modulators, GPR40 agonists, GPR120 modulators, high affinity nicotinic acid receptor (HM74A) activators, SGLT1 inhibitors, carnitine palmitoyl transferase inhibitors or modulators, fructose 1, 6-bisphosphatase inhibitors, aldose reductase inhibitors, mineralocorticoid receptor inhibitors, TORC2 inhibitors, 2 and/or 5 inhibitors, PKC isoforms (e.g., PKC α, PKC β, PKC γ) inhibitors, fatty acid palmitoyl transferase inhibitors, serine palmitoyl transferase inhibitors, A GPR81 modulator, a GPR39 modulator, a GPR43 modulator, a GPR41 modulator, a GPR105 modulator, a kv1.3 modulator, a retinol binding protein 4 modulator, a glucocorticoid receptor modulator, a somatostatin receptor modulator, a PDHK2 or PDHK4 inhibitor or modulator, a MAP4K4 inhibitor, a modulator of the IL1 family including IL1 β, an HMG-CoA reductase inhibitor, a squalene synthetase inhibitor, a fibrate, a bile acid sequestrant, an ACAT inhibitor, an MTP inhibitor, a lipoxygenase inhibitor, a cholesterol absorption inhibitor, a PCSK9 modulator, a cholesteryl ester transfer protein inhibitor, and a rxralpha modulator.

Embodiment B1 is a further embodiment of embodiment B, wherein the disease or disorder is fatty liver.

Embodiment B2 is a further embodiment of embodiment B, wherein the disease or condition is nonalcoholic fatty liver disease.

Embodiment B3 is a further embodiment of embodiment B, wherein the disease or disorder is non-alcoholic steatohepatitis.

Embodiment B4 is a further embodiment of embodiment B, wherein the disease or disorder is non-alcoholic steatohepatitis with liver fibrosis.

Embodiment B5 is a further embodiment of embodiment B, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis.

Embodiment B6 is a further embodiment of embodiment B wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

Embodiment B7 is a further embodiment of embodiment B wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

Embodiment B8 is a further embodiment of any one of embodiments B1 to B7 wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorobenzyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment B9 is a further embodiment of embodiment B8 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment B10 is a further embodiment of embodiment B9 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment B11 is a further embodiment of any one of embodiments B1 to B7 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment B12 is a further embodiment of embodiment B11 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment B13 is a further embodiment of embodiment B12 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment B14 is a further embodiment of any one of embodiments B1 to B13, wherein 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, is 2-amino-2- (hydroxymethyl) propan-1 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, 3-dialkoxide.

Embodiment B15 is a further embodiment of embodiment B14 wherein the 2-amino-2- (hydroxymethyl) propane-1, 3-diol salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is in crystalline form.

Embodiment B16 is a further embodiment of embodiment B15 wherein the ratio of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid to 2-amino-2- (hydroxymethyl) propane-1, 3-diol in the crystalline form is 1: 1.

Embodiment B17 is a further embodiment of embodiment B15 or B16 wherein the crystalline form is an anhydrous crystalline form.

Embodiment B18 is a further embodiment of embodiment B17 wherein the crystalline anhydrous form has a PXRD pattern comprising peaks at diffraction angles of 9.6, 10.7, and 11.32 theta, ± 0.2 ° 2 theta.

Embodiment B19 is a further embodiment of embodiment B17 or B18 wherein the anhydrous crystalline form has a crystal size comprised between 1511, 1561 and 1615cm-1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment B20 is an embodiment B17 to B19A further embodiment of any one of the above, wherein the anhydrous crystalline form has chemical shifts comprised at 22.9, 146.2, and 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment B21 is a further embodiment of embodiment B20, wherein the crystalline anhydrate form has analytical parameters selected from the group consisting of: included at 1511 and 1615cm-1,±2cm-1And at least one chemical shift comprised at 22.9, 146.2 or 161.9ppm, ± 0.2ppm 13C ssNMR spectrum.

Embodiment B22 is a further embodiment of embodiment B15 or B16 wherein the crystalline form is a trihydrate crystal.

Embodiment B23 is a further embodiment of embodiment B22 wherein the trihydrate crystals have a PXRD pattern comprising peaks at diffraction angles of 8.4, 9.0, and 10.52 theta, ± 0.2 ° 2 theta.

Embodiment B24 is a further embodiment of embodiment B22 or B23 wherein the trihydrate crystals have a crystalline form comprised between 1507, 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment B25 is a further embodiment of any one of embodiments B22 to B24, wherein the trihydrate crystals have chemical shifts comprised between 19.2, 149.5 and 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment B26 is a further embodiment of embodiment B25 wherein the trihydrate crystals have analytical parameters selected from the group consisting of:

PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta,

contained in 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (A), and

comprising at least one chemical shift in the range of 19.2, 149.5 or 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment B27 is a further embodiment of embodiment B25 wherein the trihydrate crystals have a component selected from the group consisting of Analyzing parameters: PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta and PXRD patterns containing peaks at 1507, 1557 or 1610cm-1,±2cm-1At least one peak shift of the raman spectrum.

Embodiment B28 is a further embodiment of embodiment B25 wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns comprising peaks at diffraction angles of 8.4 and 9.02 theta, + -0.2 DEG 2 theta and PXRD patterns comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm, + -0.2 ppm13C ssNMR spectrum.

Embodiment B29 is a further embodiment of any one of embodiment B or embodiments B1 to B28, the method further comprising administering at least one additional agent.

Embodiment C1 is a further embodiment of embodiment C wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment C2 is a further embodiment of embodiment C1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment C3 is a further embodiment of embodiment C2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment C4 is a further embodiment of embodiment C wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment C5 is a further embodiment of embodiment C4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

Or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment C6 is a further embodiment of embodiment C5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment C7 is a further embodiment of any one of embodiment C1 to B6, wherein 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, or a pharmaceutically acceptable salt thereof, is 2-amino-2- (hydroxymethyl) propan-1 of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, 3-dialkoxide.

Embodiment C8 is a further embodiment of embodiment C7 wherein the 2-amino-2- (hydroxymethyl) propane-1, 3-diol salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is in crystalline form.

Embodiment C9 is a further embodiment of embodiment C8 wherein the ratio of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid to 2-amino-2- (hydroxymethyl) propane-1, 3-diol in the crystalline form is 1: 1.

Embodiment C10 is a further embodiment of embodiment C8 or C9 wherein the crystalline form is an anhydrous crystalline form.

Embodiment C11 is a further embodiment of embodiment C10 wherein the crystalline anhydrous form has a PXRD pattern comprising peaks at diffraction angles of 9.6, 10.7, and 11.32 theta, ± 0.2 ° 2 theta.

Embodiment C12 is a further embodiment of embodiment C10 or C11 wherein the anhydrous crystalline form has a crystal size comprised between 1511, 1561 and 1615cm-1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment C13 is a further embodiment of any one of embodiments C10 to C12, wherein the anhydrous crystalline form has chemical shifts comprised at 22.9, 146.2, and 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment 14 is a further embodiment of embodiment C13 wherein the anhydrous crystalline form has analytical parameters selected from the group consisting of: included at 1511 and 1615cm-1,±2cm-1And at least one chemical shift comprised at 22.9, 146.2 or 161.9ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment C15 is a further embodiment of embodiment C8 or BC9 wherein the crystalline form is a trihydrate crystal.

Embodiment C16 is a further embodiment of embodiment C15 wherein the trihydrate crystals have a PXRD pattern comprising peaks at diffraction angles of 8.4, 9.0, and 10.52 theta, ± 0.2 ° 2 theta.

Embodiment C17 is a further embodiment of embodiment C15 or C16 wherein the trihydrate crystals have a crystalline form comprised between 1507, 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (a).

Embodiment C18 is a further embodiment of any one of embodiments C15 to C16, wherein the trihydrate crystals have chemical shifts comprised between 19.2, 149.5 and 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment C19 is a further embodiment of embodiment C18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of:

PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta,

contained in 1557 and 1610cm-1,±2cm-1Raman spectrum of the peak shift of (A), and

comprising at least one chemical shift in the range of 19.2, 149.5 or 163.8ppm, ± 0.2ppm13C ssNMR spectrum.

Embodiment C20 is a further embodiment of embodiment C18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns containing peaks at diffraction angles of 8.4 and 9.02 theta, ± 0.2 ° 2 theta and PXRD patterns containing peaks at 1507, 1557 or 1610cm-1,±2cm-1At least one peak shift of the raman spectrum.

Embodiment C21 is a further embodiment of embodiment C18 wherein the trihydrate crystals have analytical parameters selected from the group consisting of: PXRD patterns comprising peaks at diffraction angles of 8.4 and 9.02 theta, + -0.2 DEG 2 theta and PXRD patterns comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm, + -0.2 ppm 13C ssNMR spectrum.

Embodiment C22 is a further embodiment of embodiment C or any one of embodiments C1 to C21, the method further comprising administering at least one additional agent.

In each embodiment of the method of treatment of the present invention (including embodiment B, embodiments B1 to B29, embodiment C and embodiments C1 to C29), each GLP-1R agonist and ACC inhibitor may be present in the same composition or in separate compositions. The combined amount of GLP-1R agonist and ACC inhibitor is therapeutically effective for the methods described herein. Even when the GLP-1R agonist and the ACC inhibitor are present in separate compositions, they may be administered simultaneously or sequentially; and when they are administered sequentially, they may be administered in any order.

GLP-1R agonist/KHKi combinations

Embodiments D, E and F relate to a combination of a GLP-1R agonist (including GLP-1R agonists described herein) and a KHKi inhibitor [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof.

Embodiment D1 is a further embodiment of embodiment D wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment D2 is a further embodiment of embodiment D or D1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment D3 is a further embodiment of embodiment D2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment D4 is a further embodiment of embodiment D wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; 3

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment D5 is a further embodiment of embodiment D4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment D6 is a further embodiment of embodiment D5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment D7 is a further embodiment of embodiment D or any one of embodiments D1 to D6 wherein the [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof is [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid.

Embodiment D8 is a further embodiment of embodiment D7 wherein the [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid is in crystalline form.

Embodiment D9 is a further embodiment of embodiment D8 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 10.4, 15.0, and 21.42 theta, ± 0.2 ° 2 theta.

Embodiment D10 is a further embodiment of embodiment D8 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 15.019.6, 21.4, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment D11 is a further embodiment of embodiment D8 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 10.4, 11.5, 15.0, 19.6, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment D12 is a further embodiment of embodiment D or any one of embodiments D1 to D11 wherein the composition further comprises at least one additional agent.

Embodiment E1 is a further embodiment of embodiment E, wherein the disease or disorder is fatty liver.

Embodiment E2 is a further embodiment of embodiment E, wherein the disease or disorder is nonalcoholic fatty liver disease.

Embodiment E3 is a further embodiment of embodiment E, wherein the disease or disorder is non-alcoholic steatohepatitis.

Embodiment E4 is a further embodiment of embodiment E, wherein the disease or disorder is nonalcoholic steatohepatitis with liver fibrosis.

Embodiment E5 is a further embodiment of embodiment E, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis.

Embodiment E6 is a further embodiment of embodiment E wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

Embodiment E7 is a further embodiment of embodiment E, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

Embodiment E8 is a further embodiment of any one of embodiments E1 to E7, wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorobenzyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment E9 is a further embodiment of embodiment E8 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment E10 is a further embodiment of embodiment E9 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment E11 is a further embodiment of embodiment E or any one of embodiments E1 to E7 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment E12 is a further embodiment of embodiment E11 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment E13 is a further embodiment of embodiment E12 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment E14 is a further embodiment of embodiment E or any one of embodiments E1 to E13, wherein the [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof is [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid (free acid).

Embodiment E15 is a further embodiment of embodiment E14 wherein [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid is in crystalline form.

Embodiment E16 is a further embodiment of embodiment E15 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 10.4, 15.0, and 21.42 theta, ± 0.2 ° 2 theta.

Embodiment E17 is a further embodiment of embodiment E15 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 15.019.6, 21.4, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment E18 is a further embodiment of embodiment E15, wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 10.4, 11.5, 15.0, 19.6, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment E19 is a further embodiment of embodiment E or any one of embodiments E1 to E18, the method further comprising administering at least one additional agent.

Embodiment F1 is a further embodiment of embodiment F wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment F2 is a further embodiment of embodiment F1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment F3 is a further embodiment of embodiment F2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment F4 is a further embodiment of embodiment F wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment F5 is a further embodiment of embodiment F4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

Or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment F6 is a further embodiment of embodiment F5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment F7 is a further embodiment of embodiment F or any one of embodiments F1 to F6, wherein the [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid or a pharmaceutically acceptable salt thereof is [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid.

Embodiment F8 is a further embodiment of embodiment F7 wherein [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid is in crystalline form.

Embodiment F9 is a further embodiment of embodiment F8 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 10.4, 15.0, and 21.42 theta, ± 0.2 ° 2 theta.

Embodiment F10 is a further embodiment of embodiment F8 wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 9.0, 15.019.6, 21.4, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment F11 is a further embodiment of embodiment F8, wherein the crystalline form has a PXRD pattern comprising peaks at diffraction angles of 10.4, 11.5, 15.0, 19.6, and 26.52 theta, ± 0.2 ° 2 theta.

Embodiment F12 is a further embodiment of embodiment F or any one of embodiments F1 to F11 wherein the method further comprises administering at least one additional agent.

In each of the embodiments of the methods of treatment of the present invention (including embodiment E, embodiments E1 to E19, embodiment F and embodiments F1 to F12), each GLP-1R agonist and KHK inhibitor may be present in the same composition or in separate compositions. The combined amount of the GLP-1R agonist and the KHK inhibitor is therapeutically effective for the methods described herein. Even when the GLP-1R agonist and the KHK inhibitor are present in separate compositions, they may be administered simultaneously or sequentially; and when they are administered sequentially, they may be administered in any order.

GLP-1R agonist/DGAT 2 inhibitor combinations

Embodiments G, H and I relate to a combination of a GLP-1R agonist (including GLP-1R agonists described herein) and the DGAT2 inhibitor (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide or a pharmaceutically acceptable salt thereof.

Embodiment G1 is a further embodiment of embodiment G wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment G2 is a further embodiment of embodiment G or G1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment G3 is a further embodiment of embodiment G2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment G4 is a further embodiment of embodiment G wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

Or a pharmaceutically acceptable salt thereof.

Embodiment G5 is a further embodiment of embodiment G4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment G6 is a further embodiment of embodiment G5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment G7 is a further embodiment of embodiment G or any one of embodiments G1 to G6, wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, is (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide.

Embodiment G8 is a further embodiment of embodiment G7 wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is in crystalline form.

Embodiment G9 is a further embodiment of embodiment G8 wherein the crystalline form has a PXRD pattern (form 1) comprising peaks at diffraction angles of 5.3, 7.7, 15.42 theta, ± 0.2 ° 2 theta.

Embodiment G10 is a further embodiment of embodiment G8 wherein the crystalline form has a PXRD pattern (form 2) comprising peaks at diffraction angles of 6.5, 9.3, 13.62 Θ, ± 0.2 ° 2 Θ.

Embodiment G11 is a further embodiment of embodiment G or any one of embodiments G1 to G10, wherein the composition further comprises at least one additional agent.

Embodiment H1 is a further embodiment of embodiment H, wherein the disease or disorder is fatty liver.

Embodiment H2 is a further embodiment of embodiment H, wherein the disease or disorder is non-alcoholic fatty liver disease.

Embodiment H3 is a further embodiment of embodiment H, wherein the disease or disorder is non-alcoholic steatohepatitis.

Embodiment H4 is a further embodiment of embodiment H, wherein the disease or disorder is nonalcoholic steatohepatitis with liver fibrosis.

Embodiment H5 is a further embodiment of embodiment H, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis.

Embodiment H6 is a further embodiment of embodiment H wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

Embodiment H7 is a further embodiment of embodiment H, wherein the disease or disorder is non-alcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

Embodiment H8 is a further embodiment of embodiment H or any one of embodiments H1 to H7 wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorobenzyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment H9 is a further embodiment of embodiment H8 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment H10 is a further embodiment of embodiment H9 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment H11 is a further embodiment of embodiment H or any one of embodiments H1 to H7 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment H12 is a further embodiment of embodiment H11 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment H13 is a further embodiment of embodiment H12 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment H14 is a further embodiment of any one of embodiments H1 to H13, wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, is (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide.

Embodiment H15 is a further embodiment of embodiment H14 wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is in crystalline form.

Embodiment H16 is a further embodiment of embodiment H15 wherein the crystalline form has a PXRD pattern (form 1) comprising peaks at diffraction angles of 5.3, 7.7, 15.42 Θ, ± 0.2 ° 2 Θ.

Embodiment H17 is a further embodiment of embodiment H15 wherein the crystalline form has a PXRD pattern (form 2) comprising peaks at diffraction angles of 6.5, 9.3, 13.62 Θ, ± 0.2 ° 2 Θ.

Embodiment H18 is a further embodiment of any one of embodiments H1 to E17, the method further comprising administering at least one additional agent.

Embodiment I1 is a further embodiment of embodiment I wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment I2 is a further embodiment of embodiment I1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment I3 is a further embodiment of embodiment I2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment I4 is a further embodiment of embodiment I wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment I5 is a further embodiment of embodiment I4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

Or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment I6 is a further embodiment of embodiment I5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment I7 is a further embodiment of any one of embodiment I or embodiments I1 to I6, wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, is (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide.

Embodiment I8 is a further embodiment of embodiment I7 wherein (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is in crystalline form.

Embodiment I9 is a further embodiment of embodiment I8 wherein the crystalline form has a PXRD pattern (form 1) comprising peaks at diffraction angles of 5.3, 7.7, 15.42 theta, ± 0.2 ° 2 theta.

Embodiment I10 is a further embodiment of embodiment I8 wherein the crystalline form has a PXRD pattern (form 2) comprising peaks at diffraction angles of 6.5, 9.3, 13.62 Θ, ± 0.2 ° 2 Θ.

Embodiment I11 is a further embodiment of any one of embodiment I1 to I10 wherein the composition further comprises at least one additional agent.

In each of the embodiments of the methods of treatment of the present invention (including embodiments E, H1 to H18, embodiment F, and embodiments I1 to I11), each GLP-1R agonist and DGAT2 inhibitor may be present in the same composition or in separate compositions. The combined amount of GLP-1R agonist and DGAT2 inhibitor is therapeutically effective for the methods described herein. Even when the GLP-1R agonist and the DGT2 inhibitor are present in separate compositions, they may be administered simultaneously or sequentially; and when they are administered sequentially, they may be administered in any order.

GLP-1R agonist/FXR agonist combinations

Embodiments J, K and L relate to a combination of a GLP-1R agonist (including GLP-1R agonists described herein) and the FXR agonist 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid (also known as tropezol), or a pharmaceutically acceptable salt thereof.

Embodiment J1 is a further embodiment of embodiment J wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment J2 is a further embodiment of embodiment J or J1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment J3 is a further embodiment of embodiment J2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment J4 is a further embodiment of embodiment J wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment J5 is a further embodiment of embodiment J4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment J6 is a further embodiment of embodiment J5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment J7 is a further embodiment of embodiment J or any one of embodiments J1 to J6, wherein the 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof is 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro- 1, 3-benzothiazole-6-carboxylic acid.

Embodiment J8 is a further embodiment of embodiment J or any one of embodiments J1 to J7, wherein the composition further comprises at least one additional agent.

Embodiment K1 is a further embodiment of embodiment K, wherein the disease or disorder is fatty liver.

Embodiment K2 is a further embodiment of embodiment K, wherein the disease or disorder is non-alcoholic fatty liver disease.

Embodiment K3 is a further embodiment of embodiment K, wherein the disease or disorder is non-alcoholic steatohepatitis.

Embodiment K4 is a further embodiment of embodiment K, wherein the disease or disorder is nonalcoholic steatohepatitis with liver fibrosis.

Embodiment K5 is a further embodiment of embodiment K, wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis.

Embodiment K6 is a further embodiment of embodiment K wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with hepatocellular carcinoma.

Embodiment K7 is a further embodiment of embodiment K wherein the disease or disorder is nonalcoholic steatohepatitis with cirrhosis and with metabolic-related diseases.

Embodiment K8 is a further embodiment of embodiment K or any one of embodiments K1 to K7, wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorobenzyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment K9 is a further embodiment of embodiment K8 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment K10 is a further embodiment of embodiment K9 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment K11 is a further embodiment of embodiment K or any one of embodiments K1 to K7 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment K12 is a further embodiment of embodiment K11 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment K13 is a further embodiment of embodiment K12 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment K14 is a further embodiment of embodiment K or any one of embodiments K1 to K13, wherein 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof is 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro- 1, 3-benzothiazole-6-carboxylic acid.

Embodiment K15 is a further embodiment of embodiment K or any one of embodiments K1 to K14, the method further comprising administering at least one additional agent.

Embodiment L1 is a further embodiment of embodiment L wherein the GLP-1R agonist is 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof.

Embodiment L2 is a further embodiment of embodiment L1 wherein the GLP-1R agonist is a pharmaceutically acceptable salt of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid.

Embodiment L3 is a further embodiment of embodiment L2 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt.

Embodiment L4 is a further embodiment of embodiment L wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-4-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (pyridin-3-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-5-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-1, 2, 3-triazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof.

Embodiment L5 is a further embodiment of embodiment L4 wherein the GLP-1R agonist is selected from the group consisting of:

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid;

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2S) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid;

2- ({4- [ (2R) -2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid; and

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2,

or a pharmaceutically acceptable salt thereof. In a further embodiment, the GLP-1R agonist is a pharmaceutically acceptable salt.

Embodiment L6 is a further embodiment of embodiment L5 wherein the pharmaceutically acceptable salt is the tris (hydroxymethyl) aminomethane salt. In other embodiments, the tris (hydroxymethyl) aminomethane salt is in a crystalline form, such as the crystalline form described herein.

Embodiment L7 is a further embodiment of any one of embodiments L1 to L6 wherein 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid or a pharmaceutically acceptable salt thereof is 2- [ (1R,3R,5S) -3- ({ 5-cyclopropyl-3- [2- (trifluoromethoxy) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro- 1, 3-benzothiazole-6-carboxylic acid.

Embodiment L8 is a further embodiment of embodiment L or any one of embodiments L1 to L7 wherein the composition further comprises at least one additional agent.

In each embodiment of the method of treatment of the invention (including embodiment K, embodiments K1 to K15, embodiment L and embodiments L1 to L8), each GLP-1R agonist and FXR agonist may be present in the same composition or in separate compositions. The combined amount of GLP-1R agonist and FXR agonist is therapeutically effective for the methods described herein. Even when the GLP-1R agonist and the FXR agonist are present in separate compositions, they may be administered simultaneously or sequentially; and when they are administered sequentially, they may be administered in any order.

Combination medicament

The compounds in the combinations of the invention (in the compositions or methods) may be administered alone, or as separate agents or in fixed dose combinations or together with one or more additional therapeutic agents. When administered in combination, the components may be administered sequentially, in any order, at the same time (i.e., simultaneously) or at different time points. Thus, each component may be administered separately, but in close enough time proximity, to provide the desired therapeutic effect. Thus, the methods of treatment described herein include the administration of two or more agents in a combination using a combination of agents.

The combination medicament is administered to a patient or mammal in a therapeutically effective amount. By "therapeutically effective amount" is meant an amount of a compound of the invention that is effective to treat a desired disease/disorder (e.g., NASH) when administered to a mammal, either alone or in combination with an additional therapeutic agent.

Examples of suitable antidiabetic agents include (e.g., insulin, metformin, DPPIV inhibitors, GLP-1 agonists, analogs and mimetics, SGLT1, and SGLT2 inhibitors). Suitable antidiabetic agents include acetyl-CoA carboxylase- (ACC) inhibitors (such as those described in WO2009144554, WO2003072197, WO2009144555 and WO 2008065508), diacylglycerol O-acyltransferase 1(DGAT-1) inhibitors (such as those described in WO09016462 or WO2010086820, AZD7687 or LCQ908), monoacylglycerol O-acyltransferase inhibitors, Phosphodiesterase (PDE) -10 inhibitors, AMPK activators, sulfonylureas (e.g., acetohexamide), chlorpropamide (chlorpamide), hydropropamide (diabeside), glyburide (glibenclamide), glibenclamide, glimepiride (gliclazide), taxide (gliperide), gliquidone (gliquidone), glibenclamide (glibenclamide), glibenclamide (gliclazide), glibenclamide (glibenclamide), and glibenclamide (glibenclamide), Tolazamide (tolazamide) and tolbutamide (tolbutamide)), meglitinides, alpha-amylase inhibitors (e.g., amylastatin (tenamistat), auristatin (trestatin) and AL-3688), alpha-glucosidase inhibitors (e.g., acarbose), alpha-glucosidase inhibitors (e.g., lipo (adiposine), crilagogliose (camigiose), emiglitate (emiglitate), miglitol (miglitol), voglibose (voglibose), pradimicin (pradimicin) -Q and sabotatin (salbestatin)), PPAR γ agonists (e.g., balaglitazone (balaglitazone), cycloglitazone (ciglitazone), darglitazone (darglitazone), enon (englitazone), glitazone (tigolozone), pioglitazone (2440-gix), and glitazone (e.g., PPAR-1539), glitazone (2440, 1539, and/or gla), PPAR-0933, e.g., glitazone (gla), PPAR-r, tigone (e), glitazone (e.g., tigolone (2433), glitazone (e), glitazone (e.g., tigolone (e), glitazone (e), PPAR-r (e.g., glitazone (p), glitazone (e., KRP-297, L-796449, LR-90, MK-0767, and SB-219994), biguanides (e.g., metformin), glucagon-like peptide 1(GLP-1) modulators, such as agonists (e.g., exendin-3 and exendin-4), liraglutide, albiglutide, exenatideAlbiuretic peptides, lixividepsipeptides, dolabrotalin, somaglutelin, NN-9924, TTP-054, protein tyrosine phosphatase-1B (PTP-1B) inhibitors (e.g., curvatamine (rodussquesine), ceteuraldehyde (hysosal) extract and compounds disclosed by Zhang, S. et al, Drug Discovery Today,12(9/10),373-381 (2007)), SIRT-1 activators (e.g., resveratrol (resvertrol), JNK 2245840 or GSK184072), dipeptidyl peptidase IV (DPP-IV) inhibitors (e.g., those described in WO2005116014, sitagliptin, vildagliptin, alogliptin, polyglastin, linagliptin and saxagliptin (saxagliptin)), insulin secretagogues, fatty acid oxidation inhibitors, A2, amino kinase-jjjn inhibitors (WO 343937) such as glucokinase 01037, inhibitors (WO 34 GKa) and inhibitors (Zhang, S. et al, S WO2010103438, WO2010013161, WO2007122482, TTP-399, TTP-355, TTP-547, AZD1656, ARRY403, MK-0599, TAK-329, AZD5658 or GKM-001), insulin mimetics, glycogen phosphorylase inhibitors (e.g., GSK1362885), VPAC2 receptor inhibitors Agonists, SGLT2 inhibitors (such as those described in E.C. Chao et al, Nature Reviews Drug Discovery 9,551-5,559 (July 2010), including dapagliflozin, canagliflozin, engagliflozin, tolagliflozin (tofogliflozin) (CSG452), egagliflozin, ASP-1941, THR1474, TS-071, ISIS388626 and LX4211, and those described in WO 2010023594), glucagon receptor modulators (such as those described in Demong, D.E. et al, Annual report in Medicinal Chemistry 2008,43,119,137), GPR119 modulators, particularly agonists (such as those described in WO2010140092, WO 0122010128425, 2018414, WO 20157, Jones R149, J.149, Jones et al, J.R. 2009, J.9, J.51,82, EP-76,102,82, EP-76,80, EP-9,82, EP-9, EP-80, EP-9, EP-3, EP-80, EP-9, EP-S-9, EP-S3, preferably-S3, S-S, TGR5 (also known as GPBAR1) receptor modulators, in particular agonists (such as those described in Current Topics in Medicinal Chemistry,2010,10(4),386-396 and INT777, by Zhong, m.), GPR40 agonists (such as those described in Medicinal Chemistry,2008,43,75-85, by annular Reports in Medicinal Chemistry), including but not limited to TAK-875, GPR120 modulators, in particular agonists, high affinity nicotinic acid receptor (HM74A) activators and SGLT1 inhibitors (such as GSK 1614235). A further representative list of antidiabetic agents which may be combined with the compounds of the present invention may be found, for example, on page 28 line 35 to page 30 line 19 of WO 2011005611. Preferred antidiabetic agents are metformin and DPP-IV inhibitors (e.g., sitagliptin, vildagliptin, alogliptin, doxogliptin, linagliptin and saxagliptin). Other antidiabetic agents may include carnitine palmitoyl transferase inhibitors or modulators, fructose 1, 6-bisphosphatase inhibitors, aldose reductase inhibitors, mineralocorticoid receptor inhibitors, TORC2 inhibitors, CCR2 and/or CCR5 inhibitors, PKC isoform (e.g., PKC α, PKC β, PKC γ) inhibitors, fatty acid synthase inhibitors, serine palmitoyl transferase inhibitors, GPR81 modulators, GPR39 modulators, GPR43 modulators, GPR41 modulators, GPR105 modulators, kv1.3 modulators, retinol binding protein 4 modulators, glucocorticoids Sterol receptor modulators, somatostatin receptor modulators (e.g., SSTR1, SSTR2, SSTR3, and SSTR 5); PDHK2 or PDHK4 inhibitors or modulators, MAP4K4 inhibitors, modulators of the IL1 family including IL1 β, RXR α modulators. In addition, suitable anti-diabetic agents include the mechanisms listed by Expert opin the ther paper, 2010,20(12),1627-51 of Carpino, p.a., Goodwin, b.

Suitable antiobesity agents include 11 beta-hydroxysteroid dehydrogenase-1 (11 beta-HSD type 1) inhibitors, stearoyl-CoA desaturase-1 (SCD-1) inhibitors, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors such as sibutramine (sibutramine), sympathomimetics, beta3Adrenergic agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (OB protein), leptin analogs, leptin agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin (lipstatin), i.e., orlistat), appetite inhibitors (such as bombesin agonists), neuropeptide-Y antagonists (e.g., NPY Y5 antagonists), PYY 3-36(including analogs thereof), thyromimetic agents, dehydroepiandrosterone or analogs thereof, glucocorticoid agonist or antagonist, orexin antagonist, glucagon-like peptide-1 agonist, ciliary neurotrophic factor (such as those available from Regeneron Pharmaceuticals, inc., Tarrytown, NY and Procter)&Axokine from Gamble Company, Cincinnati, OHTM) Human agouti-related protein (AGRP) inhibitors, ghrelin antagonists, histamine 3 antagonists or inverse agonists, neuregulin U agonists, MTP/apolipoprotein B inhibitors (e.g., gut-selective MTP inhibitors such as desloratadine), opioid antagonists, orexin antagonists, naltrexone in combination with bupropion (buproprion), and the like.

Preferred anti-obesity agents for use in the combination aspect of the invention include gut-selective MTP inhibitors (e.g. desloratadine, miritapide and implitapide), R56918(CAS number 403987) and CAS number 913541-47-6) CCKa agonists (e.g., N-benzyl-2- [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4, 5-dihydro-2, 3,6,10 b-tetraazabenzo [ e ] a described in PCT publication No. WO 2005/116034 or US publication No. 2005-0267100A 1) ]Azulen-6-yl]-N-isopropylacetamide), 5HT2c agonists (e.g. lorcaserin), MCR4 agonists (e.g. the compounds described in US 6,818,658), lipase inhibitors (e.g. Cetilistat), PYY3-36(As used herein, "PYY3-36"includes analogs, such as pegylated PYY3-36Such as those described in US publication 2006/0178501), opioid antagonists (e.g., naltrexone), combinations of naltrexone and bupropion, oleoyl estrone (CAS No. 180003-17-2), obinepitide (TM30338), pramlintide (pramlintide)Tesofensine (NS2330), leptin, liraglutide, bromocriptine, orlistat, exenatideAOD-9604(CAS number 221231-10-3), phentermine (phentermine) and topiramate (topiramate) (trade name: Qsymia) and sibutramine. The compounds of the invention and combination therapies are preferably administered in conjunction with exercise and a coherent diet.

The compounds of the present invention may be used in combination with the following cholesterol modulating agents (including cholesterol lowering agents): such as lipase inhibitors, HMG-CoA reductase inhibitors, HMG-CoA synthase inhibitors, HMG-CoA reductase gene expression inhibitors, HMG-CoA synthase gene expression inhibitors, MTP/apolipoprotein B secretion inhibitors, CETP inhibitors, bile acid absorption inhibitors, cholesterol synthesis inhibitors, squalene synthetase inhibitors, squalene epoxidase inhibitors, squalene cyclase inhibitors, combined squalene epoxidase/squalene cyclase inhibitors, fibrates, nicotinic acid, ion exchange resins, antioxidants, ACAT inhibitors or bile acid sequestrants or agents such as milbemes (mipomensen).

Examples of suitable cholesterol/lipid lowering agents and lipid profile therapies include: HMG-CoA reductase inhibitors (e.g., pravastatin (pravastatin), lovastatin (lovastatin), atorvastatin (atorvastatin), simvastatin (simvastatin), fluvastatin (fluvastatin), NK-104 (also known as itavastatin (itavastatin) or Nivastatin (nisvastatin) or Nibestatin (nisstatin)) and ZD-4522 (also known as rosuvastatin (rosuvastatin) or atorvastatin (atavastatin) or visastatin (visastatin))); a squalene synthetase inhibitor; a fibrate; bile acid sequestrants (such as quintesla (quetran)); an ACAT inhibitor; an MTP inhibitor; a lipoxygenase inhibitor; cholesterol absorption inhibitors; and cholesteryl ester transfer protein inhibitors. Other atherosclerotic agents include PCSK9 modulators.

In another embodiment, the compounds of the invention may be co-administered with an agent for the treatment of nonalcoholic steatohepatitis (NASH) and/or nonalcoholic fatty liver disease (NAFLD), such as orlistat, TZD and other insulin sensitizers, FGF21 analogs, metformin, ethyl omega-3-carboxylates (e.g., Lovaza (Lovaza)), fibrates, HMG CoA-reductase inhibitors, ezetimibe (ezetimbe), Probucol (Probucol), ursodeoxycholic acid, TGR5 agonists, FXR agonists, vitamin E, betaine, pentoxifylline, CB1 antagonists, carnitine, N-acetyl cysteine, reduced glutathione, lorcaserin, naltrexone and bupropion combinations, SGLT2 inhibitors, phentermine, topiramate, incretin (GLP and GIP) analogs, and angiotensin receptor blockers.

In another embodiment, the additional agent is selected from the group consisting of: cysteamine or a pharmaceutically acceptable salt thereof, cystamine or a pharmaceutically acceptable salt thereof, an antioxidant compound, lecithin, a vitamin B complex, a bile salt formulation, a cannabinoid-1 (CB1) receptor antagonist, a cannabinoid-1 (CB1) receptor inverse agonist, a peroxisome proliferator activated receptor activity modulator, benzothiazepineOr a benzothiepine compound, an RNA antisense construct that inhibits protein tyrosine phosphatase PTPRU, heteroatom-linked substituted piperidines and derivatives thereof, an aziridine derivative capable of inhibiting stearoyl-coenzyme α δ -9 desaturase, an acyl amide compound having secretagogue activity or inducer activity of adiponectin, a quaternary ammonium compound, glatiramer acetate, a pentraxin protein, an HMG-CoA reductase inhibitor, N-acetyl cysteine, an isoflavone compound, a macrolide antibiotic, a galectin inhibitor, an antibody, or any combination of the above.

Additional therapeutic agents include anticoagulants or coagulation inhibitors, antiplatelet or platelet inhibitors, thrombin inhibitors, thrombolytic or fibrinolytic agents, antiarrhythmic agents, antihypertensive agents, calcium channel blockers (L-and T-type), cardiac glycosides, diuretics, mineralocorticoid receptor antagonists, NO-yielding agents (such as organic nitrates), NO-promoting agents (such as phosphodiesterase inhibitors), cholesterol/lipid lowering agents and lipid profile therapies, antidiabetics, antidepressants, anti-inflammatory agents (steroids and non-steroids), anti-osteoporosis agents, hormone replacement therapies, oral contraceptives, antiobesity agents, anxiolytic agents, antiproliferative agents, antineoplastic agents, antiulcer agents and antigastroesophageal reflux disease agents, growth hormones and/or growth hormone secretagogues, thyroid mimetics (including thyroid hormone receptor antagonists), Anti-infective, antiviral, antibacterial and antifungal agents.

Including those used in the ICU environment such as dobutamine (dobutamine), dopamine, epinephrine (epinephrine), nitroglycerin (nitroglycerin), nitroprusside (nitropraside), and the like.

Including combination agents useful in the treatment of vasculitis, such as azathioprine (azathioprine), cyclophosphamide, mycophenolate mofetil (mycophenolate mofetil), rituximab (rituximab), and the like.

In another embodiment, the present invention provides wherein the additional agent (third agent) is at least one agent selected from the group consisting of: factor Xa inhibitors, anticoagulants, antiplatelet agents, thrombin inhibitors, thrombolytic agents, and fibrinolytic agents. Exemplary factor Xa inhibitors include apixaban (apixaban) and rivaroxaban (rivaroxaban). Examples of suitable anticoagulants for use in combination with the compounds of the invention include heparin (e.g., unfractionated heparin and low molecular weight heparins such as enoxaparin (enoxaparin) and dalteparin).

In another preferred embodiment, the additional agent (third agent) is at least one agent selected from the group consisting of: warfarin (warfarin), dabigatran (dabigatran), unfractionated heparin, low molecular weight heparin, synthetic pentose, hirudin, argatroban (argatroban), aspirin (aspirin), ibuprofen (ibuprofen), naproxen (naproxen), sulindac (sulindac), indomethacin (indomethacin), mefenamic acid (mefenamate), droxicam (droxicam), diclofenac (diclofenac), fensulazone (sulfyrazone), piroxicam (piroxicam), ticlopidine (clopidogrel), gray (clopidogrel), tirofiban (tirofiban), eptifibatidine (eptifibatidine), eptidine (epibatidine), eptifibatidine (epibatidine), fibatidine (epibatidine), asparatbazide (epitaxine), acizumab (abciximab), melagatran (melagatran), modified hirudin (histolytica), plasmin (plasminogen, plasminogen activator, plasminogen activator.

The preferred third agent is at least one antiplatelet agent. Particularly preferred antiplatelet agents are aspirin and clopidogrel.

The term antiplatelet agent (or platelet inhibitor), as used herein, refers to an agent that inhibits platelet function, for example, by inhibiting platelet aggregation, adhesion, or granule secretion. The agents include, but are not limited to, various known nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamic acid, droxicam, diclofenac, fensulazone, piroxicam, and pharmaceutically acceptable salts or prodrugs thereof. Among the NSAIDs, aspirin (acetylsalicylic acid or ASA) and COX-2 inhibitors (such as CELEBREX or piroxicam) are preferred. Other suitable platelet inhibitors include IIb/IIIa antagonists (e.g., tirofiban, eptifibatide, and abciximab), thromboxane-a 2-receptor antagonists (e.g., ifetroban), thromboxane a 2-synthase inhibitors, PDE-III inhibitors (e.g., pradal (Pletal), dipyridamole (dipyridamole)), and pharmaceutically acceptable salts or prodrugs of the above agents.

The term antiplatelet agent (or platelet inhibitor), as used in the present application, is also intended to include ADP (adenosine diphosphate) receptor antagonists, preferably purinergic receptor P 2Y1And P2Y12Antagonist of (1), with P2Y12Even more preferably. Preferred P2Y12Receptor antagonists include ticagrelor (ticagrelor), prasugrel (prasugrel), ticlopidine and clopidogrel, including pharmaceutically acceptable salts or prodrugs thereof. Clopidogrel is an even more preferred agent. Ticlopidine and clopidogrel are also preferred compounds because they are known to be gentle to the digestive tract when used.

The term thrombin inhibitor (or antithrombin agent) as used in this application denotes a serine protease thrombin inhibitor. Various thrombin-mediated processes, such as thrombin-mediated platelet activation (i.e., for example, platelet aggregation and/or granule secretion of plasminogen activator inhibitor-1 and/or serotonin) and/or fibrin formation, are disrupted by inhibition of thrombin. Many thrombin inhibitors are known to those skilled in the art, and such inhibitors are contemplated for use in combination with the compounds of the present invention. Such inhibitors include, but are not limited to, boroarginine (boroargine) derivatives, boropeptides, dabigatran, heparin, hirudin, argatroban (argatroban), and melagatran, including pharmaceutically acceptable salts and prodrugs thereof. Boroarginine derivatives and boropeptides include N-acetyl and peptide derivatives of boronic acids, such as C-terminal alpha-amino boronic acid derivatives of lysine, ornithine, arginine, homoarginine, and their corresponding isothiouronium (isothiouronium) analogs. The term hirudin as used in this application includes suitable derivatives or analogues of hirudin, referred to herein as hirulog (hirulog), such as hirudin disulfate. The term thrombolytic or fibrinolytic agent (or thrombolytic or fibrinolytic) as used in the present application denotes an agent that dissolves blood clots (thrombi). Such agents include tissue plasminogen activator (natural or recombinant) and modified forms thereof, anistreplase, urokinase, streptokinase, Tenecteplase (TNK), lanoteplase (lanoteplase) (nPA), factor vila inhibitors, PAI-1 inhibitors (i.e., inactivators of tissue plasminogen activator inhibitors), alpha 2-antifibrinolytic enzyme inhibitors, and anisylated plasminogen streptokinase activator complexes, including pharmaceutically acceptable salts or prodrugs thereof. The term anistreplase as used in this application refers to an anisylated plasminogen streptokinase activator complex as described, for example, in EP 028,489, the disclosure of which is hereby incorporated by reference into this application. The term urokinase as used in this application is intended to mean both dual and single chain urokinase, the latter also being referred to as prourokinase in this application.

Examples of suitable antiarrhythmic agents include: class I agents (such as propafenone); class II agents (such as metoprolol, atenolol, carvedilol, and propranolol); class III agents (such as sotalol (sotalol), dofetilide (dofetilide), amiodarone (amiodarone), azimilide (azimilide), and ibutilide (ibutilide)); class IV agents (such as diltiazem)(dithezem) and verapamil (verapamil)); k+Channel openers such as IAch inhibitors and IKur inhibitors (e.g. those disclosed in WO 01/40231).

The compounds of the present invention may be used in combination with an antihypertensive agent, and the hypertensive activity is readily determined by one skilled in the art according to standard assays (e.g., measuring blood pressure). Examples of suitable antihypertensive agents include: an alpha adrenergic blocker; beta adrenergic blockers; calcium channel blockers (e.g., diltiazem)Verapamil, nifedipine (nifedipine), and amlodipine (amlodipine)); vasodilators (e.g., hydrazine (hydrazine)), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, chlorothiazide, trichlormethiazide, polythiazide, benthiazide, triamcinolone acetonide, chlorothalolone, tolteromide, pyroximide, massomine, bumetamide, triamcinolone acetonide, chlorothiadone, torasemide, furosemide, triamcinolone, tramethamine, bumetamide, triamcinolone acetonide, triamcinolone, spironolactone, and the like); a renin inhibitor; ACE inhibitors (e.g., captopril (captopril), zofenopril (zofenopril), fosinopril (fosinopril), enalapril (enalapril), enalapril (ceranopril), cilazapril (cilazopril), delapril (delapril), pentopril (pentopril), quinapril (quinapril), ramipril (ramipril), lisinopril (lisinopril)); AT-1 receptor antagonists (e.g., losartan (losartan), ebisatan (irbesartan), valsartan (valsartan)); ET receptor antagonists (e.g., sitaxsentan (sitaxsentan), atrasentan (atrsentan), and the compounds disclosed in U.S. patent nos. 5,612,359 and 6,043,265); dual ET/AII antagonists (e.g., the compounds disclosed in WO 00/01389); neutral Endopeptidase (NEP) inhibitors; vasopeptidase inhibitors (bis-NEP-ACE inhibitors) (e.g., gemotrilat (gemopatrilat) and nitrate). An exemplary anti-angina agent is ivabradine (ivabradine).

Examples of suitable calcium channel blockers (L-form or T-form) include diltiazemVerapamil, nifedipine, amlodipine and mibefradil (mybefradil).

Examples of suitable cardiac glycosides include rehmannia glutinosa (digitalis) and ouabain (ouabain).

In one embodiment, the compounds of the inventionMay be co-administered with one or more diuretics. Examples of suitable diuretics include (a) loop diuretics such as furosemide (such as LASIX)TM) Torasemide (such as DEMADEX)TM) Bemetanide (such as BUMEX)TM) And ethacrynic acid (such as EDECRIN)TM) (ii) a (b) Thiazine diuretics, such as chlorothiazide (such as DIURIL)TM、ESIDRIXTMOr HYDRODIRILTM) Hydrochlorothiazide (such as MICROZIDE)TMOr ORETICTM) Benzylthiazine, hydroflumethiazide (such as salulon)TM) Benflumethiazide, methylchlorothiazide, polythiazide, trichlormethiazide and indapamide (such as LOZOL)TM) (ii) a (c) Benzo [ c ]]Diuretic of the pyrrolidone type, such as chlorthalidone (e.g. HYGROTON)TM) And metolazone (metolazone) (such as zarxolyn)TM) (ii) a (d) A quinazoline-type diuretic such as quinethazolinone (quinethazone); and (e) potassium sparing diuretics such as triamterene (such as DYRENIUM)TM) And amiloride (such as MIDAMOR) TMOr MODURETICTM)。

In another embodiment, a compound of the invention may be co-administered with a loop diuretic. In yet another embodiment, the loop diuretic is selected from furosemide and torasemide. In yet another embodiment, the compounds of the present invention may be co-administered with furosemide. In yet another embodiment, the compounds of the invention may be co-administered with torsemide, optionally in a controlled or modified release form thereof.

In another embodiment, the compounds of the invention may be co-administered with a thiazide diuretic. In yet another embodiment, the thiazide-type diuretic is selected from the group consisting of chlorothiazide and hydrochlorothiazide. In yet another embodiment, the compounds of the present invention may be co-administered with chlorothiazide. In yet another embodiment, the compounds of the present invention may be co-administered with hydrochlorothiazide.

In another embodiment, the compounds of the present invention may be co-administered with a phthalimidine-type diuretic. In yet another embodiment, the phthalimidine-type diuretic is chlorthalidone. Examples of suitable mineralocorticoid receptor antagonists include spironolactone and eplerenone (eplerenone). Examples of suitable phosphodiesterase inhibitors include: PDE III inhibitors (such as cilostazol); and PDE V inhibitors such as sildenafil (sildenafil).

It will be appreciated by those skilled in the art that the compounds of the present invention may also be used in conjunction with other cardiovascular or cerebrovascular therapies, including PCI, stent implantation, drug-coated stents, stem cell therapy, and medical devices, such as implanted pacemakers, defibrillators, or cardiac resynchronization therapy.

The dosage of each therapeutic agent (e.g., a GLP-1R agonist) and any additional therapeutic agents described herein will generally depend on a number of factors, including the health of the subject to be treated, the extent of treatment desired, the nature and kind of concurrent treatment (if any), and the frequency of treatment and nature of the effect desired. The dosage range for each therapeutic agent is generally in the range of about 0.001mg to about 100mg per kilogram of individual body weight per day, preferably in the range of about 0.1mg to about 10mg per kilogram of individual body weight per day. However, some variability in the general dosage range may also be required depending on the age and weight of the individual to be treated, the intended route of administration, the particular anti-obesity agent to be administered, and the like. Determination of the dosage range and optimal dosage for a particular patient to have the benefit of this invention is well within the ability of those of ordinary skill in the art.

According to the treatment methods of the present invention, the compounds of the present invention or a combination of the compounds of the present invention and at least one additional agent (referred to herein as a "combination") are administered, preferably in the form of a pharmaceutical composition, to an individual in need of such treatment. In a combined aspect of the invention, a compound of the invention and at least one other agent (e.g., another anti-obesity agent) may be administered alone or in a pharmaceutical composition comprising both. Oral administration is generally preferred for such administration.

When a compound of the invention is administered in combination with at least one other agent, such administration may be sequential in time or simultaneous in time. The combination of drugs is generally preferred for simultaneous administration. The compounds of the invention and additional agents for sequential administration may be administered in any order. Oral administration is generally preferred for such administration. Oral and simultaneous administration are particularly preferred for this administration. When the compound of the invention and the additional agent are administered sequentially, the administration of each agent may be the same or different.

According to the method of the invention, the compound or combination of the invention is preferably administered in the form of a pharmaceutical composition. Thus, the compounds or combinations of the present invention may be administered to a patient individually or together in any conventional oral, rectal, transdermal, parenteral (e.g., intravenous, intramuscular, or subcutaneous), intracisternal, intravaginal, intraperitoneal, topical (e.g., powders, ointments, creams, sprays, or lotions), intrabuccal, or nasal dosage form (e.g., sprays, drops, or inhalants).

The compounds or combinations of the present invention may be administered alone, but will generally be administered in admixture with one or more suitable pharmaceutical excipients, adjuvants, diluents or carriers, which are known in the art and selected with respect to the intended route of administration and standard pharmaceutical practice. The compounds or combinations of the present invention may be formulated to provide immediate, delayed, modified, sustained, pulsed or controlled release dosage forms, depending on the desired route of administration and the specificity of the release profile commensurate with the therapeutic requirements.

The pharmaceutical compositions comprise a compound or combination of the invention in an amount generally in the range of from about 1% to about 75%, 80%, 85%, 90% or even 95% (by weight) of the composition, often in the range of from about 1%, 2% or 3% to about 50%, 60% or 70%, more often in the range of from about 1%, 2% or 3% to less than 50%, such as about 25%, 30% or 35%.

Methods for preparing various pharmaceutical compositions having a specific amount of active compound are known to those skilled in the art. See, for example, Remington, The Practice of Pharmacy, Lippincott Williams and Wilkins, Baltimore Md.20. th ed.2000.

Compositions suitable for parenteral injection typically include pharmaceutically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers or diluents (including solvents and vehicles) include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol and the like), suitable mixtures thereof, triglycerides (including vegetable oils such as olive oil) and injectable organic esters (such as ethyl oleate). A preferred carrier is caprylic/capric acid ester of glycerol or propylene glycol available from the miglyol.rtm. brand of Condea Vista co., Cranford, n.j. (e.g., miglyol.rtm.812, miglyol.rtm.829, miglyol.rtm.840). Proper fluidity can be maintained, for example, by the use of a coating (such as lecithin), by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.

The compositions for parenteral injection may also contain excipients such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of microbial contamination of the composition can be accomplished with various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical composition can be brought about by the use of agents capable of delaying absorption, for example, aluminum monostearate and gelatin.

Solid dosage forms for oral administration include capsules, tablets, chewables, lozenges, pills, powders, and multiparticulate formulations (granules). In such solid dosage forms, the compound or combination of the present invention is mixed with at least one inert excipient, diluent or carrier. Suitable excipients, diluents or carriers include materials such as sodium citrate or dicalcium phosphate and/or (a) one or more fillers or extenders (e.g. microcrystalline cellulose (from FMC corpTMObtained), starch, lactose, sucrose, mannitol, silicic acid, xylitol, sorbitol, dextrose, dibasic calcium phosphate, dextrin, α -cyclodextrin, β -cyclodextrin, polyethylene glycol, medium chain fatty acids, titanium oxide, magnesium oxide, aluminum oxide, and the like); (b) one or more binders (e.g., carboxymethylcellulose, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, gelatin, gum arabic, ethylcellulose, polyvinyl alcohol, pullulan, pregelatinized Starch, agar, tragacanth, alginates, gelatin, polyvinylpyrrolidone, sucrose, acacia, and the like); (c) one or more humectants (e.g., glycerin and the like); (d) one or more disintegrating agents (e.g., agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, sodium carbonate, sodium lauryl sulfate, sodium starch glycolate (ex Edward Mendell Co. as Explotab;)TMObtained), crospolyvinylpyrrolidone, croscarmellose sodium type A (as Ac-di-sol)TMProcurement), polacrilin (polyacrilin) potassium (ion exchange resin) and the like); (e) one or more solution retarding agents (e.g., paraffin and the like); (f) one or more absorption accelerators (e.g., quaternary ammonium compounds and the like); (g) one or more wetting agents (e.g., cetyl alcohol, glyceryl monostearate, and the like); (h) one or more adsorbents (e.g., kaolin, bentonite, and the like); and/or (i) one or more lubricants (e.g., talc, calcium stearate, magnesium stearate, stearic acid, polyoxyethylene stearate, cetyl alcohol, talc, hydrogenated castor oil, sucrose esters of fatty acids, dimethylpolysiloxane, microcrystalline wax, yellow beeswax, white beeswax, solid polyethylene glycol, sodium lauryl sulfate, and the like). In the case of capsules and tablets, the dosage forms may also contain buffering agents.

Solid compositions of similar form may also be employed as fillers in soft or hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

Solid dosage forms (such as tablets, dragees, capsules and granules) can be prepared with coatings and shells such as enteric coatings and other coatings well known in the art. They may also contain opacifying agents and may also have such compositions that they release the compounds of the invention and/or additional agents in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The drug may also be in one or more microencapsulated forms, if appropriate, with the above-mentioned excipients.

For tablets, the active agentTypically constitute less than 50% (by weight) of the formulation, for example less than about 10%, such as 5% or 2.5% (by weight). The main part of the formulation comprises fillers, diluents, disintegrants, lubricants and optionally flavouring agents. The composition of such excipients is well known in the art. Typically, the filler/diluent comprises a mixture of two or more of the following components: microcrystalline cellulose, mannitol, lactose (all types), starch and dicalcium phosphate. The filler/diluent mixture typically comprises less than 98% of the formulation, and preferably less than 95%, for example 93.5%. Preferably the disintegrant comprises Ac-di-sol TM、ExplotabTMStarch and sodium lauryl sulfate. When present, disintegrants often comprise less than 10% or less than 5%, for example about 3% of the formulation. Preferably, the lubricant is magnesium stearate. When present, lubricants often comprise less than 5% or less than 3%, e.g., about 1%, of the formulation.

Tablets may be made by standard tableting methods, for example, direct compression or wet, dry or melt granulation, melt congealing and extrusion. The tablet core may be single or multi-layered and may be coated with suitable protective films as known in the art.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the compounds or combinations of the present invention, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, e.g., ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (e.g., cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil, sesame seed oil, and the like), miglyole. rtm (available from CONDEA Vista co., Cranford, n.j.), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of such materials, and the like.

In addition to such inert diluents, the compositions can also include excipients such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Of compounds or combinations of the inventionOral liquid dosage forms comprise solutions in which the active compound is completely dissolved. Examples of solvents include all the pharmaceutically antecedent solvents suitable for oral administration, in particular those which allow the compounds of the invention to exhibit good solubility therein, such as polyethylene glycol, polypropylene glycol, edible oils and systems based on glyceryl and glyceryl esters. Glyceryl and glyceryl ester based systems may include, for example, the following branded products (and corresponding generic name products): captexTM355 EP (glyceryl tricaprylate/caprate from Abitec, Columbus Ohio), CrodamolTMGTC/C (medium chain triglycerides, from Croda, Cowick Hall, UK) or LabrafacTMCC (Medium chain Triglycerides, from Gattefose), CaptexTM500P (glyceryl triacetate), i.e. glyceryl triacetate (triacetin), from Abitec, CapmulTMMCM (Medium chain mono-and diglycerides from Abitec), MigyolTM812 (caprylic/capric triglyceride from Condea, Cranford N.J.), Migyol TM829 (caprylic/capric/succinic triglyceride from Condea), MigyolTM840 (propylene glycol dicaprylate/dicaprate from Condea), LabrafilTMM1944CS (oleoyl macrogol-6 glycerides from Gattefose), PeceolTM(Glycerol monooleate from Gattefose) and MaisineTM35-1 (glycerol monooleate from Gattefose). Of particular interest are medium chains (about C)8To C10) Triglyceride oils. These solvents often make up the major portion of the composition, i.e., greater than about 50%, often greater than about 80%, such as about 95% or 99%. Adjuvants and additives may also be included with the solvent, primarily as taste masking agents, palatability and flavoring agents, antioxidants, stabilizers, texture and viscosity modifiers, and cosolvents.

Suspensions, in addition to a compound or combination of the invention, may additionally contain carriers such as suspending agents, for example ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of such substances and the like.

Compositions for rectal or vaginal administration preferably comprise suppositories which can be prepared by mixing the compounds or combinations of the invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ordinary room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity, thus releasing the active ingredient.

Dosage forms for topical administration of the compounds or combinations of the present invention include ointments, creams, lotions, powders and sprays. The medicament is mixed with a pharmaceutically acceptable excipient, diluent or carrier and any preservatives, buffers or propellants which may be required.

When the compound is poorly soluble in water (e.g., less than about 1 μ g/mL), then a liquid composition dissolved in a non-aqueous solvent, such as the medium chain triglyceride oils discussed above, is a preferred dosage form for the compound.

Solid amorphous dispersions, including those formed by spray drying, are also preferred dosage forms of the poorly soluble compounds of the present invention. By "solid amorphous dispersion" is meant a solid material in which at least a portion of the poorly soluble compound is amorphous and dispersed in a water soluble polymer. By "amorphous" is meant that the poorly soluble compound is not in a crystalline form. By "crystalline form" is meant a compound that exhibits a three-dimensional long-range order of at least 100 repeating units in each dimension. Thus, the term amorphous is intended to include not only substantially disordered materials, but also materials that may have a somewhat lesser degree of order, but less than three dimensions and/or only within a short distance. Amorphous materials can be characterized by techniques known in the art, such as powder x-ray diffraction (PXRD) crystallography, solid state NMR, or thermal analysis techniques, such as Differential Scanning Calorimetry (DSC).

At least a majority (i.e., at least about 60 weight percent) of the poorly soluble compound in the solid amorphous dispersion is preferably amorphous. The compounds may be present in relatively pure amorphous domains or regions within the solid amorphous dispersion as solid solutions of the compounds uniformly distributed throughout the polymer or in the stated states or any combination of their states with intermediates in between. The solid amorphous dispersion is preferably substantially homogeneous so that the amorphous compound is dispersed as uniformly as possible throughout the polymer. As used herein, "substantially homogeneous" means that the portion of the compound present in the relatively pure amorphous domains or regions within the solid amorphous dispersion is relatively small, on the order of less than 20% by weight of the total amount of drug, and preferably less than 10% by weight.

Water-soluble polymers suitable for use in solid amorphous dispersions should be inert in the sense that they do not chemically react in an adverse manner with poorly soluble compounds, are pharmaceutically acceptable, and have at least some solubility in aqueous solutions at physiologically relevant pH (e.g., 1 to 8). The polymer may be neutral or ionizable and should have an aqueous solubility of at least 0.1mg/mL over at least a portion of the pH range of 1 to 8.

Water soluble polymers suitable for use with the present invention may be cellulosic or non-cellulosic materials. The polymer may be neutral or ionizable in aqueous solution. Among the polymers, ionizable cellulose polymers are preferred, and ionizable cellulose polymers are more preferred.

Exemplary water-soluble polymers include hydroxypropylmethylcellulose acetate succinate (HPMCAS), Hydroxypropylmethylcellulose (HPMC), hydroxypropylmethylcellulose phthalate (HPMCP), carboxymethylethylcellulose (CMEC), Cellulose Acetate Phthalate (CAP), cellulose acetate 1,2, 4-trimellitate (CAT), polyvinylpyrrolidone (PVP), Hydroxypropylcellulose (HPC), Methylcellulose (MC), block copolymers of ethylene oxide and propylene oxide (PEO/PPO, also known as poloxamers), and mixtures thereof. Particularly preferred polymers include HPMCAS, HPMC, HPMCP, CMEC, CAP, CAT, PVP, poloxamer and mixtures thereof. Most preferred is HPMCAS. See European patent application publication No. 0901786A 2, the disclosure of which is incorporated herein by reference.

The solid amorphous dispersion may be prepared according to any method for forming a solid amorphous dispersion that results in at least a majority (at least 60%) of the poorly soluble compound being in an amorphous state. Such methods include mechanical methods, thermal methods, and solvent methods. Exemplary mechanical methods include grinding and extrusion; the melting method comprises high-temperature fusion, solvent modified fusion and melting congealing method; and solvent methods including non-solvent precipitation, spray coating and spray drying. See, for example, the following U.S. patents, the relevant disclosures of which are incorporated herein by reference: U.S. Pat. Nos. 5,456,923 and 5,939,099, which describe forming a dispersion in an extrusion process; 5,340,591 and 4,673,564 which illustrate the formation of dispersions by a milling process; and 5,707,646 and 4,894,235 which illustrate the formation of dispersions by the melt congealing process. In a preferred process, the solid amorphous dispersion is formed by spray drying, as disclosed in european patent application publication No. 0901786 a 2. In this process, the compound and polymer are dissolved in a solvent, such as acetone or methanol, and then the solvent is rapidly removed from the solution by spray drying to form a solid amorphous dispersion. The solid amorphous dispersion prepared may contain up to about 99 wt% of the compound, e.g., 1 wt%, 5 wt%, 10 wt%, 25 wt%, 50 wt%, 75 wt%, 95 wt%, or 98 wt%, as desired.

The solid dispersion may be used as a dosage form per se, or it may be used as a manufacturing application product (MUP) in the preparation of other dosage forms, such as capsules, tablets, solutions or suspensions. Examples of aqueous suspensions are 1: 1(w/w) compound/aqueous suspension of HPMCAS-HF spray-dried dispersion containing 2.5mg/mL compound in 2% polysorbate-80. Solid dispersions for tablets or capsules are usually mixed with other excipients or adjuvants typically found in such dosage forms. For example, an exemplary filler for a capsule contains 2: 1(w/w) compound/HPMCAS-MF spray dried dispersion (60%), lactose (fast flow) (15%), microcrystalline cellulose (e.g., avicel. sup. (R0-102)) (15.8%), sodium starch (7%), sodium lauryl sulfate (2%) and magnesium stearate (1%).

HPMCAS polymers were obtained from Shin-Etsu Chemical co., LTD, Tokyo, Japan as low, medium and high aqoa. Higher MF and HF are generally preferred.

The following paragraphs describe exemplary formulations, dosages, and the like for non-human animals. Administration of the combinations described in this application can be effected orally or parenterally.

The amount of each component of the combination described in this application is administered together or in combination with another agent such that an effective dose is received. The daily dose orally administered to an animal is typically between about 0.01 and about 1,000mg/kg body weight, for example between about 0.01 and about 300mg/kg body weight or between about 0.01 and about 100mg/kg body weight or between about 0.01 and about 50mg/kg body weight, or between about 0.01 and about 25mg/kg, or between about 0.01 and about 10mg/kg, or between about 0.01 and about 5 mg/kg.

Conveniently, the compound (or combination) of the invention may be carried in drinking water such that a therapeutic dose of the compound is taken with a daily supply of water. The compounds may be metered directly into drinking water, preferably in the form of a liquid, water-soluble concentrate (such as an aqueous solution of a water-soluble salt).

The compounds (or combinations) of the invention may also conveniently be added directly to the feed, either as such or in the form of an animal feed supplement (also known as a premix or concentrate). Premixes or concentrates of the compounds in excipients, diluents or carriers are more commonly used to contain the agent in the feed. Suitable excipients, diluents or carriers are liquids or solids as desired, such as water, various meals (such as alfalfa meal, soybean meal, cottonseed oil meal, linseed oil meal, corn cobs and corn meal, molasses, urea, bone meal) and mineral mixtures (such as are commonly used in poultry feed). Particularly effective excipients, diluents or carriers are the respective animal feed itself; i.e. a small portion of the feed. The carrier facilitates uniform distribution of the compound in the final feed of the premix with which it is mixed. The compounds are preferably mixed well in the premix and subsequently in the feed. In this aspect, the compound can be dispersed or dissolved in a suitable oily medium (such as soybean oil, corn oil, cottonseed oil, and the like) or in a volatile organic solvent, and then combined with the carrier. It will be appreciated that the proportion of the compound in the concentrate can vary widely, as the amount of compound in the final feed can be adjusted by mixing the premix with the feed in appropriate proportions to achieve the desired level of compound.

High titer concentrates can be mixed by feed manufacturers with proteinaceous carriers (such as the soybean oil meal and other meals described above) to produce concentrated supplements that are suitable for direct feeding to animals. In such cases, the animal is permitted to consume the usual diet. Alternatively, such concentrated supplements may be added directly to the feed to produce a nutritionally balanced final feed containing therapeutically effective levels of the compounds of the invention. The mixture is mixed thoroughly in standard procedures (such as in a double barrel mixer) to ensure homogeneity.

If the supplement is used as a topping dressing for feed, it likewise helps to ensure the uniformity of distribution of the compound on the topping of the prepared feed.

Drinking water and feed effective in increasing lean meat deposition and improving lean meat to fat ratio are typically prepared by mixing the compounds of the invention with a sufficient amount of animal feed to provide about 10 in the feed or water-3To about 500ppm of compound.

Preferred medicated pig, cattle, sheep and goat feed will typically contain from about 1 to about 400 grams of a compound (or combination) of the invention per ton of feed, with the optimum amount for the animal often being from about 50 to about 300 grams per ton of feed.

Preferred poultry and domestic pet feeds often contain from about 1 to about 400 grams, and preferably from about 10 to about 400 grams of a compound (or combination) of the invention per ton of feed.

The compounds (or combinations) of the present invention for parenteral administration to an animal can be prepared in the form of pastes or pellets and are often administered as implants under the skin of the head or ear of the animal where increased lean meat deposition and improved lean meat to fat ratios are sought.

Paste formulations may be prepared by dispersing the drug in a pharmaceutically acceptable oil such as peanut oil, sesame oil, corn oil or the like.

The pellets containing a therapeutically effective amount of each component of the combination described herein may be added with diluents (such as carbowax, carnauba wax, and the like) and lubricants (such as magnesium or calcium stearate) to improve the pelletizing process.

Of course, it is understood that more than one pill can be administered to an animal to achieve a desired dosage level that provides increased lean meat deposition and an improved desired ratio of lean meat to fat. Moreover, implantation may be performed periodically during treatment of the animal to maintain appropriate drug levels in the animal.

The present invention has several advantageous veterinary features. For pet owners or veterinarians desiring to increase lean meat and/or trim undesirable fat in a pet animal, the present invention provides a way to accomplish this desire. For poultry, beef cattle and swine breeders, the method of the present invention yields leaner animals that win higher sales prices from the meat industry.

Examples

Unless otherwise noted, starting materials are typically obtained from commercial sources such as Aldrich Chemicals Co. (Milwaukee, WI), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Ltd. (Cornwall, England), and Tyger Scientific (Princeton, NJ). Certain common abbreviations and acronyms have been used and may include: AcOH (acetic acid), DBU (1, 8-diazabicyclo [5.4.0 ]]Undec-7-ene), CDI (1,1 ' -carbonyldiimidazole), DCM (dichloromethane), DEA (diethylamine), DIPEA (N, N-diisopropylethylamine), DMAP (4-dimethylaminopyridine), DMF (N, N ' -dimethylformamide), DMSO (dimethyl sulfoxide), EDCI (N- (3-dimethylaminopropyl) -N ' -ethylcarbodiimide), Et2O (diethyl ether), EtOAc (ethyl acetate), EtOH (ethanol), G or G (G), HATU (2- (1H-7-azabenzotriazol-1-yl) -1,1,3, 3-tetramethyluronium hexafluorophosphate), HBTU (O-benzotriazol-1-yl-N, N, N ', N' -tetramethyluronium hexafluorophosphate), HOBT (1-hydroxybenzotriazole), H or H (hr), IPA (isopropanol), KHMDS (potassium hexamethyldisilazide), MeOH (methanol), L or L (liter), mL (mL) MTBE (tert-butyl methyl ether), mg (mg), NaBH (OAc) 3(sodium triacetoxyborohydride), NaHMDS (sodium hexamethyldisilazide), NMP (N-methylpyrrolidone), RH (relative humidity), RT or RT (room temperature, which is the same as ambient temperature (about 20 to 25 ℃)), SEM ([2- (trimethyl-25 ℃))Silicon-based) ethoxy groups]Methyl), TEA (triethylamine), TFA (trifluoroacetic acid), THF (tetrahydrofuran), and T3P (propanephosphonic anhydride).

1The H Nuclear Magnetic Resonance (NMR) spectrum is consistent with the proposed structure in all cases. Characteristic chemical shifts (. delta.) as compared to residual proton signal in deuterated solvents (CHCl at 7.27 ppm)3(ii) a CD at 3.31ppm2HOD) is given in parts per million and reported using the conventional abbreviations for the names of the major peaks: e.g., s, singlet; d, double peak; t, triplet; q, quartet; m, multiplet; br, broad peak.

ssNMR means solid state NMR.

PXRD means powder X-ray diffraction.

The term "substantially the same" when used to describe an X-ray powder diffraction pattern is intended to include patterns in which the peaks are within a standard deviation of +/-0.2 DEG 2 theta.

As used herein, the term "substantially pure" with respect to a particular crystalline form means that the crystalline form includes less than 10%, preferably less than 5%, preferably less than 3%, preferably less than 1% by weight of any other physical form of the same compound.

The reaction is carried out in air or, when oxygen-or moisture-sensitive reagents or intermediates are used, under an inert atmosphere (nitrogen or argon). Where appropriate, the reaction apparatus is dried under dynamic vacuum using a heat gun and anhydrous solvent (Sure-Seal from Aldrich Chemical Company, Milwaukee, Wisconsin)TMProduct or DriSolv from EMD Chemicals, Gibbstown, NJTMProduct). Commercial solvents and reagents were used without further purification. When indicated, the reaction was heated by microwave irradiation using Biotage Initiator or Personal Chemistry emerys Optimizer microwaves. The progress of the reaction is monitored analytically using Thin Layer Chromatography (TLC), liquid chromatography-mass spectrometry (LCMS), High Performance Liquid Chromatography (HPLC) and/or gas chromatography-mass spectrometry (GCMS). TLC was carried out on pre-coated silica gel plates with fluorescent display (excitation wavelength of 254 nm) and under UV light and/or with I2、KMnO-4、CoCl2Phosphomolybdic acid and/or ammonium cerium molybdate stain development. LCMS data were acquired on an Agilent 1100 Series instrument with a Leap Technologies autosampler, a Gemini C18 column, MeCN/water gradient, and TFA, formic acid, or ammonium hydroxide modifiers. Column eluents were analyzed using a Waters ZQ mass spectrometer scanning from 100 to 1200Da using both positive and negative ion models. Other similar instruments are also used. HPLC data were acquired on an Agilent 1100 Series instrument using a Gemini or XBridge C18 column, MeCN/water gradient, and TFA or ammonium hydroxide modifier. GCMS data were obtained using a Hewlett Packard 6890 oven with HP 6890 syringe, HP-1 column (12m × 0.2mm × 0.33 μm), and helium carrier gas. Samples were analysed on a HP 5973 mass selective detector scanning from 50 to 550Da using electron liberation. Purification was performed by Medium Performance Liquid Chromatography (MPLC) using an Isco CombiFlash company, AnaLogix IntelliFlash 280, Biotage SP1 or Biotage Isolera One instrument and a pre-filled Isco redisep or Biotage Snap silica cartridge. Using a Berger or Thar instrument; ChiralPAK-AD, -AS, -IC, Chiralcel-OD or-OJ column; and CO with MeOH, EtOH, iPrOH or MeCN 2Mixtures (alone or with TFA or iPrNH)2Modification) by chiral Supercritical Fluid Chromatography (SFC). UV detection was used to trigger fraction collection.

Mass spectral data were reported by LCMS analysis. Mass Spectrometry (MS) was performed by Atmospheric Pressure Chemical Ionization (APCI), electrospray ionization (ESI), electron impact ionization (EI), or Electron Scattering (ES) free sources. Mass spectral data are reported by liquid chromatography-mass spectrometry (LCMS), Atmospheric Pressure Chemical Ionization (APCI), or gas chromatography-mass spectrometry (GCMS) instruments. Symbol denotes a chlorine isotope pattern observed in the mass spectrum. Proton nuclear magnetic spectrometry (1H NMR) chemical shifts are given in parts per million from the low magnetic field of tetramethylsilane and recorded on a 300, 400, 500, or 600MHz Varian spectrometer. Chemical shifts are expressed in parts per million (ppm, δ) of the reference deuterated solvent residual peak. The peak shape is illustrated below: s, singlet; d, double peak; t, triplet; q, quartet; quin, quintuple peak; m, multiplet; br s, broad singlet; app, appearance. Analytical SFC data were acquired on a Berger analyzer instrument as described above. Optically activeThe degree data were obtained on a Perkinelmer type 343 polarimeter using a 1dm cell. Silica gel chromatography is mainly performed using a high pressure Biotage or ISCO system using columns pre-filled by various market suppliers including Biotage and ISCO. Micro-analysis was performed by Quantitative Technologies inc. and was within 0.4% of the calculated values.

Unless otherwise noted, chemical reactions were carried out at room/ambient temperature (about 23 degrees celsius).

The compounds and intermediates described below are generally named using the nomenclature convention provided by ChemBioDraw Ultra, version 12.0 (Cambridge soft corp., Cambridge, Massachusetts). The nomenclature convention provided in ChemBioDraw Ultra, 12.0 edition is well known to those skilled in the art, and it is believed that the nomenclature convention provided in ChemBioDraw Ultra, 12.0 edition generally complies with IUPAC (international union of pure and applied chemistry) recommendations of organic chemistry nomenclature and CAS indexing rules. Unless otherwise noted, all reactants were obtained on the market, without further purification, or prepared using methods known in the literature.

Chiral separation methods are used to separate enantiomers or diastereomers of some of the intermediates during the preparation of the compounds of the invention. When chiral separation is performed, the separated enantiomers are designated ENT-1 or ENT-2 (or DIAST-1 or DIAST-2) depending on the order in which they elute. In some embodiments, the enantiomer designated ENT-1 or ENT-2 may be used as a starting material to prepare other enantiomers or diastereomers. In such cases, the resulting enantiomers as prepared are designated, depending on their starting materials, ENT-X1 and ENT-X2, respectively; likewise, the prepared diastereomers are designated DIAST-X1 and DIAST-X2, respectively, depending on their starting materials. In syntheses using a variety of intermediates, the DIAST-Y and DIAST-Z nomenclature is similarly used.

With respect to compounds having two chiral centers, stereoisomers at each stereocenter are separated at different times. The designation of ENT-1 or ENT-2 (or DIAST-1 or DIAST-2) for intermediates or examples refers to the elution sequence in which the separation is performed at this step. It is understood that when a stereoisomer of a chiral center is isolated in a compound having two or more centers, the isolated enantiomers are diastereomers of each other. By way of example, but not limitation, examples 15 and 16 have two chiral centers. When intermediate C36 was separated into ENT-1 (giving intermediate P17) and ENT-2 (giving intermediate P18), the chiral center of the cyclopropyl moiety was separated. C70 was then prepared using P18, which had a mixture of one enriched stereoisomer on the cyclopropyl chiral carbon and a stereoisomer on the dioxolane carbon. C70 was then separated into DIAST-Y1 on the dioxolane carbon to give intermediate C71, and DIAST-Y2 on the dioxolane carbon to give intermediate C72, both of which were enriched in a single stereoisomer. Example 15 was then prepared using C71, the name of which was identified as 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate [ from P18 via C71 ]. In the preparation, after the mixture is subjected to a separation procedure, a chiral center is identified near that center as "abs," with the understanding that the separated enantiomers may not be pure enantiomers. Typically, the enriched enantiomers at each chiral center constitute > 90% of the separated material. Preferably, the enriched enantiomers at each center make up > 98% of the mixture.

In some embodiments, the optical rotation of an enantiomer is measured using a polarimeter. According to their observed optical data (or their specific optical data), the clockwise rotating enantiomer is labeled as the (+) -enantiomer and the counterclockwise rotating enantiomer is labeled as the (-) -enantiomer. Racemic compounds are shown by unshown or illustrated stereochemistry, or by the paracomponents (+/-); in the latter case, the stereochemistry indicated represents the relative (rather than absolute) configuration of the substituents of the compound.

The reaction continued through the detectable intermediate is typically followed by LCMS and allowed to proceed to full conversion before the addition of subsequent reagents. Reaction conditions (reaction time) may be varied with respect to the synthesis of procedures with reference to other examples or methodsAnd temperature). The reaction is usually followed by thin layer chromatography or mass spectrometry, and if appropriate by work-up. Purification may vary from experiment to experiment: the solvent and solvent ratio for the eluent/gradient are generally selected to provide the appropriate RfOr a retention time. All starting materials in the preparations and examples are commercially available or may be prepared by methods known in the art or as described herein.

The terms "concentration", "evaporation" and "concentration in vacuo" refer to the removal of solvent on a rotary evaporator under reduced pressure at a bath temperature below 60 ℃. The abbreviations "min" and "h" represent "minutes" and "hours", respectively. The term "TLC" refers to thin layer chromatography, "room or ambient temperature" refers to a temperature between 18 to 25 ℃, "GCMS" refers to gas chromatography-mass spectrometry, "LCMS" refers to liquid chromatography-mass spectrometry, "UPLC" refers to ultra performance liquid chromatography, and "HPLC" refers to high pressure liquid chromatography, "SFC" refers to supercritical fluid chromatography.

The hydrogenation can be carried out at a specified temperature under pressurized hydrogen in a Parr shaker or in Thales-nano H-Cube flow hydrogenation devices under total hydrogen and a flow rate of between 1 and 2 mL/min.

HPLC, UPLC, LCMS, GCMS and SFC retention times were measured using the methods noted in the procedures.

Preparation examples and examples of intermediates

Preparation P1

4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester (P1)

Step 1.2 Synthesis of bromo-6- [ (4-chloro-2-fluorophenyl) (hydroxy) methyl ] phenol (C1)

This experiment was performed in two batches of the same scale. N-butyllithium (2.5M solution in hexanes; 32.8mL, 82.0mmol) was slowly added to 1-bromo-4-chloro-2-fluorobenzene (17.2g, 82.1 mmo) in diethyl ether (100mL) l) at-70 ℃ while maintaining the temperature of the reaction mixture below-60 ℃. After the reaction mixture was stirred at-70 ℃ for 20 minutes, a solution of 3-bromo-2-hydroxybenzaldehyde (5.5g, 27mmol) in diethyl ether (100mL) was added slowly while maintaining the reaction temperature below-60 ℃. After stirring at-70 ℃ for an additional 1h, the reaction was quenched at-70 ℃ with the addition of aqueous ammonium chloride (50mL) and the resulting mixture was diluted with water (100 mL). At this point the two batches were combined and extracted with ethyl acetate (400 mL); the organic layer was washed with saturated aqueous solution of sodium chloride (200mL), dried over sodium sulfate, filtered and concentrated in vacuo. Chromatography on silica gel (gradient: 0% to 7% ethyl acetate in petroleum ether) afforded C1 as a white solid. Combined yields: 15.7g, 47.4mmol, 88%.1H NMR (400MHz, chloroform-d) δ 7.44(dd, J ═ 8.0,1.5Hz,1H),7.37(dd, J ═ 8.1,8.1Hz,1H),7.15(br dd, J ═ 8.5,2.1Hz,1H),7.12-7.05(m,2H),6.80(dd, J ═ 7.8,7.8Hz,1H),6.78(s,1H),6.31(d, J ═ 4.8Hz,1H),3.02(br d, J ═ 4.9Hz, 1H).

Step 2.4 Synthesis of bromo-2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxole (C2)

A solution of sodium periodate (25.4g, 119mmol) in water (105mL) was added to a solution of C1(15.7g, 47.4mmol) in methanol (450mL) and the reaction mixture was stirred at 30 ℃ for 16 h, then concentrated in vacuo. After the residue was diluted with dichloromethane (500mL), it was washed with water (500 mL). The dichloromethane solution was then dried over sodium sulfate, filtered and concentrated in vacuo. Purification via silica gel chromatography (eluent: petroleum ether) gave C2 as a white solid. Yield: 10.0g, 30.3mmol, 64%. From experiments carried out in the same manner but on a smaller scale, the following were obtained 1H NMR data.1H NMR(400MHz,DMSO-d6)δ7.67-7.61(m,2H),7.50(s,1H),7.43(br dd,J=8,2Hz,1H),7.09(dd,J=8.3,1.1Hz,1H),7.01(dd,J=7.9,1.1Hz,1H),6.86(dd,J=8.1,8.1Hz,1H)。

Step 3.4 Synthesis of tert-butyl- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] -3, 6-dihydropyridine-1 (2H) -carboxylate (C3)

Will contain in 1, 4-diC2(8.00g, 24.3mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (9.01g, 29.1mmol), sodium carbonate (5.15g, 48.6mmol) in dioxane (80mL) and water (32mL), and [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride [ Pd (dppf) Cl2;888mg,1.21mmol]The reaction flask of the suspension was evacuated and charged with nitrogen. This evacuation cycle was repeated twice, and then the reaction mixture was stirred at 90 ℃ for 16 hours. After removal of the solvent in vacuo, the residue was partitioned between ethyl acetate (200mL) and water (200 mL). The organic layer was washed with saturated aqueous solution of sodium chloride (100mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. Chromatography on silica gel (gradient: 0% to 4.3% ethyl acetate in petroleum ether) afforded the product, which was combined with material from an analogous reaction using C2(2.00g, 6.07mmol) to give C3 as a pale yellow gum. Combined yields: 10.3g, 23.8mmol, 78%.1H NMR (400MHz, chloroform-d) δ 7.53(dd, J ═ 8.3,7.8Hz,1H),7.23-7.16(m,3H),6.88-6.83(m,2H),6.81-6.76(m,1H),6.34-6.28(br m,1H),4.10-4.05(m,2H),3.61(br dd, J ═ 6,5Hz,2H),2.59-2.50(br m,2H),1.48(s, 9H).

Step 4.4 Synthesis of tert-butyl 4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (P1)

A solution of C3(10.3g, 23.8mmol) and tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst; 1.54g, 1.66mmol) in methanol (100mL) was stirred at 50 ℃ under hydrogen (45psi) for 18 h. The reaction mixture was then filtered through a pad of celite, the filtrate was concentrated under reduced pressure and subjected to silica gel chromatography (gradient: 0% to 9% ethyl acetate in petroleum ether). The resulting material was combined with material from a similar reaction using C3(1.67g, 3.87mmol) to give P1 as a colourless gum. Combined yields: 10.3g, 23.7mmol, 86%. LCMS M/z 456.1 [. M + Na ]+]。1H NMR (400MHz, chloroform-d) δ 7.52(dd, J ═ 8.5,7.6Hz,1H),7.23-7.17(m,2H),7.16(s,1H),6.83(dd, J ═ 7.8,7.8Hz,1H),6.78-6.69(m,2H),4.35-4.10(br m,2H),2.89-2.71(m,3H),1.89-1.77(m,2H),1.77-1.63(m,2H),1.47(s, 9H).

Preparation P2

4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester (P2)

Step 1.4 Synthesis of bromo-2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole (C4)

1- (4-chloro-2-fluorophenyl) ethanone (316g, 1.83mol) and p-toluenesulfonic acid (6.02g, 35.0mmol) were added to a solution of 3-bromobenzene-1, 2-diol (330g, 1.75mol) in toluene (1.5L). The reaction apparatus was equipped with Dean-Stark trap and the reaction mixture was heated at 140 ℃ for 60 hours whereupon the solution was concentrated in vacuo and purified using silica gel chromatography (eluent: petroleum ether); c4 was obtained as a mixture of yellow oil and solid. Yield: 158g, 460mmol, 26%.1H NMR (400MHz, chloroform-d): δ 7.54(dd, J ═ 8.4,8.4Hz,1H),7.17-7.10(m,2H),6.95(dd, J ═ 7.9,1.4Hz,1H),6.75(dd, a component of ABX pattern, J ═ 7.8,1.4Hz,1H),6.70(dd, a component of ABX pattern, J ═ 7.9,7.9Hz,1H),2.11(d, J ═ 1.1Hz, 3H).

Step 2.4 Synthesis of tert-butyl- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -3, 6-dihydropyridine-1 (2H) -carboxylate (C5)

Tert-butyl 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylate (62g, 200mmol) and sodium carbonate (100g, 940mmol) were added to a solution of C4(58.0g, 169mmol) in 1, 4-dioxane (600 mL). Adding [1, 1' -bis (diphenylphosphino) ferrocene ]After palladium (II) dichloride (6.0g, 8.2mmol), the reaction mixture was heated to 90 ℃ and stirred for 16 hours. Water (500mL) was then added and the resulting mixture was extracted with ethyl acetate (2X 500 mL). The combined organic layers were washed with saturated aqueous sodium chloride (2 × 500mL), dried over sodium sulfate, filtered and concentrated in vacuo. Chromatography on silica gel (gradient: 0% to 9% ethyl acetate in petroleum ether) afforded C5 as a yellow oil. Yield:56.0g,126mmol,75%。1h NMR (400MHz, chloroform-d) δ 7.50(dd, J ═ 8.2,8.2Hz,1H),7.17-7.09(m,2H),6.83-6.77(m,2H),6.74(dd, a component of ABX pattern, J ═ 5.4,3.6Hz,1H),6.39-6.33(br m,1H),4.14-4.08(m,2H),3.70-3.56(m,2H),2.66-2.45(m,2H),2.07(d, J ═ 1.1Hz,3H),1.50(s, 9H).

Step 3.4 Synthesis of tert-butyl- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (P2)

Tris (triphenylphosphine) rhodium (I) chloride (wilkinson's catalyst; 8.10g, 8.75mmol) was added to a solution of C5(56.0g, 126mmol) in methanol (200mL) and the reaction mixture was heated to 50 ℃ under hydrogen (45psi) for 18 h. It was then cooled to 25 ℃ and filtered through celite. The filtrate was concentrated in vacuo and purified twice using silica gel chromatography (first column-gradient: 0% to 9% ethyl acetate in petroleum ether; second column-gradient: 0% to 2% ethyl acetate in petroleum ether) to afford P2 as a yellow solid. Yield: 37.0g, 82.6mmol, 66%. LCMS M/z 392.1 solid-solid [ (M-2-methylpropan-1-ene) + H ]+1H NMR (400MHz, chloroform-d) δ 7.51(dd, J ═ 8.3,8.0Hz,1H),7.17-7.09(m,2H),6.77(dd, components of ABC pattern, J ═ 7.8,7.8Hz,1H),6.70(dd, components of ABC pattern, J ═ 7.7,1.3Hz,1H),6.66(dd, components of ABC pattern, J ═ 7.8,1.3Hz,1H),4.37-4.13(br m,2H),2.92-2.73(m,3H),2.05(d, J ═ 1.1Hz,3H),1.90-1.63(m,4H),1.49(s, 9H).

Preparation P3

4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, P-toluenesulfonate (P3)

Step 1 isolation of tert-butyl 4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (C6) and tert-butyl 4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (C7)

P2(75.2g, 168mmol) was separated into its component enantiomers by SFC (supercritical fluid chromatography) [ column: 5 μm for Chiral Technologies Chiralpak AD-H; mobile phase: 4:1 carbon dioxide/(2-propanol containing 0.2% of 1-aminopropan-2-ol) ]. The first eluting compound is designated C6 and the second eluting enantiomer is designated C7. The absolute configuration indicated was assigned based on single crystal X-ray structural determination with C8, which C8 is derived from C6 (see below).

C6-yield: 38.0g, 84.8mmol, 50%. Retention time 3.64 min [ column: chiral Technologies Chiral pak AD-H, 4.6X 250mm, 5 μm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol containing 0.2% 1-aminopropan-2-ol; gradient: 5% of B over 1.00 min followed by 5% to 60% of B over 8.00 min; flow rate: 3.0 ml/min; back pressure: 120 bar ].

C7-yield: 36.8g, 82.2mmol, 49%. Retention time 4.19 minutes (conditions for analytical SFC are the same as for C6).

Step 2.Synthesis of 4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, P-toluenesulfonate (P3)

A solution of C7(1.62g, 3.62mmol) in ethyl acetate (36mL) was treated with p-toluenesulfonic acid monohydrate (791mg, 4.16mmol) and heated at 45 ℃. After 23 hours, the reaction mixture was allowed to cool to room temperature and the solid was collected via filtration. It was washed with a mixture of ethyl acetate and heptane (1: 1; 2X 15mL) to give P3 as a white solid. Yield: 1.37g, 2.63mmol, 73%. LCMS M/z 348.1 [. M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.53(v br s,1H),8.29(v br s,1H),7.65-7.55(m,2H),7.47(d,J=8.1Hz,2H),7.35(dd,J=8.4,2.0Hz,1H),7.11(d,J=7.8Hz,2H),6.88-6.81(m,2H),6.75-6.68(m,1H),3.42-3.33(m,2H),3.11-2.93(m,3H),2.29(s,3H),2.03(s,3H),1.98-1.82(m,4H)。

Conversion of C6 to 4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, mesylate (C8) for determination of absolute stereochemistry

P-toluenesulfonic acid (377mg, 2.19mmol) was added to a solution of C6(490mg, 1.09mmol) in ethyl acetate (5.5mL) and the reaction mixture was stirred at room temperature overnight. After dilution with additional ethyl acetate, the reaction mixture was washed sequentially with aqueous sodium bicarbonate, water and saturated aqueous sodium chloride, dried over sodium sulfate, filtered and concentrated in vacuo. Yield: 375mg, 1.08mmol, 99%.1H NMR (400MHz, methanol-d)4)δ7.59(dd,J=8.3.8.3Hz,1H),7.27(dd,J=10.9,2.0Hz,1H),7.20(br dd,J=8.4,2.1Hz,1H),6.81-6.75(m,1H),6.74-6.67(m,2H),3.18-3.09(m,2H),2.88-2.77(m,1H),2.77-2.67(m,2H),2.02(d,J=0.7Hz,3H),1.85-1.73(m,4H)。

A0.1M solution of the free base (4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine) in ethyl acetate was prepared and subjected to salt screening. Only the formation of the mesylate salt is illustrated herein. A mixture of methanesulfonic acid (25. mu.L, 39. mu. mol) and substrate solution (0.1M; 0.25mL, 25. mu. mol) was stirred overnight. Sufficient methanol was then added to dissolve the solids present, and ethyl acetate (3mL) was added. The resulting solution was allowed to slowly evaporate without stirring to give crystals of C8; one of these crystals was used for the single crystal X-ray structural determination described below.

Single crystal X-ray texture determination of C8

Single crystal X-ray analysis

Data collection was performed on a Bruker D8 Quest diffractometer at room temperature. Data collection is represented by ω and Scanning the image.

By using SHELX software package to obtain orthorhombic space group P212121The intrinsic phasing of (a) resolves the structure. The structure is then refined with a full matrix least squares method. All non-hydrogen atoms were found and refined using anisotropic shift parameters.

Formation of the mesylate salt was confirmed via N1_ H1X _ O4 proton transfer.

The hydrogen atoms located on nitrogen and oxygen were found from the fourier difference plot and refined with the constrained distance. The remaining hydrogen atoms are positioned at the calculated positions and allowed to sit on their carrier atoms. The final refinement includes the anisotropic displacement parameters of all hydrogen atoms.

Analysis of the absolute structure using likelihood methods (hoft), 2008) was performed using platon (spek). The results show that the absolute structure has been correctly assigned; the method calculates the probability of the structure being correct to be 100%. The Hoff parameter is reported as 0.02 with an esd of 0.0012, and the Parson's parameter is reported as 0.07 with an esd of 0.009. The absolute configuration at C7 is identified as (R).

The asymmetric unit consists of one molecule of the protonated free base of C8 and one molecule of deprotonated methanesulfonic acid. The final R index was 4.6%. The resulting difference fourier reveals the electron density without loss or dislocation.

The relevant crystal, data collection and refinement information is summarized in table a. The atomic coordinates, bond lengths, bond angles, and displacement parameters are listed in tables B through D.

Software and reference documents

SHELXTL,Version 5.1,Bruker AXS,1997。

PLATON.A.L.Spek,J.Appl.Cryst.2003,36,7-13。

MERCURY,C.F.Macrae,P.R.Edington,P.McCabe,E.Pidcock,G.P.Shields,R.Taylor,M.Towler,and J.van de Streek,J.Appl.Cryst.2006,39,453-457。

OLEX2, o.v.dolomanov, l.j.bourhis, r.j.gildea, j.a.k.howard, and h.puschmann, j.appl.cryst.2009, 42, 339-.

R.w.w.hooft, l.h.straver, and a.l.spek, j.appl.cryst.2008, 41, 96-103.

H.D.Flack,Acta Cryst.1983,A39,867-881。

Crystal data and structure refinement of table a.c 8.

C8 atomic coordinates (x 10)4) And equivalent isotropic displacement parameterU (eq) is defined as orthogonal UijOne third of the trace of the tensor.

C. C8 bond lengthAngle of harmony key [ ° ]]

For generating symmetric transformations of equivalent atoms.

An anisotropic displacement of a. c8 in table d.a. Pueraria brevis 25976The anisotropic displacement factor takes the form: -2 pi2[h2 a*2U11+...+2 h k a b U12]。

Preparation of P3, di-P-toluoyl-L-tartrate

4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, di-P-toluoyl-L-tartrate (P3, di-P-toluoyl-L-tartrate)

A solution of C13, free base (519mg, 1.49mmol) and di-p-toluoyl-L-tartaric acid (278mg, 0.719mmol) in acetonitrile (7.5mL) was stirred at 50 ℃ for 1.5 h. The mixture was allowed to cool to room temperature at 0.2 ℃/min. After 15 hours at room temperature, the mixture was heated to 65 ℃ and charged with acetonitrile (15 mL). The mixture was allowed to cool to room temperature at 0.2 ℃/min. After 15 hours at room temperature, the mixture was heated to 54 ℃. After 3 hours, the solid was collected by filtration and dried in a vacuum oven at 35 ℃ under nitrogen to give P3, di-P-toluoyl-L-tartrate (217mg, 0.296mmol, 20%, 82% ee) as a white solid.

A solution of P3, di-P-toluoyl-L-tartrate (217mg, 0.296mmol, 82% ee) in acetonitrile (8.0mL) at 50 ℃ was allowed to cool to room temperature at 0.2 ℃/min. After 15 hours, the solid was collected by filtration and dried in a vacuum oven at 35 ℃ under nitrogen to give P3, di-P-toluoyl-L-tartrate (190mg, 0.259mmol, 88%, 88% ee) as a white solid. LCMS M/z 348.1 [. M + H ]]+1H NMR(400MHz,DMSO-d6) δ 8.9-8.5(br s,2H),7.79(d, J ═ 8.1Hz,4H),7.64-7.54(m,2H),7.34(dd, J ═ 8.4,2.1Hz,1H),7.26(d, J ═ 8.0Hz,4H),6.87-6.78(m,2H),6.69(dd, J ═ 6.7,2.5Hz,1H),5.58(s,2H),3.37-3.28(m,2H, assumed values; partially masked by water peaks), 3.05-2.89(m,3H),2.33(s,6H),2.02(s,3H),1.92-1.80(m, 4H). Retention time: peak 1(4.97 min, minor peak) and peak 2(5.31 min, major peak) { column: chiralpak IC-U3.0X 50mm, 1.6 μm; mobile phase A: carbon dioxide; mobile phase B: isopropylamine 0.1% in methanol; gradient: 10% of B over 5.00 minutes followed by 45% of B over 0.6 minutes; flow rate: 1.7 ml/min; back pressure: 130 bar }.

Preparation P4

4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester (P4)

A suspension of P2(2.00g, 4.46mmol), zinc cyanide (734mg, 6.25mol), zinc (70.1mg, 1.07mmol), 1' -bis (diphenylphosphino) ferrocene (dppf; 198mg, 0.357mmol) and tris (dibenzylideneacetone) dipalladium (0) (164mg, 0.179mmol) in N, N-dimethylacetamide (20mL) was stirred at 120 ℃ for 16 h and then filtered. The filtrate was mixed with water (50mL) and extracted with ethyl acetate (3 × 50 mL); the combined organic layers were then washed sequentially with water (30mL) and saturated aqueous sodium chloride (20mL) and concentrated in vacuo. Chromatography on silica gel (gradient: 0% to 30% ethyl acetate in petroleum ether) afforded a solid, which was treated with acetonitrile (15mL) and water (15mL) and subjected to lyophilization. This gave P4 as a pale yellow solid. Yield: 1.17g, 2.67mmol, 60%. LCMS M/z 461.3[ M + Na ]+]。1H NMR (400MHz, chloroform-d) 7.71(dd, J ═ 7.7,7.6Hz,1H),7.45(dd, J ═ 8.0,1.6Hz,1H),7.42(dd, J ═ 10.0,1.5Hz,1H),6.79(dd, a component of the ABC pattern, J ═ 7.7,7.6Hz,1H),6.72(dd, a component of the ABC pattern, J ═ 7.8,1.3Hz,1H),6.68(dd, a component of the ABC pattern, J ═ 7.8,1.3Hz,1H),4.37-4.14(br m,2H),2.91-2.73(m,3H),2.07(d, J ═ 1.1, 3H),1.89-1.62(m, 1H), 1.49H (s, 49).

Preparation examples P5 and P6

4-bromo-2-phenyl-1, 3-benzodioxole, ENT-1(P5) and 4-bromo-2-phenyl-1, 3-benzodioxole, ENT-2(P6)

Step 1.2 Synthesis of bromo-6- [ hydroxy (phenyl) methyl ] phenol (C9)

Phenyllithium (1.9M solution in 1-butoxybutane; 78.5mL, 149mmol) was slowly added to a-70 ℃ solution of 3-bromo-2-hydroxybenzaldehyde (10.0g, 49.7mmol) in tetrahydrofuran (70mL) at a rate to maintain the reaction temperature below-60 ℃. The resulting suspension was stirred at-70 ℃ for 1 hour, then allowed to warm to room temperature overnight, and then poured into 0 ℃ aqueous ammonium chloride (30 mL). The mixture was extracted with ethyl acetate (3 × 30mL) and the combined organic layers were washed with saturated aqueous sodium chloride (30mL), dried over sodium sulfate, filtered and concentrated in vacuo. Chromatography on silica gel (gradient: 0% to 5% ethyl acetate in petroleum ether) afforded C9 as a yellow solid. Yield: 6.11g, 21.9mmol, 44%.1H NMR (400MHz, chloroform-d) δ 7.45-7.28(m,6H),7.22-7.18(m,1H),7.06(br d, J ═ 7.7Hz,1H),6.77(dd, J ═ 7.9,7.8Hz,1H),6.06(br s,1H),2.89(br s, 1H).

Step 2.4 Synthesis of bromo-2-phenyl-1, 3-benzodioxole (C10)

A solution of sodium periodate (11.7g, 54.7mmol) in water (175mL) was added to a solution of C9(6.11g, 21.9mmol) in methanol (370 mL). The reaction mixture was stirred at 30 ℃ for 40 hours, whereupon most of the methanol was removed via concentration in vacuo. The resulting mixture was extracted with dichloromethane (5 × 100mL) and the combined organic layers were washed sequentially with aqueous sodium sulfite (100mL) and saturated aqueous sodium chloride (100mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. Chromatography on silica gel (eluent: petroleum ether) gave C10 as a colorless oil. Yield: 4.50g, 16.2mmol, 74%. LCMS M/z 278.5 (bromine isotope pattern observed) [ M + H [)]+1H NMR (400MHz, chloroform-d) δ 7.62-7.57(m,2H),7.49-7.43(m,3H),7.04(s,1H),7.00(dd, J ═ 8.0,1.4Hz,1H),6.79(dd, component of ABX pattern, J ═ 7.8,1.4Hz,1H),6.75(dd, component of ABX pattern, J ═ 7.9,7.8Hz, 1H).

Step 3.4-bromo-2-phenyl-1, 3-benzodioxole, ENT-1(P5) and 4-bromo-2-phenyl-1, 3-benzodioxole, ENT-2(P6) separations

Enantiomer containing C10(5.00g, 18.0mmol) was purified using SFC [ column: chiral Technologies Chiral cel OD, 10 μm; mobile phase: 3:1 carbon dioxide/(methanol containing 0.1% ammonium hydroxide) ]. The first eluting enantiomer was designated ENT-1(P5) and the second eluting enantiomer was designated ENT-2 (P6); both were obtained as yellow oil.

Yield of P5: 2.20g, 7.94mmol, 44%. LCMS M/z 277.0 (bromine isotope pattern observed) [ M + H [)]+1H NMR (400MHz, chloroform-d) δ 7.63-7.55(m,2H),7.51-7.42(m,3H),7.04(s,1H),7.00(dd, J ═ 8.0,1.3Hz,1H),6.80(dd, component of ABX pattern, J ═ 7.8,1.4Hz,1H),6.75(dd, component of ABX pattern, J ═ 7.9,7.8Hz, 1H). Retention time 3.28 min (column: Chiral Technologies ChiralCel OD-H, 4.6 x 150mm, 5 μm; mobile phase a: carbon dioxide; mobile phase B: methanol with 0.05% diethylamine; gradient: 5% to 40% B over 5.5 min; flow rate: 2.5 ml/min).

Yield of P6: 2.00g, 7.22mmol, 40%. LCMS M/z 276.9 (bromine isotope pattern observed) [ M + H [)]+1H NMR (400MHz, chloroform-d) δ 7.63-7.55(m,2H),7.50-7.42(m,3H),7.04(s,1H),7.00(dd, J ═ 8.0,1.4Hz,1H),6.80(dd, component of ABX pattern, J ═ 7.8,1.4Hz,1H),6.75(dd, component of ABX pattern, J ═ 7.9,7.9Hz, 1H). Retention time 3.73 minutes (analytical conditions were the same as those for P5).

Preparation P7

4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester (P7)

Step 1.2- (4-bromo-2-methyl-1, 3-benzodioxol-2-yl) -5-chloropyridine (C11) Synthesis

5-chloro-2-ethynylpyridine (1.80g, 13.1mmol), 3-bromobenzene-1, 2-diol (2.47g, 13.1mmol) and deca in toluene (25mL)A mixture of tricarbonyltriruthenium (167mg, 0.261mmol) was degassed for 1 minute and then heated at 100 ℃ for 16 hours. The reaction mixture was diluted with ethyl acetate (30mL) and filtered through a pad of celite; the filtrate was concentrated in vacuo and purified using silica gel chromatography (gradient: 0% to 1% ethyl acetate in petroleum ether) to afford C11 as a yellow oil. Yield: 1.73g, 5.30mmol, 40%. LCMS M/z 325.6 (bromine-chlorine isotope pattern observed) [ M + H [ ]]+1H NMR (400MHz, chloroform-d) δ 8.63(dd, J ═ 2.4,0.7Hz,1H),7.71(dd, component of ABX pattern, J ═ 8.4,2.4Hz,1H),7.60(dd, component of ABX pattern, J ═ 8.4,0.7Hz,1H),6.97(dd, J ═ 8.0,1.4Hz,1H),6.76(dd, component of ABX pattern, J ═ 7.8,1.4Hz,1H),6.72(dd, component of ABX pattern, J ═ 8.0,7.8Hz,1H),2.10(s, 3H).

Step 2.4 Synthesis of tert-butyl- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] -3, 6-dihydropyridine-1 (2H) -carboxylate (C12)

Reacting [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (388mg, 0.530mmol) was added to a suspension of C11(1.73g, 5.30mmol), tert-butyl 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylate (1.64g, 5.30mmol) and cesium carbonate (5.18g, 15.9mmol) in 1, 4-dioxane (35mL) and water (6 mL). The reaction mixture was stirred at 90 ℃ for 4 hours, whereupon it was diluted with ethyl acetate (30mL) and water (5 mL). The organic layer was concentrated in vacuo and the residue was subjected to silica gel chromatography (gradient: 0% to 5% ethyl acetate in petroleum ether) to afford C12 as a yellow gum. Yield: 1.85g, 4.31mmol, 81%. LCMS M/z 451.0 [. M + Na ] +]。1H NMR (400MHz, chloroform-d) δ 8.62(dd, J ═ 2.5,0.8Hz,1H),7.69(dd, a component of ABX pattern, J ═ 8.4,2.4Hz,1H),7.57(dd, a component of ABX pattern, J ═ 8.4,0.8Hz,1H),6.84-6.79(m,2H),6.78-6.73(m,1H),6.39-6.33(br m,1H),4.13-4.07(m,2H),3.68-3.58(m,2H),2.60-2.51(br m,2H),2.07(s,3H),1.49(s, 9H).

Step 3.4 Synthesis of tert-butyl- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (P7)

Methanol (1)00mL) of C12(2.61g, 6.08mmol) and tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst; 563mg, 0.608mmol) was degassed under vacuum and then purged with hydrogen; this evacuation-purge cycle was carried out three times in total. The reaction mixture was then stirred at 60 ℃ under hydrogen (50psi) for 16 hours, whereupon it was filtered. The filtrate was concentrated in vacuo and the residue was purified using silica gel chromatography (gradient: 0% to 10% ethyl acetate in petroleum ether); the resulting material was combined with a similar hydrogenation of the material obtained with C12 (110mg, 0.256mmol) to give P7 as a pale yellow gum. Combined yields: 2.05g, 4.76mmol, 75%. LCMS M/z 431.3 [. M + H ]]+1H NMR (400MHz, chloroform-d) δ 8.62(d, J ═ 2.3Hz,1H),7.69(dd, a component of the ABX pattern, J ═ 8.4,2.4Hz,1H),7.57(d, half of the AB quartet, J ═ 8.4Hz,1H),6.79(dd, a component of the ABC pattern, J ═ 7.8,7.7Hz,1H),6.72(dd, a component of the ABC pattern, J ═ 7.8,1.3Hz,1H),6.68(br d, a component of the ABC pattern, J ═ 7.9Hz,1H),4.32-4.12(br m,2H),2.91-2.73(m,3H),2.05(s,3H),1.90-1.62(m,4H),1.48(s, 9H).

Preparation examples P8 and P9

4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester, ENT-1(P8) and 4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylic acid tert-butyl ester, ENT-2(P9)

Separation of P7(500mg, 1.16mmol) into its component enantiomers was performed using SFC { column: phenomenex Lux Amylose-1, 5 μm; mobile phase: 9:1 carbon dioxide/[ 2-propanol with 0.2% (7M ammonia in methanol) ] }. The first eluting enantiomer was designated ENT-1(P8) and the second eluting enantiomer was designated ENT-2 (P9).

Yield of P8: 228mg, 0.529mmol, 46%. Retention time 4.00 minutes { column: phenomenex Lux Amylose-1, 4.6X 250mm, 5 μm; mobile phase A: carbon dioxide; mobile phase B: [ 2-propanol containing 0.2% (7M ammonia in methanol) ]; gradient: 5% of B over 1.00 min followed by 5% to 60% of B over 8.00 min; flow rate: 3.0 ml/min; back pressure: 120 bar }.

Yield of P9: 229mg, 0.531mmol, 46%. Retention time 4.50 minutes (analytical conditions were the same as those for P8).

Preparation P10

{4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetic acid (P10)

Step 1.4 Synthesis of- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, p-toluenesulfonate (C13)

A solution of P2(5.0g, 11mmol) and P-toluenesulfonic acid (4.81g, 27.9mmol) in ethyl acetate (100mL) was stirred at 60 ℃ for 2 hours, whereupon it was concentrated in vacuo to give C13 as a yellow gum. This material was taken directly to the next step. LCMS M/z347.9 [. M + H ]]+

Step 2 Synthesis of Ethyl {4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetate (C14)

Potassium carbonate (7.71g, 55.8mmol) and ethyl bromoacetate (1.86g, 11.2mmol) were added to a solution of C13 (from the previous step;. ltoreq.11 mmol) in acetonitrile (150mL) and the reaction mixture was stirred at 55 ℃ for 16 h. It was then filtered, the filtrate concentrated in vacuo and purified using silica gel chromatography (gradient: 0% to 30% ethyl acetate in petroleum ether) to give C14 as a yellow gum. To be provided with1H NMR analysis showed that this was not completely pure material. Yield: 3.57g, 8.23mmol, 75% in 2 steps.1H NMR (400MHz, chloroform-d); peak of C14 only: δ 7.52(dd, J ═ 8.4,8.0Hz,1H),7.17-7.07(m,2H),6.77(dd, a component of the ABC pattern, J ═ 7.8,7.8Hz,1H),6.72-6.67(m,2H),4.21(q, J ═ 7.1Hz,2H),3.27(s,2H),3.07(m,2H),2.70(tt, J ═ 12.1,3.8Hz,1H),2.35(ddd, J ═ 11.5,11.5,2.7Hz, 1H) ,2H),2.04(d,J=1.1Hz,3H),2.02-1.76(m,4H),1.29(t,J=7.1Hz,3H)。

Step 3 Synthesis of {4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetic acid (P10)

A solution of C14(3.57g, 8.23mmol) and aqueous sodium hydroxide (3M; 13.7mL, 41.1mmol) in a mixture of methanol (80mL) and tetrahydrofuran (40mL) was stirred at 25 ℃ for 16 h. After removal of the solvent in vacuo, the aqueous residue was acidified to pH 7 with the addition of 1M hydrochloric acid and then extracted with a mixture of dichloromethane and methanol (10:1, 2X 100 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure to give P10 as a yellow solid. Yield: 2.95g, 7.27mmol, 88%. LCMS M/z 406.2 [. sup. + H ]]+1H NMR (400MHz, methanol-d)4) δ 7.61(dd, J ═ 8.3,8.3Hz,1H),7.29(dd, J ═ 10.9,2.0Hz,1H),7.22(ddd, J ═ 8.4,2.0,0.8Hz,1H),6.82(dd, a component of the ABC pattern, J ═ 8.3,7.1Hz,1H),6.78-6.72(m,2H),3.65-3.54(br m,2H),3.51(s,2H),3.04-2.88(m,3H),2.23-2.07(m,2H),2.07-1.93(m,2H),2.04(d, J ═ 1.1Hz, 3H).

Preparation P11

2- (chloromethyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P11)

Step 1.3- [ (2-methoxyethyl) amino ] -4-nitrobenzoic acid methyl ester (C15) Synthesis

Triethylamine (40.7g, 402mmol, 55.8mL) was added to a colorless solution of methyl 3-fluoro-4-nitrobenzoate (50g, 250mmol) in tetrahydrofuran (400mL), followed by dropwise addition of 2-methoxyethylamine (30.2g, 402mmol) in tetrahydrofuran (100mL) at room temperature. The resulting yellow solution was stirred at 55 ℃ for 18 hours. The solution was cooled to room temperature and concentrated under reduced pressure to remove tetrahydrofuran. The resulting yellow solid was dissolved in ethyl acetate (800mL) and washed with saturated aqueous ammonium chloride solution (250 mL). The aqueous phase was separated and extracted with ethyl acetate (200 mL). Combining the organic layersWashed with saturated aqueous solution of sodium chloride (3 × 250mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give C15(60.2g, 94%) as a yellow solid.1H NMR (600MHz, chloroform-d) Δ 8.23(d,1H),8.17(br s,1H),7.58(d,1H),7.25(dd,1H),3.95(s,3H),3.69-3.73(m,2H),3.56(m,2H),3.45(s, 3H); LCMS M/z 255.4[ M + H ]]+

Step 2.4 Synthesis of methyl 4-amino-3- [ (2-methoxyethyl) amino ] benzoate (C16)

Pd/C (10g, 94mmol) was added to a solution of C15(30g, 118mmol) in methanol (500 mL). The reaction was stirred at room temperature under 15psi of hydrogen for 18 hours. The black suspension was filtered through celite and the filter cake was washed with methanol (500 mL). The combined filtrates were concentrated in vacuo to give C16(26.5g, quantitative) as a brown oil, which solidified upon standing. 1H NMR (400MHz, chloroform-d) Δ 7.48(dd,1H),7.36(d,1H),6.69(d,1H),3.87(s,3H),3.77(br s,2H),3.68(t,2H),3.41(s,3H),3.32(t, 2H); LCMS M/z 224.7[ M + H ]]+

Step 3.2- (chloromethyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P11) Synthesis

2-chloro-1, 1, 1-trimethoxyethane (3.31mL, 24.6mmol) was added to a solution of C16(5.00g, 22.3mmol) in tetrahydrofuran (100mL), followed by p-toluenesulfonic acid monohydrate (84.8mg, 0.446 mmol). The reaction mixture was heated at 45 ℃ for 5 hours, whereupon it was concentrated in vacuo; the residual oil was dissolved in ethyl acetate (10mL) and heated until a solution was formed. This was stirred slowly while cooling to room temperature overnight. The precipitate was collected via filtration and washed with heptane to afford P11 as a gray solid. Yield: 5.73g, 20.3mmol, 91%.1H NMR (600MHz, chloroform-d) δ 8.12(br s,1H),8.01(br d, J ═ 8.6Hz,1H),7.79(d, J ═ 8.4Hz,1H),4.96(s,2H),4.52(t, J ═ 5.1Hz,2H),3.96(s,3H),3.74(t, J ═ 5.1Hz,2H),3.28(s, 3H).

Step 4.2- (chloromethyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, Synthesis of the hydrochloride salt (P11, HCl salt)

A solution of C16(5.0g, 24mmol) in 1, 4-dioxane (100mL) was heated To 100 ℃, a solution of chloroacetic anhydride (4.1g, 24.5mmol) in 1, 4-dioxane (60mL) was added over a period of 10 hours via an addition funnel and the reaction mixture was stirred at 100 ℃ for an additional 12 hours. On alternate days, the reaction was cooled to room temperature and the 1, 4-dioxane was removed under reduced pressure. The crude reaction mixture was dissolved in ethyl acetate and washed with saturated aqueous sodium bicarbonate. The ethyl acetate layer was separated, dried over sodium sulfate and filtered. A 4M hydrogen chloride solution in 1, 4-dioxane (1.1 eq) was added to a constantly stirring ethyl acetate solution. The hydrochloride salt of P11 precipitated as a pale yellow solid. The suspension was stirred for 1 hour, then the hydrochloride salt of P11 was collected by filtration to give a yellow solid (6.1g, 86%).1H NMR(600MHz,CD3OD)δ8.64(s,1H),8.30(d,1H),7.92(d,1H),5.32(s,2H),4.84(m,2H),3.99(s,3H),3.83(t,2H),3.31(s,3H)。LCMS m/z 283.2[M+H]+

Preparation P12

1- (2-methoxyethyl) -2- (piperazin-1-ylmethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P12)

Step 1.2 Synthesis of methyl- { [4- (tert-butoxycarbonyl) piperazin-1-yl ] methyl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C17)

Compound P11(1.59g, 5.62mmol) was added to a 15 ℃ mixture of tert-butyl piperazine-1-carboxylate (1.00g, 5.37mmol) and potassium carbonate (2.97g, 21.5mmol) in acetonitrile (15mL) and the reaction mixture was stirred at 55 ℃ for 12 hours. Then combined with a similar reaction using P11 and tert-butyl piperazine-1-carboxylate (200mg, 1.07mmol) and the mixture was filtered. After concentration of the filtrate in vacuo, the residue was purified via chromatography on silica gel (gradient: 0% to 60% ethyl acetate in petroleum ether) to give C17 as a light yellow solid. Combined yields: 2.30g, 5.32mmol, 83%. LCMS M/z 433.0[ M + H ] ]+1H NMR (400MHz, chloroform-d) δ 8.12(d, J ═ 1.5Hz,1H),7.96(dd, J ═ 8.4,1.5Hz,1H),7.73(d, J ═ 8.5Hz,1H),4.58(t, J ═ 5).4Hz,2H),3.95(s,3H),3.89(s,2H),3.73(t,J=5.4Hz,2H),3.46-3.37(br m,4H),3.28(s,3H),2.54-2.44(br m,4H),1.45(s,9H)。

Step 2.1 Synthesis of methyl 1- (2-methoxyethyl) -2- (piperazin-1-ylmethyl) -1H-benzimidazole-6-carboxylate (P12)

A solution of hydrogen chloride in ethyl acetate (20mL) was added to a solution of C17(2.30g, 5.32mmol) in dichloromethane (80 mL). The reaction mixture was stirred at 20 ℃ for 2 hours and then concentrated in vacuo. The residue was diluted with water (20mL), adjusted to pH 9 to 10 by the addition of saturated aqueous sodium bicarbonate solution and extracted with a mixture of ethyl acetate and methanol (10:1, 15 × 50 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo to afford P12 as a light yellow solid. Yield: 1.68g, 5.05mmol, 95%. LCMS M/z332.8[ M + H ]]+1H NMR (400MHz, chloroform-d) δ 8.13(br s,1H),7.96(br d, J ═ 8.5Hz,1H),7.72(d, J ═ 8.5Hz,1H),4.59(t, J ═ 5.5Hz,2H),3.95(s,3H),3.86(s,2H),3.75(t, J ═ 5.5Hz,2H),3.29(s,3H),2.87(t, J ═ 4.8Hz,4H),2.50(br m, 4H).

Preparation P13

6-bromo-2- (chloromethyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine (P13)

Step 1.5 Synthesis of bromo-N- (2-methoxyethyl) -2-nitropyridin-3-amine (C18)

A solution of 5-bromo-3-fluoro-2-nitropyridine (400mg, 1.81mmol) and 2-methoxyethylamine (408mg, 5.43mmol) in tetrahydrofuran (10mL) was stirred at 25 ℃ for 2 hours, whereupon it was diluted with ethyl acetate (100mL) and washed with water (50 mL). The organic layer was washed with saturated aqueous solution of sodium chloride (50mL), dried over magnesium sulfate, filtered and concentrated to give C18 as a yellow solid. Yield: 430mg, 1.56mmol, 86%.

Step 2.5 bromo-N3Synthesis of- (2-methoxyethyl) pyridine-2, 3-diamine (C19)

Mixing methanol (10mL) with water (2mL)A solution of C18(430mg, 1.56mmol), ammonium chloride (833mg, 15.6mmol) and iron powder (870mg, 15.6mmol) in this material was stirred at 80 ℃ for 30 minutes. The resulting suspension was poured into water (50mL) and extracted with ethyl acetate (2 × 50 mL); the combined organic layers were dried over magnesium sulfate, filtered and concentrated to give C19 as a brown solid. Yield: 350mg, 1.42mmol, 91%.1H NMR (400MHz, chloroform-d) δ 7.63(d, J ═ 2.1Hz,1H),6.88(d, J ═ 2.0Hz,1H),4.33-4.19(br s,2H),3.65(dd, J ═ 5.6,4.6Hz,2H),3.40(s,3H),3.22(br t, J ═ 5Hz, 2H).

Step 3.6 Synthesis of bromo-2- (chloromethyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine (P13)

A solution of C19(400mg, 1.63mmol) in 1, 4-dioxane (8mL) was treated with chloroacetyl chloride (0.284mL, 3.57mmol) and stirred at room temperature until LCMS analysis showed complete conversion of C19 to the intermediate amide. After removal of 1, 4-dioxane in vacuo, the residue was dissolved in trifluoroacetic acid (8mL) and heated at 80 ℃ for 18 hours, whereupon the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting oil was dissolved in ethyl acetate (50mL) and neutralized with the addition of saturated aqueous sodium bicarbonate solution. The aqueous layer was extracted with ethyl acetate (20mL) and the combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. Silica gel chromatography (gradient: 0% to 80% ethyl acetate in heptane) afforded P13 as a solid. Yield: 176mg, 0.578mmol, 35%. LCMS M/z 306.1 (bromine-chlorine isotope pattern observed) [ M + H [ ]]+1H NMR (600MHz, chloroform-d) δ 8.58(br s,1H),7.89(br s,1H),4.92(s,2H),4.44(t, J ═ 5.0Hz,2H),3.71(t, J ═ 5.0Hz,2H),3.28(s, 3H).

Preparation P14

2- { [4- (2, 3-dihydroxyphenyl) piperidin-1-yl ] methyl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P14)

Step 1 Synthesis of [ (3-bromobenzene-1, 2-diyl) bis (oxymethynedioxyethane-2, 1-diyl) ] bis (trimethylsilane) (C20)

The reaction was carried out in two batches of the same scale. N, N-diisopropylethylamine (37.8mL, 217mmol) was added dropwise to a solution of 3-bromobenzene-1, 2-diol (10.0g, 52.9mmol) in tetrahydrofuran (300 mL). After the mixture was stirred at 20 ℃ for 10 minutes, [2- (chloromethoxy) ethyl group was added dropwise over 5 minutes](trimethyl) silane (19.2mL, 108mmol) and stirring was continued at room temperature (18 ℃ C.) for 16 h. N, N-diisopropylethylamine (27.6mL, 158mmol) was added, followed by dropwise addition of [2- (chloromethoxy) ethyl at room temperature (18 deg.C)](trimethyl) silane (14.0mL, 79.1 mmol). After a further 2.5 hours at room temperature, the reaction mixture was filtered and the filtrate was concentrated in vacuo. At this point, the crude products from the two batches were combined and purified using silica gel chromatography (gradient: 0% to 7% ethyl acetate in petroleum ether) to give C20 as a colorless oil. To be provided with1H NMR analysis showed that this was not completely pure material. Combined yields: 22.9g, 50.9mmol, 48%.1H NMR (400MHz, chloroform-d), only C20 peak: δ 7.19(dd, J ═ 8.1,1.5Hz,1H),7.12(dd, J ═ 8.3,1.4Hz,1H),6.90(dd, J ═ 8.2.8.2Hz,1H),5.26-5.19(m,4H),4.00-3.92(m,2H),3.80-3.73(m,2H),1.00-0.91(m,4H),0.03(s,9H),0.00(s, 9H).

Step 2.4 Synthesis of tert-butyl 4- (2, 3-bis { [2- (trimethylsilyl) ethoxy ] methoxy } phenyl) -3, 6-dihydropyridine-1 (2H) -carboxylate (C21)

C20(6.11g, 13.6mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (5.04g, 16.3mmol), aqueous sodium carbonate solution (1M; 40.8mL, 40.8mmol) and [1, 1' -bis (diphenylphosphino) ferrocene in 1, 4-dioxane (100mL) were combined]The reaction vessel for the suspension of palladium (II) dichloride (497mg, 0.679mmol) was evacuated and charged with nitrogen. This vacuum cycle was repeated twice, then the reaction mixture was stirred at 85 ℃ for 16 hours, whereupon the reaction mixture was diluted with water (40mL) and extracted with ethyl acetate (3X 150 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. Purification via silica gel chromatography (gradient: 0% to 8% methanol in dichloromethane) gave C21 as a yellow oil. Yield: 5.47g,9.91mmol,73%。1H NMR (600MHz, chloroform-d) δ 7.10(br d, J ═ 8.2Hz,1H),6.98(dd, J ═ 7.9,7.9Hz,1H),6.81(br d, J ═ 7.7Hz,1H),5.79(br s,1H),5.23(s,2H),5.07(s,2H),4.03(br s,2H),3.83-3.74(m,4H),3.59(br s,2H),2.52(br s,2H),1.49(s,9H),1.01-0.89(m,4H),0.01(s, 9H).

Step 3.4 Synthesis of tert-butyl 4- (2, 3-bis { [2- (trimethylsilyl) ethoxy ] methoxy } phenyl) piperidine-1-carboxylate (C22)

A solution of C21(12.5g, 22.6mmol) in methanol (300mL) was treated with 10% palladium on carbon (2.94g, 2.76mmol) and hydrogenated at 40psi and 25 deg.C for 16 h. Analysis at this point by LCMS showed conversion to product: LCMS M/z 576.0[ M + Na ]+]. After the reaction mixture was filtered and the filter cake was washed with methanol (2 × 100mL), the combined filtrates were concentrated in vacuo to afford C22 as a colorless oil. Yield: 11.2g, 20.1mmol, 89%.1H NMR (400MHz, chloroform-d) δ 7.05-6.97(m,2H),6.83(dd, J ═ 6.9,2.5Hz,1H),5.22(s,2H),5.13(s,2H),4.38-4.10(br m,2H),3.90-3.82(m,2H),3.81-3.73(m,2H),3.22(tt, J ═ 12.2,3.5Hz,1H),2.79(br dd, J ═ 12.8,12.8Hz,2H),1.78(br d, J ═ 13Hz,2H),1.65-1.52(m,2H),1.48(s,9H),1.04-0.91(m,4H),0.03(s,9H),0.00(s,9H), 9H (m, 9H).

Step 4.4 Synthesis of 4- (2, 3-bis { [2- (trimethylsilyl) ethoxy ] methoxy } phenyl) piperidine (C23)

2, 6-lutidine (2.39g, 22.3mmol) was added to a room temperature (15 deg.C) solution of C22(7.23g, 13.0mmol) in dichloromethane (90mL), followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (3.80g, 17.1 mmol). The reaction mixture was stirred at 15 ℃ for 16 h, whereupon additional 2, 6-lutidine (909mg, 8.48mmol) and trimethylsilyl trifluoromethanesulfonate (1.45g, 6.52mmol) were added. After stirring at room temperature (15 ℃) for a further 5 hours, LCMS analysis of the reaction mixture showed the presence of the product: LCMS M/z 454.1[ M + H ] ]+. The reaction mixture was concentrated in vacuo and the residue was washed sequentially with aqueous ammonium chloride (3 × 100mL) and saturated aqueous sodium chloride (100mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give C23(6.6g) as a brown oil. The substance is taken directly to the nextIn the step (2).

Step 5 Synthesis of methyl 2- { [4- (2, 3-bis { [2- (trimethylsilyl) ethoxy ] methoxy } phenyl) piperidin-1-yl ] methyl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C24)

P11(3.08g, 10.9mmol) was added to a solution of C23 (from previous step; 6.6g,. ltoreq.13 mmol) in acetonitrile (150mL), followed by potassium carbonate (10.1g,. ltoreq.13 mmol) and the reaction mixture stirred at room temperature (15 ℃ C.) for 16 h. Analysis at this point by LCMS showed the presence of product: LCMS M/z 700.2[ M + H ]]+. The reaction mixture was filtered and the filtrate was concentrated in vacuo; purification via silica gel chromatography (gradient: 34% to 56% ethyl acetate in petroleum ether) gave C24 as a yellow oil. Yield: 5.4g, 7.7mmol, obtained in 2 steps to 59%.1H NMR (400MHz, chloroform-d) δ 8.16-8.12(m,1H),7.96(dd, J ═ 8.5,1.5Hz,1H),7.73(d, J ═ 8.5Hz,1H),7.04-6.96(m,2H),6.86(dd, J ═ 6.7,2.6Hz,1H),5.21(s,2H),5.12(s,2H),4.63(t, J ═ 5.5Hz,2H),3.95(s,3H),3.93-3.83(m,4H),3.80-3.72(m,4H),3.31(s,3H),3.17-3.06(m,1H),2.99(br d, J ═ 11.2Hz,2H), 2.35-2.35 (m, 2.81H), 2.81 (m,1H), 0.01-7H (H), 1H (1H), 0.05-7H, 1H), 1H (H), 1H, 9H, 1H), and a peak.

Step 6.2 Synthesis of methyl- { [4- (2, 3-dihydroxyphenyl) piperidin-1-yl ] methyl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (P14)

A solution of hydrogen chloride in 1, 4-dioxane (4M; 96mL, 384mmol) was added to a room temperature (18 ℃) solution of C24(6.40g, 9.14mmol) in 1, 4-dioxane (120 mL). After the addition was complete, the reaction mixture was stirred at room temperature (18 ℃) for 16 h, combined with a similar reaction using C24(1.00g, 1.43mmol) and concentrated in vacuo. The residue was treated with a mixture of dichloromethane and methanol (20:1, 150mL) and stirred at room temperature (18 ℃) for 1 hour, whereupon the solid was collected via filtration (4.85 g). This material was treated with water (100mL) and the mixture was adjusted to pH 7 to 8 with the addition of aqueous sodium bicarbonate, stirred at room temperature (18 ℃) for 30 minutes and filtered. The filter cake was washed with water (2 × 20mL), then mixed with methanol (100mL) and concentrated in vacuo. Mixing the obtained material with stoneOil ether (100mL) and stirred at room temperature (18 ℃ C.) for 30 minutes. After filtration, the filter cake was mixed with toluene (30mL) and concentrated in vacuo to give P14 as a gray solid. Combined yields: 2.92g, 6.64mmol, 63%. LCMS M/z440.1[ M + H ] ]+1H NMR(400MHz,DMSO-d6)δ8.21(d,J=1.6Hz,1H),7.81(dd,J=8.5,1.6Hz,1H),7.66(d,J=8.5Hz,1H),6.64-6.51(m,3H),4.63(t,J=5.3Hz,2H),3.88(s,3H),3.84(s,2H),3.75(t,J=5.3Hz,2H),3.22(s,3H),2.97-2.78(m,3H),2.18(br dd,J=11,11Hz,2H),1.75-1.64(m,2H),1.64-1.49(m,2H)。

Preparation P15

2- (chloromethyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester (P15)

The entire procedure was carried out on a large scale. The reactor is evacuated to-0.08 to-0.05 MPa, usually before the reaction and after the addition of reagents, and then filled with nitrogen to normal pressure. This process is typically repeated 3 times and then the oxygen level is assessed to ensure that it is ≦ 1.0%. The mixture used in the extraction and washing process of the organic layer is typically stirred for 15 to 60 minutes, then allowed to settle over 15 to 60 minutes, and the layers are separated.

Step 1 Synthesis of (2S) -2- [ (benzyloxy) methyl ] oxetane (C25)

This reaction was carried out in three batches of about the same scale. 2-methylpropan-2-ol (774.7kg) was charged to a 2000L glass lined reactor. Potassium tert-butoxide (157.3kg, 1402mol) was added via the solid addition funnel and the mixture was stirred for 30 minutes. Trimethyl sulphoxide iodide (308.2kg, 1400mol) is then added in the same manner and the reaction mixture is heated at 55 to 65 ℃ for 2 to 3 hours, whereupon (2S) -2- [ (benzyloxy) methyl is added at a rate of 5 to 20 kg/hour]Ethylene oxide (92.1kg, 561 mol). After the reaction mixture was maintained at 55 ℃ to 65 ℃ for 25 hours, it was cooled to 25 ℃ to 35 ℃ and filtered through celite (18.4 kg). The filter cake was washed with tert-butyl methyl ether (3X 340kg) and the mixture was washed The filtrate was transferred to a 5000L reactor, treated with pure water (921kg) and stirred at 15 to 30 ℃ for 15 to 30 minutes. The organic layer was then washed twice with a solution of sodium chloride (230.4kg) in pure water (920.5kg) and concentrated at ≦ 45 ℃ under reduced pressure (≦ -0.08 MPa). N-heptane (187kg) was added and the resulting mixture was concentrated at 45 ℃ or less under reduced pressure (-0.08 MPa or less); the organic phase was purified using silica gel chromatography (280kg) with sodium chloride (18.5kg) on the top of the column. The crude material was loaded onto the column using n-heptane (513kg) and then eluted with a mixture of n-heptane (688.7kg) and ethyl acetate (64.4 kg). The three batches were combined to give 85% pure C25(189.7kg, 906mmol, 54%) as a pale yellow oil.1H NMR (400MHz, chloroform-d), only C25 peak: δ 7.40-7.32(m,4H),7.32-7.27(m,1H),4.98(dddd, J ═ 8.1,6.7,4.9,3.7Hz,1H),4.72-4.55(m,4H),3.67(dd, a component of ABX pattern, J ═ 11.0,4.9Hz,1H),3.62(dd, a component of ABX pattern, J ═ 11.0,3.7Hz,1H),2.72-2.53(m, 2H).

Step 2 Synthesis of (2S) -oxetan-2-ylmethanol (C26)

10% palladium on carbon (30.7kg) was added via an addition funnel to a 10 ℃ to 30 ℃ solution of 85% pure C25 (from previous step; 185.3kg, 884.8mol) in tetrahydrofuran (1270kg) in a 3000L stainless steel autoclave reactor. The addition funnel was rinsed with pure water and tetrahydrofuran (143kg), and the rinse was added to the reaction mixture. After the reactor contents were purged with nitrogen, they were similarly purged with hydrogen, increased in pressure to 0.3 to 0.5MPa, and then vented to 0.05 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.3 to 0.4 MPa. The reaction mixture was then heated to 35 ℃ to 45 ℃. After the hydrogen pressure was maintained at 0.3 to 0.5MPa for a period of 13 hours, the mixture was vented to 0.05MPa, purged five times with nitrogen, thereby increasing the pressure to 0.15 to 0.2MPa, and then vented to 0.05 MPa. After the mixture was cooled to 10 to 25 ℃, it was filtered and the reactor was rinsed with tetrahydrofuran (2 x 321 kg). Soaking the filter cake twice with the flushing liquid, and then filtering; concentration under reduced pressure (. ltoreq. -0.06MPa) at ≦ 40 deg.C gave C26(62.2kg, 706mol, 80%) in tetrahydrofuran (251 kg).

Step 3.4 Synthesis of (2S) -oxetan-2-ylmethyl 4-methylbenzenesulfonate (C27)

4- (dimethylamino) pyridine (17.5kg, 143mol) was added to a 10 ℃ to 25 ℃ solution of C26 (from previous step; 62.2kg, 706mol) in tetrahydrofuran (251kg) and triethylamine (92.7kg, 916mol) in dichloromethane (1240 kg). After 30 minutes p-toluenesulfonyl chloride (174.8kg, 916.9mol) was added portionwise at intervals of 20 to 40 minutes and the reaction mixture was stirred at 15 to 25 ℃ for 16 hours and 20 minutes. Adding pure water (190 kg); after stirring, the organic layer was washed with an aqueous sodium bicarbonate solution (prepared using 53.8kg of sodium bicarbonate and 622kg of pure water), and then with an aqueous ammonium chloride solution (prepared using 230kg of ammonium chloride and 624kg of pure water). After a final wash with pure water (311kg), the organic layer was filtered through a stainless steel Nutsche filter pre-filled with silica gel (60.2 kg). The filter cake was soaked in dichloromethane (311kg) for 20 minutes and then filtered; the combined filtrates are concentrated under reduced pressure (less than or equal to-0.05 MPa) and at a temperature of less than or equal to 40 ℃ until 330 to 400L remains. Tetrahydrofuran (311kg) was then added at 15 ℃ to 30 ℃ and the mixture was concentrated in the same way to a final volume of 330 to 400L. The addition of tetrahydrofuran and concentration was repeated to a further volume of 330 to 400L to give a solution of C27(167.6kg, 692mmol, 98%) in tetrahydrofuran (251.8kg) as a pale yellow oil. 1H NMR (400MHz, chloroform-d), only C27 peak: δ 7.81(d, J ═ 8.4Hz,2H),7.34(d, J ═ 8.1Hz,2H),4.91(ddt, J ═ 8.0,6.7,3.9Hz,1H),4.62-4.55(m,1H),4.53-4.45(m,1H),4.14(d, J ═ 3.9Hz,2H),2.75-2.63(m,1H),2.60-2.49(m,1H),2.44(s, 3H).

Step 4 Synthesis of (2S) -2- (azidomethyl) oxetane (C28)

N, N-dimethylformamide (473kg), sodium azide (34.7kg, 534mol) and potassium iodide (5.2kg, 31mol) were combined in a 3000L glass-lined reactor at 10 ℃ to 25 ℃. After addition of C27(83.5kg, 344.6mol) in tetrahydrofuran (125.4kg), the reaction mixture was heated to 55 ℃ to 65 ℃ for 17 hours and 40 minutes, whereupon it was cooled to 25 ℃ to 35 ℃ and nitrogen was bubbled through from the bottom valve for 15 minutes. Tert-butyl methyl ether (623kg) and pure water (840kg) were then added, and the resulting aqueous layer was extracted twice with tert-butyl methyl ether (312kg and 294 kg). The combined organic layers were washed with pure water (2 × 419kg) while maintaining the temperature at 10 ℃ to 25 ℃, yielding C28(31.2kg, 276mol, 80%) in the above organic layer solution (1236.8 kg).

Step 5.1 Synthesis of- [ (2S) -oxetan-2-yl ] methylamine (C29)

10% palladium on carbon (3.7kg) was added via addition funnel to C28 in tetrahydrofuran (328kg) in a 3000L stainless steel autoclave reactor [ from previous step; 1264kg (31.1kg of C28, 275mol) ]From 10 ℃ to 30 ℃. The addition funnel was rinsed with tetrahydrofuran (32kg) and the rinse was added to the reaction mixture. After the reactor contents were purged with nitrogen, they were similarly purged with hydrogen, increased in pressure to 0.05 to 0.15MPa, and then vented to 0.03 to 0.04 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.05 to 0.07 MPa. The reaction temperature was increased to 25 to 33 ℃, and the hydrogen pressure was maintained at 0.05 to 0.15MPa for 22 hours while replacing the hydrogen gas every 3 to 5 hours. The mixture was then purged five times with nitrogen, thereby increasing the pressure to 0.15 to 0.2MPa, and then vented to 0.05 MPa. After filtration, the reactor was rinsed with tetrahydrofuran (92kg and 93kg) and the filter cake was then soaked. The combined filtrates were concentrated under reduced pressure (. ltoreq. -0.07MPa) and. ltoreq.45 ℃ to give C29(18.0kg, 207mol, 75%) in tetrahydrofuran (57.8 kg).1H NMR(400MHz,DMSO-d6) Only C29 peak: δ 4.62(ddt, J ═ 7.6,6.6,5.1Hz,1H),4.49(ddd, J ═ 8.6,7.3,5.6Hz,1H),4.37(dt, J ═ 9.1,5.9Hz,1H),2.69(d, J ═ 5.1Hz,2H),2.55-2.49(m,1H),2.39(m, 1H).

Step 6.Synthesis of methyl 4-nitro-3- { [ (2S) -oxetan-2-ylmethyl ] amino } benzoate (C30)

Potassium carbonate (58.1kg, 420mol) was added to a solution of methyl 3-fluoro-4-nitrobenzoate (54.8kg, 275mol) in tetrahydrofuran (148kg) in a 100L glass-lined reactor and the mixture was stirred for 10 minutes. A solution of C29(29.3kg, 336mol) in tetrahydrofuran (212.9kg) was added and the reaction mixture was stirred at 20 ℃ to 30 ℃ for 12 hours whereupon ethyl acetate (151kg) was added and the mixture was filtered through silica gel (29 kg). The filter cake was washed with ethyl acetate (150kg and 151kg)Washed and the combined filtrates are concentrated under reduced pressure (< 0.08MPa) and ≤ 45 deg.C to a volume of 222-281L. After the mixture was cooled to 10 ℃ to 30 ℃, n-heptane (189kg) was added, stirring was carried out for 20 minutes and the mixture was concentrated under reduced pressure (. ltoreq. -0.08MPa) and. ltoreq.45 ℃ to a volume of 222L. N-heptane (181kg) was added to the mixture at a reference rate of 100 to 300 kg/hour and stirring was continued for 20 minutes. The mixture was sampled until 5% tetrahydrofuran remained and 10% to 13% ethyl acetate remained. The mixture is heated to 40 ℃ to 45 ℃ and stirred for 1 hour, whereupon it is cooled to 15 ℃ to 25 ℃ at a rate of 5 ℃ to 10 ℃ per hour, and then stirred for 1 hour at 15 ℃ to 25 ℃. Filtration using a stainless steel centrifuge to provide a filter cake, which was rinsed with a mixture of ethyl acetate (5.0kg) and n-heptane (34kg), then stirred with tetrahydrofuran (724kg) at 10 ℃ to 30 ℃ for 15 minutes; a yellow solid consisting mostly of C30(57.3kg, 210mol, 76%) was provided by filtration. 1H NMR(400MHz,DMSO-d6)8.34(t,J=5.8Hz,1H),8.14(d,J=8.9Hz,1H),7.63(d,J=1.7Hz,1H),7.13(dd,J=8.9,1.8Hz,1H),4.99(dddd,J=7.7,6.7,5.3,4.1Hz,1H),4.55(ddd,J=8.6,7.3,5.8Hz,1H),4.43(dt,J=9.1,6.0Hz,1H),3.87(s,3H),3.67-3.61(m,2H),2.67(dddd,J=11.1,8.6,7.7,6.2Hz,1H),2.57-2.47(m,1H)。

Step 7.2 Synthesis of methyl 2- (chloromethyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (P15)

A solution of C30 (from previous step; 51.8kg, 190mol) in tetrahydrofuran (678kg) in a 3000L autoclave reactor was treated with 10% palladium on carbon (5.2kg) at 10 ℃ to 30 ℃. The addition tube was rinsed with tetrahydrofuran (46kg) and the rinse was added to the reaction mixture. After the reactor contents were purged with nitrogen, they were similarly purged with hydrogen, increased in pressure to 0.1 to 0.2MPa, and then vented to 0.02 to 0.05 MPa. This hydrogen purge was repeated 5 times, whereupon the hydrogen pressure was increased to 0.1 to 0.25 MPa. The reaction mixture was stirred at 20 to 30 ℃ and the mixture was purged three times with nitrogen and then five times with hydrogen every 2 to 3 hours; after each final hydrogen change, the hydrogen pressure was increased to 0.1 to 0.25 MPa. After a total reaction time of 11.25 hours, the reaction mixture was vented to normal pressure, purged five times with nitrogen, thereby increasing the pressure to 0.15 to 0.2MPa, and then vented to 0.05 MPa. It was then filtered and the filter cake was washed twice with tetrahydrofuran (64kg and 63 kg); the combined washing liquid and filtrate are concentrated to 128 to 160L volume under reduced pressure (less than or equal to-0.08 MPa) and at the temperature of less than or equal to 40 ℃. Tetrahydrofuran (169kg) was added and the mixture was re-concentrated to a volume of 128 to 160L; this procedure was repeated a total of 4 times to give a solution of intermediate methyl 4-amino-3- { [ (2S) -oxetan-2-ylmethyl ] amino } benzoate.

Tetrahydrofuran (150kg) was added to this solution followed by 2-chloro-1, 1, 1-trimethoxyethane (35.1kg, 227mol) and p-toluenesulfonic acid monohydrate (1.8kg, 9.5 mol). After the reaction mixture was stirred for 25 minutes, it was heated at 40 ℃ to 45 ℃ for 5 hours, whereupon it was concentrated under reduced pressure to a volume of 135 to 181L. 2-propanol (142kg) was added and the mixture was re-concentrated to a volume of 135 to 181L, whereupon 2-propanol (36.5kg) and pure water (90kg) were added and stirring was continued until a solution was obtained. It was filtered with a line liquid filter and then treated with pure water (447kg) at 20 ℃ to 40 ℃ at a reference rate of 150 to 400 kg/hour. After the mixture was cooled to 20 to 30 ℃, it was stirred for 2 hours and the solid was collected via centrifugal filtration. The filter cake was washed with a solution of 2-propanol (20.5kg) and pure water (154 kg); after drying, P15 was obtained as a white solid (32.1kg, 109mol, 57%).1H NMR (400MHz, chloroform-d) δ 8.14-8.11(m,1H),8.01(dd, J ═ 8.5,1.1Hz,1H),7.79(br d, J ═ 8.6Hz,1H),5.26-5.18(m,1H),5.04(s,2H),4.66-4.58(m,2H),4.53(dd, a component of the ABX pattern, J ═ 15.7,2.7Hz,1H),4.34(dt, J ═ 9.1,6.0Hz,1H),3.96(s,3H),2.82-2.71(m,1H),2.48-2.37(m, 1H).

Preparation P16

2- (chloromethyl) -1-methyl-1H-benzimidazole-6-carboxylic acid methyl ester (P16)

Methyl 4-amino-3- (methylamino) benzoate (206mg, 1.14mmol) was dissolved in 1, 4-dioxane(11.5mL) and treated with chloroacetyl chloride (109. mu.L, 1.37 mmol). The mixture was stirred at 100 ℃ for 3 hours and cooled to room temperature. Triethylamine (0.8mL, 7mmol) and heptane (10mL) were added and filtered. The filtrate was concentrated under reduced pressure and the crude material was purified by chromatography on silica gel (eluent: 40% ethyl acetate in heptane) to yield 120mg of P16 (44%).1H NMR (400MHz, chloroform-d) Δ 8.14(s,1H),8.01(d,1H),7.78(d,1H),4.87(s,2H),3.97(s,3H),3.94(s, 3H); LCMS M/z 239.1[ M + H ]]+

Preparation examples P17 and P18

2- (6-Azaspiro [2.5] oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, ENT-1(P17) and 2- (6-azaspiro [2.5] oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, ENT-2(P18)

Step 1.4 Synthesis of tert-butyl 4- (2-ethoxy-2-oxoethylene) piperidine-1-carboxylate (C31)

A solution of potassium tert-butoxide (65.9g, 587mmol) in tetrahydrofuran (500mL) was added to a 0 ℃ solution of ethyl (diethoxyphosphoryl) acetate (132g, 589mmol) in tetrahydrofuran (500mL) and the resulting suspension was stirred at 0 ℃ for 1 hour, whereupon it was cooled to-50 ℃. A solution of tert-butyl 4-oxopiperidine-1-carboxylate (90.0g, 452mmol) in tetrahydrofuran (1.5L) is added dropwise at-50 ℃ and the reaction mixture is allowed to warm slowly to 20 ℃ and then stirred at 20 ℃ for 16 h. After addition of water (1L), the mixture was concentrated in vacuo to remove tetrahydrofuran. The aqueous residue was extracted with ethyl acetate (2 × 800mL) and the combined organic layers were washed with saturated aqueous sodium chloride (500mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting material was washed several times with petroleum ether (200mL) to give C31 as a white solid. Yield: 95.0g, 353mmol, 78%.1H NMR (400MHz, chloroform-d) δ 5.71(s,1H),4.16(q, J ═ 7.2Hz,2H),3.55 to 3.43(m,4H),2.94(br t, J ═ 5.5Hz,2H),2.28(br t, J ═ 5.5Hz,2H),1.47(s,9H),1.28(t, J ═ 7.0Hz, 3H).

Step 2.6 Synthesis of Azaspiro [2.5] octane-1, 6-dicarboxylic acid 6-tert-butyl 1-ethyl ester (C32)

Potassium tert-butoxide (71.2g, 634mmol) was added portionwise to a solution of trimethyl sulphoxide iodide (140g, 636mmol) in dimethyl sulphoxide (800mL) at 20 ℃. After the reaction mixture was stirred at 20 ℃ for 1.5 hours, a solution of C31(95.0g, 353mmol) in dimethyl sulfoxide (800mL) was added dropwise and stirring continued at 20 ℃ for 16 hours. Then a saturated aqueous solution of sodium chloride (2.0L) was added; the resulting mixture was neutralized with the addition of ammonium chloride and extracted with ethyl acetate (3.0L). The combined organic layers were washed sequentially with water (2 × 1.0L) and saturated aqueous sodium chloride (2.0L), dried over sodium sulfate, filtered and concentrated in vacuo. Purification via silica gel chromatography (eluent: 10:1 petroleum ether/ethyl acetate) gave C32 as a yellow oil.1H NMR analysis showed the presence of foreign aliphatic material. Yield: 80g, 280mmol, 79%.1H NMR (400MHz, chloroform-d), peak only of C32: δ 4.19-4.09(m,2H),3.55-3.39(m,3H),3.27(ddd, J ═ 13.0,7.0,4.5Hz,1H),1.76-1.64(m,2H),1.56(dd, J ═ 8.0,5.5Hz,1H, false values; partially masked by water peaks), 1.47(s,9H),1.47-1.37(m,2H),1.27(t, J ═ 7.0Hz,3H),1.17(dd, J ═ 5.0,5.0Hz,1H),0.93(dd, J ═ 8.0,4.5Hz, 1H).

Step 3.6- (tert-Butoxycarbonyl) -6-azaspiro [2.5] octane-1-carboxylic acid (C33) Synthesis

Lithium hydroxide monohydrate (37.4g, 891mmol) was added in one portion to a mixture of tetrahydrofuran (500mL) and C32(80g, 280mmol) in water (500 mL). The reaction mixture was stirred at 25 ℃ for 16 h, whereupon it was diluted with water (600mL) and washed with ethyl acetate (3X 300 mL). The organic layer was discarded and the aqueous layer was acidified to pH3 to 4 with the addition of 6M hydrochloric acid. The resulting mixture was extracted with ethyl acetate (3 × 600mL) and the combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The residue was wet milled with petroleum ether (300mL) to give whiteC33 of a colored solid. Yield: 42.0g, 164mmol, 59%. LCMS M/z 278.2[ M + Na ]+]。1H NMR(400MHz,DMSO-d6) δ 12.15-12.03(br s,1H),3.43-3.25(m,3H, assumed value; partially obscured by water peaks), 3.23-3.12(m,1H),1.64-1.50(m,2H),1.52(dd, J ═ 7.5,5.5Hz,1H),1.39(s,9H),1.39-1.28(m,2H),0.96-0.88(m, 2H).

Step 4.1 Synthesis of tert-butyl 1- ({4- (methoxycarbonyl) -2- [ (2-methoxyethyl) amino ] phenyl } carbamoyl) -6-azaspiro [2.5] octane-6-carboxylate (C34)

A solution of C33(570mg, 2.23mmol), C16(500mg, 2.23mmol) and O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU; 1.27g, 3.34mmol) in N, N-dimethylformamide (10mL) was stirred at 30 ℃ for 30 minutes, whereupon triethylamine (902mg, 8.91mmol) was added and stirring continued at 30 ℃ for 16 hours. The reaction mixture was then poured into water (60mL) and extracted with ethyl acetate (3 × 50 mL). The combined organic layers were washed with saturated aqueous sodium chloride (3 × 50mL), dried over sodium sulfate, filtered and concentrated in vacuo. Chromatography on silica gel (eluent: 1:1 petroleum ether/ethyl acetate) gave C34 as a brown oil, which was taken directly into the next step.

Step 5.2 Synthesis of methyl- [6- (tert-Butoxycarbonyl) -6-azaspiro [2.5] oct-1-yl ] -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C35)

A solution of C34 (from previous step,. ltoreq.2.23 mmol) in acetic acid (15mL) was stirred at 50 ℃ for 16 h, whereupon it was concentrated in vacuo to give C35 as a brown oil. This material was used directly in the next step. LCMS M/z 444.1[ M + H ]]+

Step 6.2 Synthesis of methyl 2- (6-azaspiro [2.5] oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C36)

Trifluoroacetic acid (5mL) was added to a solution of C35 (from the previous step;. ltoreq.2.23 mmol) in dichloromethane (10mL) and the reaction mixture was stirred at 25C for 2 h. After removal of the solvent in vacuo, the residue was basified via addition of a saturated aqueous solution of potassium carbonate (40mL) and purified by filtration in a mixture of dichloromethane and methanol (10:1,3X 40 mL). The combined organic layers were dried over magnesium sulfate, filtered, concentrated in vacuo and subjected to silica gel chromatography (eluent: 10:1:0.1 dichloromethane/methanol/concentrated ammonium hydroxide) to afford C36 as a yellow solid. Yield: 640mg, 1.86mmol, obtained in three steps to 83%. LCMS M/z 344.1[ M + H ] ]+

Step 7.2- (6-Azaspiro [2.5] oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, ENT-1(P17) and 2- (6-azaspiro [2.5] oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, isolation of ENT-2(P18)

C36(630mg, 1.83mmol) was separated into its component enantiomers using SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 55:45 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide). The first eluted peak is designated ENT-1(P17) and the second eluted enantiomer is designated ENT-2 (P18); both were isolated as a pale yellow solid.

Yield of P17: 300mg, 0.874mmol, 48%. LCMS M/z 344.1[ M + H ]]+. Retention time: 5.10 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 150mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then held at 40% B over 3.0 minutes; flow rate: 2.5 ml/min).

Yield of P18: 240mg, 0.699mmol, 38%. LCMS M/z 344.1[ M + H ]]+. Retention time: 7.35 minutes (analytical conditions were the same as those for P17).

Preparation P19

4-amino-3- { [ (1-ethyl-1H-imidazol-5-yl) methyl ] amino } benzoic acid methyl ester (P19)

Step 1.3 Synthesis of- { [ (1-Ethyl-1H-imidazol-5-yl) methyl ] amino } -4-nitrobenzoic acid methyl ester (C37)

Triethylamine (3.65mL, 26.2mmol) was added to tetrahydrofuran(12mL) in a mixture with methanol (8mL) was added a solution of methyl 3-fluoro-4-nitrobenzoate (1.00g, 5.02mmol) and 1- (1-ethyl-1H-imidazol-5-yl) methylamine, dihydrochloride (1.00g, 5.05 mmol). The reaction mixture was stirred at 60 ℃ for 40 h, whereupon it was concentrated in vacuo and purified using silica gel chromatography (gradient: 0% to 2% methanol in dichloromethane) to afford C37 as an orange solid. Yield: 1.27g, 4.17mmol, 83%.1H NMR (400MHz, chloroform-d) δ 8.24(d, J ═ 8.8Hz,1H),7.98-7.91(m,1H),7.68(d, J ═ 1.7Hz,1H),7.57(br s,1H),7.33(dd, J ═ 8.8,1.7Hz,1H),7.11(br s,1H),4.53(d, J ═ 4.9Hz,2H),3.99(q, J ═ 7.3Hz,2H),3.95(s,3H),1.47(t, J ═ 7.3Hz, 3H).

Step 2.Synthesis of methyl 4-amino-3- { [ (1-ethyl-1H-imidazol-5-yl) methyl ] amino } benzoate (P19)

A mixture of wet palladium on carbon (144mg) in methanol (13mL) and C37(412mg, 1.35mmol) was stirred under a hydrogen balloon at 25 ℃ for 16 h. The reaction mixture was then filtered through a pad of celite and the filtrate was concentrated in vacuo to give P19 as a grey solid. Yield: 340mg, 1.24mmol, 92%. 1H NMR (400MHz, methanol-d)4)δ7.66(br s,1H),7.38-7.29(m,2H),6.97(br s,1H),6.67(d,J=7.9Hz,1H),4.35(s,2H),4.11(q,J=7.3Hz,2H),3.81(s,3H),1.44(t,J=7.3Hz,3H)。

Preparation P20

4-amino-3- (methylamino) benzoic acid methyl ester (P20)

Step 1.3- (methylamino) -4-nitrobenzoic acid methyl ester (D1) Synthesis

Methylamine (38.4mL, 76.8mmol, 2M in tetrahydrofuran) was added dropwise over 10 minutes to a solution of methyl 3-fluoro-4-nitrobenzoate (5.10g, 25.6mmol) in tetrahydrofuran (60 mL). The light yellow solution turned to dark orange immediately upon addition and was stirred at room temperature for 2 hours. The reaction mixture was then diluted with diethyl ether (100mL) and the organic layer was sequentially saturated with water (50mL) and sodium chloride in water(50mL) washed. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure to give 5.26g of methyl 3- (methylamino) -4-nitrobenzoate (98%) as a dark orange solid. LCMS M/z 211.1[ M + H ]]+1H NMR (400MHz, chloroform-d) δ 8.22(d, J ═ 8.9Hz,1H),8.00(br s,1H),7.56(d, J ═ 1.7Hz,1H),7.25(dd, J ═ 8.9,1.7Hz,1H, assumed values; partially obscured by solvent peaks), 3.95(s,3H),3.09(d, J ═ 5.1Hz, 3H).

Step 2.4 Synthesis of methyl amino-3- (methylamino) benzoate (P20)

A solution of D1(5.26g, 25.0mmol) in ethanol (150mL) was added to 500mL previously charged with 10% palladium on carbon (50% water; 1g) In a bottle. The mixture was shaken at room temperature under a 50psi hydrogen atmosphere for 1 hour, whereupon it was filtered and the filter cake was rinsed with ethanol (100 mL). The filtrate was concentrated under reduced pressure to give 4.38g of P20 (97%) as an off-white solid. LCMS M/z 181.1[ M + H ]]+1H NMR (400MHz, chloroform-d) δ 7.46(dd, J ═ 8.0,1.9Hz,1H),7.34(d, J ═ 1.8Hz,1H),6.68(d, J ═ 8.0Hz,1H),3.87(s,3H),3.72(br s,2H),3.21(br s,1H),2.91(s, 3H).

Preparation examples P21 and P22

5-bromo-N3-methylpyridine-2, 3-diamine (P21) and 5-bromo-N36-dimethylpyridine-2, 3-diamine (P22)

Intermediate P21 was synthesized according to literature procedures (Choi, j.y. et al, j.med.chem.2012,55,852-870). Intermediate P22 was synthesized using the same method.

Preparation P23

2- (chloromethyl) -1- [ (1-methyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid methyl ester (P23)

Step 1.3 Synthesis of- { [ (1-methyl-1H-imidazol-5-yl) methyl ] amino } -4-nitrobenzoic acid methyl ester (D2)

1- (1-methyl-1H-imidazol-5-yl) methylamine (670mg, 6.0mmol) and triethylamine (762mg, 7.53mmol) were slowly added to a colorless solution of methyl 3-fluoro-4-nitrobenzoate (1.0g, 5.0mmol) in N, N-dimethylformamide (10 mL). The reaction mixture was stirred at 60 ℃ for 16 h, whereupon it was poured into water (30mL) and extracted with dichloromethane (3X 30 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by chromatography on silica gel (eluent: 20% methanol in dichloromethane). The resulting yellow solid was wet-milled with 30:1 petroleum ether/ethyl acetate to give D2(1.2g, 82%) as a yellow solid. LCMS M/z 290.9[ M + H ] ]+1H NMR (400MHz, chloroform-d) δ 8.25(d, J ═ 8.9Hz,1H),7.98-7.92(m,1H),7.70(d, J ═ 1.7Hz,1H),7.49(s,1H),7.34(dd, J ═ 8.9,1.7Hz,1H),7.12(s,1H),4.54(d, J ═ 5.0Hz,2H),3.96(s,3H),3.67(s, 3H).

Step 2.Synthesis of methyl 4-amino-3- { [ (1-methyl-1H-imidazol-5-yl) methyl ] amino } benzoate (D3)

10% wet palladium on carbon (1g) was added to a suspension of D2(5.46g, 18.8mmol) in methanol (160 mL). The mixture was stirred at 20 ℃ under 1 atm hydrogen for 36 hours. The reaction mixture was filtered and the filter cake was rinsed with methanol (200 mL). The filtrate was concentrated under reduced pressure to give D3(4.8g, 98%) as a brown solid. LCMS M/z 260.9[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.56(s,1H),7.18(br d,J=8.1Hz,1H),7.12(br s,1H),6.87(s,1H),6.55(d,J=8.2Hz,1H),5.50(s,2H),4.84(t,J=5.2Hz,1H),4.23(d,J=5.0Hz,2H),3.73(s,3H),3.63(s,3H)。

Step 3.2 Synthesis of methyl 2- (hydroxymethyl) -1- [ (1-methyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylate (D4)

A mixture of D3(780mg, 3.00mmol) and 2-hydroxyacetic acid (342mg, 4.49mmol) in 1,3, 5-trimethylbenzene (8mL) was stirred at 140 ℃ for 14 h and at 25 ℃ for 48 h. The clear yellow solution was decanted to give a brown residue, which was dissolved in methanol (50mL) and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (eluent: 20% methanol in dichloromethane) to afford D4(318mg, 35%) as a yellow bubble. LCMS M/z 300.9[ M + H ] ]+1H NMR(400MHz,DMSO-d6)δ8.13-8.11(m,1H),7.83(dd,J=8.4,1.6Hz,1H),7.71(d,J=8.5Hz,1H),7.59(s,1H),6.58(s,1H),5.69(s,2H),4.75(s,2H),3.84(s,3H),3.53(s,3H)。

Step 4.2 Synthesis of methyl 2- (chloromethyl) -1- [ (1-methyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylate (P23)

Thionyl chloride (990mg, 0.60mL, 8.32mmol) was added dropwise to a suspension of D4(500mg, 1.66mmol) in dichloromethane (10mL) and N, N-dimethylformamide (3mL) at room temperature. The reaction mixture was stirred at room temperature for 1 hour, followed by concentration under reduced pressure. The resulting brown residue was wet milled with dichloromethane (10 mL). The solid was collected by filtration and washed with dichloromethane (5mL) to give P23(431mg, 73%) as an off-white solid. LCMS M/z 318.9 [. M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.17(s,1H),8.31(s,1H),7.93(br d,J=8.5Hz,1H),7.82(d,J=8.5Hz,1H),7.11(s,1H),5.92(s,2H),5.13(s,2H),3.87(s,3H),3.87(s,3H)。

Preparation P24

5-chloro-2- (chloromethyl) -3-methyl-3H-imidazo [4,5-b ] pyridine (P24)

Step 1.6 Synthesis of chloro-N-methyl-3-nitropyridin-2-amine (D5)

A solution of methylamine in tetrahydrofuran (2.0M; 622mL, 1.24mol) was added dropwise via syringe to 2, 6-dichloro-3-nitropyridine (200g, 1.04mol) in ethanol (1L) and Na at 0 deg.C2CO3(132g, 1.24mol) in suspension. After the addition was complete, the reaction mixture was stirred at 18 ℃ for 6 hours. The mixture was filtered and the filtrate was concentrated under reduced pressure to giveYellow solid. The crude material was purified by silica gel chromatography (gradient: 0% to 5% ethyl acetate in petroleum ether) to afford D5(158g, 81% yield) as a yellow solid. 1H NMR(400MHz,DMSO-d6)δ8.72(br s,1H),8.41(d,J=8.6Hz,1H),6.76(d,J=8.6Hz,1H),3.00(d,J=4.8Hz,3H)。

Step 2.6-chloro-N2Synthesis of (D6) methylpyridine-2, 3-diamine

Iron powder (15.4g, 276mmol) was added to a mixture of D5(15.8g, 84.2mmol) in acetic acid (100 mL). The reaction mixture was stirred at 80 ℃ for 3 hours, whereupon it was cooled to room temperature and filtered. The filter cake was washed with ethyl acetate (2 × 100). The combined organic layers were concentrated under reduced pressure and the crude material was purified by silica gel chromatography (eluent: 1:1 ethyl acetate/petroleum ether) to give D6(8.40g, 63% yield) as a brown solid.1H NMR (400MHz, chloroform-d) δ 6.79(d, J ═ 7.7Hz,1H),6.49(d, J ═ 7.7Hz,1H),3.00(s, 3H).

Step 3.5 Synthesis of chloro-2- (chloromethyl) -3-methyl-3H-imidazo [4,5-b ] pyridine (P24)

Chloroacetyl chloride (55.5mL, 698mmol) was added to a solution of D6(50.0g, 317mmol) in 1, 4-dioxane (1.2L) and the reaction mixture was stirred at 15 ℃ for 50 minutes. It was then concentrated under reduced pressure to give a brown solid, which was dissolved in trifluoroacetic acid (1.2L) and stirred at 80 ℃ for 60 hours. The mixture was concentrated under reduced pressure to give a brown oil which was diluted with ethyl acetate (1L) and neutralized with saturated aqueous solution of sodium bicarbonate added. When the evolution of carbon dioxide subsided, the layers were separated and the aqueous layer was extracted with ethyl acetate (200 mL). The organic extracts were combined, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (gradient: 10% to 25% ethyl acetate in petroleum ether) to afford P24(61.0g, 79% yield) as a yellow solid. LCMS M/z 215.7 (observed dichloro isotope pattern) [ M + H ]+1H NMR(400MHz,DMSO-d6)δ8.13(d,J=8.3Hz,1H),7.37(d,J=8.4Hz,1H),5.11(s,2H),3.84(s,3H)。

Examples 1 and 2

2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, ENT-X1, trifluoroacetate (1) [ from C39 ]; and 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, ENT-X2, trifluoroacetate (2) [ from C40]

Step 1.2 Synthesis of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C38)

This experiment was performed in two batches of the same scale. The resulting mixture was purified by adding C2(500mg, 1.52mmol), P12(530mg, 1.59mmol) and [2 ', 6' -bis (prop-2-oxy) biphenyl-2-yl group in toluene (15mL)]A reaction vessel for a mixture of (dicyclohexyl) phosphane (Ruphos; 142mg, 0.304mmol), tris (dibenzylideneacetone) dipalladium (0) (139mg, 0.152mmol) and cesium carbonate (1.48g, 4.54mmol) was evacuated and charged with nitrogen. This evacuation cycle was repeated twice, whereupon the reaction mixture was stirred at 100 ℃ for 16 hours, combined with the second batch and filtered. The filtrate was concentrated and the residue was subjected to silica gel chromatography (gradient: 0% to 60% ethyl acetate in petroleum ether) followed by preparative thin layer chromatography (eluent: 1:1 petroleum ether/ethyl acetate) to give C38 as a light yellow solid. Combined yields: 600mg, 1.03mmol, 34%. LCMS M/z 581.0 [. multidot.M + H ] ]+

Step 2.2 isolation of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate, ENT-1(C39) and methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate, ENT-2(C40)

C38(780mg, 1.34mmol) was separated into its component enantiomers using SFC [ column: chirallTechnologies Chiralpak AD, 10 μm; mobile phase: 3:2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]And (5) realizing. The first eluting enantiomer, designated ENT-1(C39), was obtained as a white solid. Yield: 282mg, 0.485mmol, 36%. LCMS M/z 581.0 [. multidot.M + H ]]+. Retention time 1.90 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6 x 50mm, 3 μm; mobile phase a: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% B over 0.20 minutes followed by 5% to 40% B over 1.4 minutes, then held at 40% B over 1.05 minutes; flow rate: 4.0 ml/min).

The second eluted enantiomer, designated ENT-2(C40), was subjected to column chromatography using SFC: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 3:2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ]And (4) second purification. This gave C40 as a light brown solid. Yield: 280mg, 0.482mmol, 36%. LCMS M/z 581.0 [. multidot.M + H ]]+. Retention time 2.18 minutes (analytical conditions were the same as those for C39).

Step 3.2 Synthesis of 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, ENT-X1, trifluoroacetate (1) [ from C39]

Aqueous lithium hydroxide (2M; 0.30mL, 0.60mmol) was added to a solution of C39(70mg, 0.12mmol) in a mixture of methanol (3mL) and tetrahydrofuran (3 mL). After the reaction mixture was stirred at 25 ℃ for 16 h, an additional aqueous lithium hydroxide solution (2M; 0.30mL, 0.60mmol) was added and stirring continued for an additional 20 h. The reaction mixture was then adjusted to pH 7 via addition of 1M hydrochloric acid and then concentrated in vacuo to remove methanol and tetrahydrofuran. The residue was adjusted to pH 5 to 6 with the addition of trifluoroacetic acid and then purified via reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase A: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile; gradient: 30% to 60% B) to give 1 as a white solid. Yield: 40.5mg, 59.5. mu. mol, 50%. LCMS M/z 567.0 [. M + H ] ]+1H NMR (400MHz, methanol-d)4)δ8.37(br s,1H),8.07(dd,J=8.5,1.5Hz,1H),7.79(d,J=8.6Hz,1H),7.59(dd,J=8.0,8.0Hz,1H),7.34(dd,J=10.2,2.0Hz,1H),7.30(br dd,J=8.3,2.0Hz,1H),7.22(s,1H),6.87(dd,J=8.1,8.1Hz,1H),6.63(br d,J=8Hz,1H),6.60(br d,J=8Hz,1H),4.70(s,2H),4.65(t,J=4.8Hz,2H),3.75(t,J=4.8Hz,2H),3.59-3.42(m,8H),3.29(s,3H)。

Step 4.Synthesis of 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, ENT-X2, trifluoroacetate (2) [ from C40]

Aqueous lithium hydroxide (2M; 0.30mL, 0.60mmol) was added to a solution of C40(69mg, 0.12mmol) in a mixture of methanol (3mL) and tetrahydrofuran (3 mL). After the reaction mixture was stirred at 25 ℃ for 16 h, an additional aqueous lithium hydroxide solution (2M; 0.30mL, 0.60mmol) was added and stirring continued for an additional 20 h. The reaction mixture was adjusted to pH 7 via addition of 1M hydrochloric acid and then concentrated in vacuo to remove methanol and tetrahydrofuran. The residue was adjusted to pH 5 to 6 with the addition of trifluoroacetic acid and then purified via reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase A: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile; gradient: 30% to 60% B) to give 2 as a white solid. Yield: 22.9mg, 33.6. mu. mol, 28%. LCMS M/z 567.0 [. M + H ]]+1H NMR (400MHz, methanol-d)4)δ8.40-8.35(m,1H),8.07(dd,J=8.6,1.5Hz,1H),7.79(d,J=8.6Hz,1H),7.59(dd,J=8.0,8.0Hz,1H),7.35(dd,J=10.2,2.0Hz,1H),7.31(br dd,J=8,2Hz,1H),7.22(s,1H),6.87(dd,J=8.3,8.0Hz,1H),6.63(br d,J=8Hz,1H),6.60(br d,J=8Hz,1H),4.68(s,2H),4.65(t,J=4.9Hz,2H),3.76(t,J=4.8Hz,2H),3.57-3.40(m,8H),3.29(s,3H)。

Example 3

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine-6-carboxylic acid, trifluoroacetate salt (3)

Step 1.4 Synthesis of- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine (C13, free base)

P-toluenesulfonic acid monohydrate (318mg, 1.67mmol) was added to a solution of P2(300mg, 0.670mmol) in ethyl acetate (3.5 mL). The reaction mixture was stirred at 60 ℃ for 1 hour, whereupon it was basified with the addition of a saturated aqueous solution of potassium carbonate (20mL) and extracted with a mixture of dichloromethane and methanol (10:1, 3X 50 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo to afford C13, the free base, as a brown solid. Yield: 230mg, 0.661mmol, 99%.

Step 2.6 Synthesis of bromo-2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine (C41)

A suspension of C13, free base (130mg, 0.374mmol), P13(130mg, 0.427mmol) and potassium carbonate (172mg, 1.24mmol) in acetonitrile (2mL) was stirred at 50 ℃ for 16 h. The reaction mixture was then purified using preparative thin layer chromatography (eluent: ethyl acetate) to give as a brown oilC41. Yield: 114mg, 0.185mmol, 49%. LCMS M/z 617.1 (bromine-chlorine isotope pattern observed) [ M + H [) ]+

Step 3.2 Synthesis of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine-6-carboxylate (C42)

A solution of C41(114mg, 0.185mmol), 1, 3-bis (diphenylphosphino) propane (15.3mg, 37.1. mu. mol), palladium (II) acetate (8.3mg, 37. mu. mol) and triethylamine (187mg, 1.85mmol) in a mixture of methanol (5mL) and N, N-dimethylformamide (1mL) was stirred at 80 ℃ under carbon monoxide (50psi) for 16 h. After the reaction mixture was diluted with ethyl acetate (50mL), it was washed with saturated aqueous solution of sodium chloride (2 × 50mL), dried over magnesium sulfate, filtered and concentrated under reduced pressure. Purification using preparative thin layer chromatography (eluent: ethyl acetate) gave C42 as a colorless oil. Yield: 60.0mg, 0.101mmol,and 55 percent. LCMS M/z 617.2 (observed chlorine isotope pattern) [ M + Na+]。

Step 4.2 Synthesis of- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1H-imidazo [4,5-b ] pyridine-6-carboxylic acid, trifluoroacetate salt (3)

Aqueous sodium hydroxide (3M; 1.0mL, 3.0mmol) was added to a solution of C42(60.0mg, 0.101mmol) in methanol (2.0mL) and the reaction mixture was stirred at 20 ℃ for 2 h. It was then adjusted to pH 7 with the addition of 1M hydrochloric acid and extracted with a mixture of dichloromethane and methanol (10:1, 3X 30 mL). The combined organic layers were dried over magnesium sulfate, filtered, concentrated in vacuo and purified using reverse phase HPLC (column: Boston Green ODS, 5 μm; mobile phase a: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile; gradient: 10% to 95% B) to give 3 as a white solid. Yield: 29.6mg, 42.6. mu. mol, 42%. LCMS M/z 581.0 [. multidot.M + H ] ]+1H NMR (400MHz, methanol-d)4) δ 9.13(d, J ═ 1.9Hz,1H),8.74(d, J ═ 1.9Hz,1H),7.63(dd, J ═ 8.3,8.3Hz,1H),7.30(dd, J ═ 10.9,2.0Hz,1H),7.24(ddd, J ═ 8.4,2.0,0.7Hz,1H),6.89-6.84(m,1H),6.82-6.77(m,2H),4.98-4.89(m,2H, assumed values; largely shielded by water peaks), 4.64(t, J ═ 4.8Hz,2H),4.04-3.92(br m,2H),3.75(dd, J ═ 5.4,4.2Hz,2H),3.51-3.39(m,2H),3.31(s,3H),3.19-3.06(m,1H),2.41-2.24(m,2H),2.24-2.12(m,2H),2.06(d, J ═ 1.0Hz, 3H).

Examples 4 and 5

Ammonium 2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (4) and ammonium 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (5).

Step 1.4 Synthesis of- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine, trifluoroacetate salt (C43)

Trifluoroacetic acid (1.3mL) was added to a solution of P1(300mg, 0.691mmol) in dichloromethane (5 mL). The reaction mixture was stirred at 29 ℃ for 2 hours, whereupon it was concentrated in vacuo to afford C43 as a brown oil, which was used directly in the next step.

Step 2.2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester (C44) and the synthesis of 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester (C45).

P15(204mg, 0.692mmol) was added to a solution of C43 (. ltoreq.0.691 mmol from the previous step) in acetonitrile (10mL), followed by potassium carbonate (956mg, 6.92 mmol). The reaction mixture was stirred at 29 ℃ for 16 hours and then filtered; the filtrate was concentrated in vacuo to give a residue which was purified by preparative thin layer chromatography (eluent: 2:1 petroleum ether/ethyl acetate) to give the diastereomeric product as a yellow gum (178 mg). Separation into two products was performed via SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 55:45 carbon dioxide/(methanol containing 0.1% ammonium hydroxide)]The process is carried out. The first eluting diastereomer obtained as a yellow oil is designated C44. Yield: 44.3mg, 74.8. mu. mol, obtained 11% in 2 steps. LCMS M/z 592.1 [. M + H ] ]+. Retention time 4.26 min (column: Chiral Technologies ChiralCel OD-3, 4.6 x 100mm, 3 μm; mobile phase a: carbon dioxide; mobile phase B: methanol with 0.05% diethylamine; gradient: 5% to 40% B over 4.5 min, then hold at 40% B over 2.5 min; flow rate: 2.8 ml/min).

The second eluted diastereomer was received via SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 3:2 carbon dioxide/(methanol containing 0.1% ammonium hydroxide)]To give the second eluted diastereomer as a colourless oil, which is then labelledShown as C45. Yield: 38mg, 64. mu. mol, in 2 steps to give 9%. LCMS M/z 592.1 [. M + H ]]+. Retention time 4.41 minutes (analytical conditions were the same as those for C44).

The absolute stereochemistry shown on dioxolane was assigned via the efficacy correlation of the 5 and 5 samples, the free acid synthesized from intermediate C48; the absolute stereochemistry of this intermediate was determined via single crystal X-ray structural determination of C49 (hemisulfate salt of C48) (see below).

Step 3.2 Synthesis of ammonium 2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (4)

Aqueous lithium hydroxide (2M; 0.80mL, 1.6mmol) was added to a solution of C44(44.3mg, 74.8. mu. mol) in a mixture of methanol (1mL) and tetrahydrofuran (1mL) and the reaction mixture was stirred at 26 ℃ for 3 hours. It was then adjusted to pH 7 with the addition of trifluoroacetic acid, the resulting mixture was concentrated in vacuo and purified using reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase a: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 30% to 50% B) to give 4 as a white solid. Yield: 26.6mg, 44.7. mu. mol, 60%. LCMS M/z 578.0 [. multidot.M + H ]]+1H NMR (400MHz, methanol-d)4) δ 8.31(d, J ═ 1.4Hz,1H),7.96(dd, J ═ 8.5,1.6Hz,1H),7.66(d, J ═ 8.5Hz,1H),7.57(dd, J ═ 8.0,8.0Hz,1H),7.34(dd, J ═ 10.1,2.0Hz,1H),7.29(br dd, J ═ 8.3,2.0Hz,1H),7.20(s,1H),6.86-6.79(m,1H),6.77(br dd, a component of an ABC pattern, J ═ 7.9,1.3Hz,1H),6.73(dd, a component of an ABC pattern, J ═ 7.5,1.4Hz,1H), 5.5-5.29.18H, 1.78 (m,1H), a pseudo-value; partially masked by water peak), 4.68(dd, J ═ 15.3,2.7Hz,1H),4.54(td, J ═ 8.0,5.9Hz,1H),4.44(dt, J ═ 9.2,5.9Hz,1H),4.02(AB quartet, J), J, 1H),4.02(AB quartet, J, 1H)AB=13.9Hz,ΔνAB=49.0Hz,2H),3.18-3.08(m,1H),3.05-2.96(m,1H),2.81-2.68(m,2H),2.56-2.45(m,1H),2.45-2.30(m,2H),2.03-1.88(m,2H),1.88-1.79(m,2H)。

Step 4.2 Synthesis of ammonium 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (5)

Aqueous lithium hydroxide (2M; 0.80mL, 1.6mmol) was added to a solution of C45(38mg, 64. mu. mol) in a mixture of methanol (1mL) and tetrahydrofuran (1mL) and the reaction mixture was stirred at 24 ℃ for 2.5 h. It was then adjusted to pH7 with the addition of 1M hydrochloric acid, the resulting mixture was concentrated in vacuo and purified using reverse phase HPLC (column: Agela Durashell C18, 5 μ M; mobile phase a: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 29% to 49% B) to give 5 as a white solid. Yield: 27.9mg, 46.9. mu. mol, 73%. LCMS M/z 577.9 [. M + H ]]+1H NMR (400MHz, methanol-d)4) δ 8.32(d, J ═ 1.4Hz,1H),7.96(dd, J ═ 8.5,1.5Hz,1H),7.66(d, J ═ 8.5Hz,1H),7.56(dd, J ═ 8.0,8.0Hz,1H),7.34(dd, J ═ 10.2,2.0Hz,1H),7.29(br dd, J ═ 8.3,2.0Hz,1H),7.20(s,1H),6.85-6.80(m,1H),6.77(dd, a component of an ABC pattern, J ═ 8.0,1.3Hz,1H),6.73(dd, a component of an ABC pattern, J ═ 7.5,1.4, 1H), 5.30-5.5.20 (m, 4H), 4.79(m, 1H); partially masked by water peak), 4.68(dd, J ═ 15.4,2.7Hz,1H),4.62-4.54(m,1H),4.44(dt, J ═ 9.2,5.9Hz,1H),4.02(AB quartet, J ═ 9.2, J, 1H),4.02(AB quartet, J ═ 1H)AB=13.9Hz,ΔνAB=44.6Hz,2H),3.18-3.09(m,1H),3.06-2.97(m,1H),2.80-2.67(m,2H),2.55-2.30(m,3H),2.02-1.78(m,4H)。

Example 5 alternative Synthesis of free acid

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid (5, free acid)

Step 1 isolation of tert-butyl 4- [ (2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (C46) and tert-butyl 4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate (C47)

P1(10g, 23mmol) was separated into its component enantiomers using reverse phase HPLC [ column: phenomenex Lux Amylose-1, 5 μm; mobile phase: 9:1 carbon dioxide/(2-propanol containing 0.2% of 1-aminopropan-2-ol) ]. The first eluting enantiomer was designated C46 and the second eluting enantiomer was designated C47; both were obtained as colorless oils. The absolute stereochemistry of C46 and C47 shown was assigned based on single crystal X-ray structural determination performed from C49 synthesized from C47 (see below).

Yield of C46: 4.47g, 10.3mmol, 45%. Retention time: 3.98 min [ column: phenomenex Lux Amylose-1, 4.6X 250mm, 5 μm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol containing 0.2% 1-aminopropan-2-ol; gradient: 5% of B over 1.00 min followed by 5% to 60% of B over 8.00 min; flow rate: 3.0 ml/min; back pressure: 120 bar ].

Yield of C47: 4.49g, 10.3mmol, 45%. Retention time: 4.32 minutes (conditions for analytical SFC were the same as for C46).

Step 2.4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine (C48) Synthesis

P-toluenesulfonic acid monohydrate (566mg, 2.98mmol) was added to a solution of C47(1.12g, 2.58mmol) in ethyl acetate (26 mL). After the reaction mixture was heated at 45 ℃ for 16 hours, it was concentrated in vacuo, dissolved in ethyl acetate and washed with a saturated aqueous solution of sodium bicarbonate. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered and concentrated under reduced pressure to give C48(947mg), LCMS M/z 334.0 [. multidot.M + H ] as a foamy white solid]+. A portion of this material, still containing some p-toluenesulfonic acid, was used in the synthesis of C50 below.

A second portion of foamy white solid (440mg) was dissolved in ethyl acetate (25mL) and washed with saturated aqueous sodium bicarbonate (2 × 15 mL); the organic layer was dried over magnesium sulfate, filtered and concentrated inConcentration in vacuo afforded C48(350mg) as a colorless oil, which contained no more p-toluenesulfonic acid. Adjusted yields: 350mg, 1.05mmol, 88%.1H NMR (400MHz, chloroform-d) δ 7.53(dd, J ═ 8.4,7.8Hz,1H),7.22 to 7.13(m,3H),6.87 to 6.80(m,1H),6.79 to 6.71(m,2H),3.23 to 3.14(m,2H),2.86 to 2.69(m,3H),1.90 to 1.68(m, 4H).

Step 3.4 Synthesis of- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidine, hemisulfate salt (C49)

A 0.1M solution of C48 (from the colorless oil above) in ethyl acetate was prepared and subjected to salt screening. Only the formation of sulfate is illustrated here. A mixture of sulfuric acid (25. mu. mol) and substrate solution (0.1M, 250. mu.L, 25. mu. mol) was heated to 45 ℃ for 1 hour, allowed to cool to room temperature and stirred for 15 hours. The resulting suspension was treated with methanol (about 150 μ L) until a solution was formed; it was allowed to evaporate slowly through the spacer until about 50. mu.L of solvent remained. The crystal obtained was analyzed by single crystal X-ray texture measurement to establish the absolute stereochemistry as shown.

Single crystal X-ray texture determination of C49

Single crystal X-ray analysis

Data collection was performed on a Bruker D8 Venture diffractometer at room temperature. Data collection is represented by ω andscanning the image.

The structure was resolved with the intrinsic phasing of triclinic family space group P1 using the SHELX software package. The structure is then refined with a full matrix least squares method. All non-hydrogen atoms were found and refined using anisotropic shift parameters.

The hydrogen atoms located on the nitrogen were found from the fourier difference map and refined with a constrained distance. The remaining hydrogen atoms are positioned at the calculated positions and allowed to sit on their carrier atoms. The final refinement includes the anisotropic displacement parameters of all hydrogen atoms.

The asymmetric unit consists of two molecules of protonated C48, one molecule of doubly deprotonated sulfuric acid, and one molecule of total water occupancy. Thus, the structure is hemisulfate and hemihydrate. The chlorofluorophenyl ring was disordered and modeled with an occupancy of 60/40, flipping the ring in two positions.

Analysis of absolute structure using likelihood method (houtt, 2008) was performed using platon (spek). The results show that the absolute structure has been correctly assigned; the probability of correct structure calculated by this method is 100.0%. The Hoff parameter is reported as 0.061 with an esd of 0.004, and the Parsen parameter is reported as 0.063 with an esd of 0.005.

The final R index was 3.1%. The resulting difference fourier reveals the electron density without loss or dislocation.

The relevant crystal, data collection and refinement information is summarized in table E. The atomic coordinates, bond lengths, bond angles, and displacement parameters are listed in tables F through H.

Software and reference data

SHELXTL,Version 5.1,Bruker AXS,1997。

PLATON,A.L.Spek,J.Appl.Cryst.2003,36,7-13。

Mercure, c.f. macroe, p.r. edington, p.mccabe, e.pidcock, g.p. shields, r.taylor, m.towler, and j.van de street, j.appl.cryst.2006, 39, 453.

OLEX2, o.v.dolomanov, l.j.bourhis, r.j.gildea, j.a.k.howard, and h.puschmann, j.appl.cryst.2009, 42, 339-.

R.w.w.hooft, l.h.straver, and a.l.spek, j.appl.cryst.2008, 41, 96-103.

H.D.Flack,Acta Cryst.1983,A39,867-881。

Crystal data and structure refinement of table e.c 49.

C49 of Table FAtomic coordinates (x 10)4) And equivalent isotropic displacement parameterU (eq) is defined as orthogonal UijOne third of the trace of the tensor.

C49 bond LengthAngle of harmony key [ ° ]]

For generating symmetric transformations of equivalent atoms.

The different characteristic displacements of table h.c49 are giy chicken \25976The anisotropic displacement factor takes the form: -2 pi2[h2 a*2U11+...+2 h k a b U12]。

Step 4.2 Synthesis of methyl 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (C50)

A solution of C48(500mg from above as a foamy white solid, corrected for p-toluenesulfonic acid: 1.25mmol) in acetonitrile (6mL) was treated with N, N-diisopropylethylamine (0.68mL, 3.9mmol) and allowed to stir at 45 ℃ for 5 min. After addition of P15(319mg, 1.08mmol), stirring was continued at 45 ℃ for 7.25 h, whereupon the reaction mixture was diluted with water (6mL) and acetonitrile (2mL) at 45 ℃. The resulting heterogeneous mixture was allowed to cool to room temperature and stirred for 72 hours. More water (5mL) was added and stirred for another 30 minutes Thereafter, the solid was collected via filtration and washed with a mixture of acetonitrile and water (15:85, 3 × 5mL) to give C50 as a white solid with a pale pink color. Yield: 605mg, 1.02mmol, 82%. LCMS M/z 592.0 [. multidot.M + H ]]+1H NMR (400MHz, chloroform-d) δ 8.17(d, J ═ 1.6Hz,1H),7.96(dd, J ═ 8.5,1.5Hz,1H),7.73(d, J ═ 8.4Hz,1H),7.51(dd, J ═ 8.0,8.0Hz,1H),7.19 (brs, 1H),7.18-7.14(m,2H),6.85-6.79(m,1H),6.76-6.71(m,2H),5.26-5.18(m,1H),4.73(dd, a component of ABX pattern, J ═ 15.3,5.9Hz,1H),4.67(dd, a component of ABX pattern, J ═ 15.3,3.5, 1H, 4.63-4.55.3, 5.9Hz,1H),4.67(dd, 2H), 3.3, 2H, 3.5.9H, 2H, 1H, 2H, 3.7.7.7H, 1H, 3H, 1H, 2H, 3H, 2H, and m,2H, 7H, 2H, 7H, 2H, 7, 1.91-1.76(m, 4H).

Step 5.2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid (5, free acid) synthesis

A suspension of C50(595mg, 1.00mmol) in methanol (10mL) was heated to 45 ℃ and treated with aqueous sodium hydroxide (1M; 2.01mL, 2.01 mmol). After 21 hours at 45 ℃, the reaction mixture was allowed to cool to room temperature; it was then treated with aqueous citric acid (1M, 1mL) to bring the pH to 5 to 6. Water (10mL) was added and the mixture was stirred for 1 hour, whereupon the solid was collected by filtration. It was washed with a mixture of methanol and water (1:10, 3X 5mL) to give a solid (433 mg). A portion of this material (300mg) was stirred at 40 ℃ for 1 hour along with a mixture of heptane and ethyl acetate (1:3, 5 mL); after cooling to room temperature with constant stirring, the solid was collected via filtration and washed with a mixture of heptane and ethyl acetate (3:1, 3 × 3mL) to give 5, the free acid as a white solid. Yield: 260mg, 0.450mmol, corresponding to 65% of the total reaction. LCMS M/z 578.0 [. multidot.M + H ] ]+1H NMR(400MHz,DMSO-d6) δ 12.75(v br s,1H),8.26(br s,1H),7.79(dd, J ═ 8.4,1.6Hz,1H),7.66-7.56(m,3H),7.40(dd, J ═ 8.3,2.0Hz,1H),7.35(s,1H),6.87-6.75(m,3H),5.13-5.03(m,1H),4.76(dd, a component of ABX pattern, J ═ 15.3,7.2Hz,1H),4.62(dd, a component of ABX pattern, J ═ 15.2,2.8Hz,1H),4.46-4.38(m,1H),4.34(d, J ═ 9.0,5.9,5.8, 1H, 9.9, 5.8, 1H), 1H, and d, 2.3 Hz,1H) 3.84(AB quartet, J)AB=13.5Hz,ΔνAB=67.7Hz,2H),3.00(br d,J=11.2Hz,1H),2.84br(d,J=11.3Hz,1H),2.71-2.56(m,2H),2.45-2.34(m,1H),2.28-2.08(m,2H),1.84-1.65(m,4H)。

This material was determined to have the same absolute configuration as example 5 above by comparing its biological activity in assay 2 to both 4 and 5, the 5, free acid sample exhibiting an EC of 25nM50(geometric mean repeated 3 times). The activity of the ammonium salts of example 4 and example 5 in assay 2, respectively, was>20000nM (2 replicates) and 20nM (geometric mean of 3 replicates).

Example 5 Synthesis of a 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt (5, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt)

A mixture of 5, free acid (0.50g, 0.86mmol) in tetrahydrofuran (4mL) was treated with aqueous 2-amino-2- (hydroxymethyl) propane-1, 3-diol (tris (hydroxymethyl) aminomethane salt, 1.0M; 0.5mL, 1.0 mmol). After 20 h, the mixture was concentrated with ethanol (2 × 6mL) in vacuo. The mixture was treated with ethanol (4 mL). After stirring for 48 hours, the solid was collected via filtration, washed with ethanol (2 × 10mL) and dried under vacuum to give 5, 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt as a white solid. Yield: 410mg, 0.586mmol, 68%. 1H NMR(600MHz,DMSO-d6) And characteristic peak: δ 8.19(s,1H),7.78(br d, J ═ 8.4Hz,1H),7.62-7.58(m,2H),7.55(d, J ═ 8.3Hz,1H),7.40(dd, J ═ 8.4,2.0Hz,1H),7.35(s,1H),6.85-6.80(m,2H),6.79(dd, J ═ 6.9,2.4Hz,1H),5.11-5.05(m,1H),4.73(dd, J ═ 15.2,7.2Hz,1H),4.60(dd, J ═ 15.3,2.9Hz,1H),4.45-4.39(m,1H),4.34(dd, J ═ 9.0,6.0, J ═ 8, 3, 3.9 Hz,1H), 13.13H, 13H, and d.5Hz,1H),2.99(br d, J ═ 11.1Hz,1H),2.85(br d, J ═ 11.3Hz,1H),2.68-2.59(m,2H),2.44-2.37(m,1H),2.25-2.18(m,1H),2.17-2.10(m,1H),1.80-1.69(m, 4H). mp-168 ℃ to 178 ℃.

Examples 6 and 7

2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid ammonium salt (6) and 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid ammonium salt (7)

Step 1.4 Synthesis of- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidine, p-toluenesulfonate (C13)

A solution of P2(150mg, 0.335mmol) and P-toluenesulfonic acid monohydrate (159mg, 0.836mmol) in ethyl acetate (2.0mL) was stirred at 60 ℃ for 3.5 h. The reaction mixture was concentrated in vacuo to afford C13 as a brown oil, which was used directly in the next step. LCMS M/z 348.1 [. M + H ] ]+

Step 2.2 Synthesis of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (C51)

P15(99.1mg, 0.336mmol) was added to a suspension of C13 (from the previous step;. ltoreq.0.335 mmol) and potassium carbonate (232mg, 1.68mmol) in acetonitrile (5.0 mL). The reaction mixture was stirred at 60 ℃ for 10 hours, whereupon it was filtered and the filtrate was concentrated in vacuo. After the residue (390mg) was combined with the material from a similar reaction using C13(≦ 0.11mmol), it was diluted with water (20mL) and extracted with a mixture of dichloromethane and methanol (10:1, 3X 30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo and subjected to preparative thin layer chromatography (eluent: 1:1 dichloro-methane)Methane/methanol) to give C51, a mixture of diastereomers as a colorless oil. Combined yields: 80.6mg, 0.133mmol, in 2 steps to give 30%. LCMS M/z 606.2 [. M + H ]]+

Step 3.2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester (C52) and 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester Isolation of the ester (C53)

C51(180mg, 0.297mmol) was separated into its component diastereomers via repeated SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65:35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]The process is carried out. The first eluting diastereomer is designated C52. Yield: 61.2mg, 0.101mmol, 34%. LCMS M/z 627.9 [. M + Na ]+]. Retention time: 5.03 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 150mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then held at 40% B over 3.0 minutes; flow rate: 2.5 ml/min).

The second eluting diastereomer is designated C53. Upon analysis, this material was confirmed to be contaminated with the corresponding ethyl ester; it is forwarded as this mixture to the hydrolysis step (yield 7). Yield: 40.0mg, 66.0. mu. mol, 22%. LCMS M/z 606.0 [. M + H ]]+. Retention time: 5.19 minutes (analytical conditions identical to those used for C52).

The absolute stereochemistry shown on dioxolane was assigned via the efficacy correlation of 7 with a 7, free acid sample synthesized from intermediate P3 (see below, example 7, alternative synthesis of free acid); the absolute stereochemistry of P3 was established via single crystal X-ray structural determination of C8 (see below).

Step 4.2- ({4- [ (2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid ammonium salt (6) Synthesis

Aqueous lithium hydroxide (2M; 0.990mL, 1.98mmol) was added to a solution of C52(60mg, 99. mu. mol) in a mixture of methanol (1.0mL) and tetrahydrofuran (1.0mL) and the reaction mixture was stirred at 20 ℃ for 16 h. Trifluoroacetic acid was added until the pH of the reaction mixture reached 7, which was then concentrated in vacuo and the residue was purified using reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase a: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 29% to 49% B) to give 6 as a white solid. Yield: 14.4 mg. 23.6. mu. mol, 24%. LCMS M/z 592.0 [. multidot.M + H ]]+1H NMR (400MHz, methanol-d)4) And characteristic peak: δ 8.35(d, J ═ 1.3Hz,1H),7.97(dd, J ═ 8.5,1.5Hz,1H),7.67(d, J ═ 8.5Hz,1H),7.58(dd, J ═ 8.3,8.3Hz,1H),7.28(dd, J ═ 10.9,2.0Hz,1H),7.21(br dd, J ═ 8.4,1.9Hz,1H),6.81-6.75(m,1H),6.74-6.68(m,2H),5.33-5.25(m,1H),4.72(dd, J ═ 15.4,2.7Hz,1H),4.49(dt, J ═ 9.1,6.0, 1H),4.03 (J ═ 9.1,6.0, 1H), four-fold (J ═ 8, J ═ 8.3Hz,1H),7 (J- AB=13.9Hz,ΔνAB=47.8Hz,2H),3.14(br d,J=11Hz,1H),3.02(br d,J=11.5Hz,1H),2.88-2.78(m,1H),2.77-2.68(m,1H),2.60-2.50(m,1H),2.47-2.32(m,2H),2.03(d,J=1.1Hz,3H),2.01-1.87(m,2H),1.87-1.78(br m,2H)。

Step 5 Synthesis of ammonium 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (7)

Aqueous lithium hydroxide (2M; 0.642mL, 1.28mmol) was added to a solution of C53(38.9mg, 64.2. mu. mol) in a mixture of methanol (1.0mL) and tetrahydrofuran (1.0 mL). After the reaction mixture was stirred at 20 ℃ for 16 h, it was adjusted to pH 7 with the addition of trifluoroacetic acid, concentrated in vacuo and purified using reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase A: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 0% to 80% B) to give 7 as a white solid. Yield: 25.1mg, 41.2. mu. mol, 64%. LCMS M/z 591.9 [. M + H ]]+1H NMR (400MHz, methanol-d)4) And characteristic peak: δ 8.34(d, J ═ 1.5Hz,1H),7.98(dd, J ═ 8.5,1.6Hz,1H),7.68(d, J ═ 8.5Hz,1H),7.58(dd, J ═ 8.3,8.3Hz,1H),7.28(dd, J ═ 10.9,2.0Hz,1H),7.20(br dd, J ═ 8.4,1.9Hz,1H),6.81-6.74(m,1H),6.74-6.67(m,2H),5.33-5.23(m,1H),4.73(dd, J ═ 15.4,2.7Hz,1H),4.68-4.61(m,1H),4.48 (J, 9.9, 1H), four-fold (AB, 1H), 1H, 4.05, 1H, J ═ 9, 1H, and J ═ 8, 1H AB=13.9Hz,ΔνAB=44.1Hz,2H),3.15(br d,J=11.7Hz,1H),3.03(br d,J=11.6Hz,1H),2.87-2.69(m,2H),2.60-2.49(m,1H),2.48-2.33(m,2H),2.03(br s,3H),2.01-1.77(m,4H)。

Example 7 alternative Synthesis of free acid

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid (7, free acid)

Step 1.2 Synthesis of methyl 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (C53)

N, N-diisopropylethylamine (15.1mL, 86.9mmol) was added to a mixture of P3(8.22g, 15.8mmol) in acetonitrile (185 mL); after stirring for 5 min, P15(4.57g, 15.5mmol) was added and the reaction mixture was heated at 45 ℃. After 4 hours, the reaction mixture was concentrated in vacuo to half its original volume, and the resulting mixture was diluted with water (100mL) and extracted with ethyl acetate (2 × 100 mL). The combined organic layers were washed with water (50mL), dried over magnesium sulfate, filtered and concentrated in vacuo. Chromatography on silica gel (gradient: 50% to 100% ethyl acetate in heptane) afforded C53 as a white solid. Yield: 8.4g, 13.9mmol, 88%. LCMS M/z 606.1 [. M + H ]]+1H NMR(600MHz,DMSO-d6)δ8.30(s,1H),7.82(br d,J=8.4Hz,1H),7.67(d,J=8.4Hz,1H),7.58-7.53(m,2H),7.33(dd,J=8.4,2.1Hz,1H),6.80-6.76(m,2H),6.76-6.72(m,1H),5.14-5.07(m,1H),4.81(dd,J=15.2,7.2Hz,1H),4.67(dd,J=15.3,2.8Hz,1H),4.51-4.44(m,1H),4.37(ddd,J=8.9,5.9,5.9Hz,1H),3.97(d,J=13.6Hz,1H),3.87(s,3H),3.78(d,J=13.5Hz,1H),3.02(br d,J=11.1Hz,1H),2.86(br d,J=11.3Hz,1H),2.74-2.60(m,2H),2.48-2.41(m,1H),2.29-2.22(m,1H),2.21-2.14(m,1H),2.02(s,3H),1.83-1.73(m,2H),1.73-1.64(m,2H)。

Step 2.2 Synthesis of 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid (7, free acid)

A mixture of C53(8.40g, 14.0mmol) in methanol (135mL) was heated to 45 ℃ and treated with aqueous sodium hydroxide (1M; 27.7mL, 27.7 mmol). After 20 hours, the reaction mixture was concentrated in vacuo to half its original volume. The resulting mixture was diluted with water (100mL) and adjusted to pH 5 to 6 using aqueous citric acid (1M, 15 mL). The resulting solid was filtered, washed with water (2 × 15mL) and transferred as a solution in ethyl acetate (50mL) to a separatory funnel; in this way residual water is removed. The organic layer was dried over magnesium sulfate, filtered, combined with four previously prepared batches from a similar procedure (the amount of C53 used in the reaction was 987mg, 1.63 mmol; 1.15g, 1.90 mmol; 8.57g, 14.1 mmol; and 12.6g, 20.8mmol) and concentrated in vacuo. The resulting sticky solid was treated with 10% ethyl acetate in heptane (500 mL). After 4 hours, the solid was collected via filtration and washed with 10% ethyl acetate in heptane (2 × 25mL) to give 7, the free acid as a white solid. The combined reaction yields were 29.4g, 0.527mmol, 74%. LCMS 592.2 [. multidot.M + H ]]+。1H NMR(600MHz,DMSO-d6):δ12.74(br s,1H),8.28(s,1H),7.80(br d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.59-7.52(m,2H),7.33(dd,J=8.4,2.1Hz,1H),6.81-6.76(m,2H),6.76-6.72(m,1H),5.14-5.07(m,1H),4.79(dd,J=15.3,7.3Hz,1H),4.65(dd,J=15.2,2.8Hz,1H),4.51-4.45(m,1H),4.38(ddd,J=9.0,5.9,5.9Hz,1H),3.96(br d,J=13.6Hz,1H),3.78(br d,J=13.5Hz,1H),3.02(br d,J=11.1Hz,1H),2.86(br d,J=11.1Hz,1H),2.74-2.60(m,2H),2.48-2.41(m,1H),2.29-2.21(m,1H),2.21-2.14(m,1H),2.02(s,3H),1.83-1.74(m,2H),1.74-1.64(m, 2H). This material was determined to have the same absolute configuration as example 7 above by comparing its biological activity in assay 2 to both 6 and 7, a sample of this 7, free acid, exhibiting an EC of 4.3nM 50(geometric mean repeated 3 times). The activity of the ammonium salts of example 6 and example 7 in assay 2 was 2400nM (geometric mean of 5 replicates) and 2.9nM (geometric mean of 8 replicates), respectively.

Example 7 Synthesis of a, 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt (7, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt)

A mixture of 7, the free acid (2.00g, 3.38mmol) in tetrahydrofuran (16mL) was treated with an aqueous solution of 2-amino-2- (hydroxymethyl) propane-1, 3-diol (tris (hydroxymethyl) aminomethane salt, 1.0M; 3.55mL, 3.55 mmol). After 18 hours, the reaction mixture was concentrated in vacuo and treated with ethanol (30 mL). After the mixture was stirred for 23 hours, the solid was collected via filtration and washed with ethyl acetate (2 × 10mL) to give 7, 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt as a white solid. Yield: 1.41g, 1.98mmol, 59%. LCMS M/z592.3 [. M + H ]]+1H NMR(600MHz,DMSO-d6) And characteristic peak: δ 8.20(s,1H),7.79(d, J ═ 8.4Hz,1H),7.59-7.52(m,3H),7.33(br d, J ═ 8.5Hz,1H),6.81-6.72(m,3H),5.14-5.07(m,1H),4.76(dd, J ═ 15.2,7.2Hz,1H),4.63(br d, J ═ 15.4Hz,1H),4.50-4.44(m,1H),4.37(ddd, J ═ 8.9,5.9,5.9Hz,1H),3.94(d, J ═ 13.4Hz,1H),3.76(d, J ═ 13.4Hz,1H),3.01(br d, J ═ 11.2H, 1H, 2H, 3.83 (br d, 2H), 2H, 3.3.3.3.3.3.3.3.3.3.3.3.3.3.3.3, 2H, 1H, 2H, 1H, 2H, and 1H, 2H, 1H, 2H, 1H, 2H, etc.). mp 175 ℃ to 180 ℃.

Example 7 acquisition of powder X-ray diffraction (PXRD) data for form I of the 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt

The white solid of tris (hydroxymethyl) aminomethane salt of example 7 was submitted for PXRD analysis and found to be a crystalline material (which is labeled as form I of this anhydrous crystalline form). Powder X-ray diffraction analysis was performed using a Bruker AXS D8 Endeavor diffractometer equipped with a Cu radiation source. The divergence slit was set at 15mm continuous illumination. The diffracted radiation was detected with a PSD-Lynx Eye detector with the detector PSD set at 2.99 degrees on. The X-ray tube voltage and current amount were set to 40kV and 40mA, respectively. Step size of 0.01 degree and step time of 1.0 second at Cu wavelength in theta-theta goniometer of 3.0 to 40.02 theta degree Data is collected. The anti-scatter screen is set to a fixed distance of 1.5 mm. The sample was rotated during data collection. The sample was prepared by placing it in a silicon low background sample holder and rotated during collection. Data were collected using Bruker DIFFRAC Plus software and analysis was performed with EVA DIFFRAC Plus software. The PXRD data file is not processed until peak search. Preliminary peak assignment was performed using a peak search algorithm in the EVA software, using peaks selected to have a threshold value of 1. In order to ensure the effectiveness, the adjustment is performed manually; to visually inspect the output of the automated dispense and adjust the peak position to the maximum peak value. Peaks with a relative intensity of 3% or more are generally selected. Peaks that are unresolved or consistent with noise are generally not selected. Typical errors associated with peak positions of PXRD are specified in USP as at most +/-0.2 deg. 2 theta (USP-941). One diffraction pattern was consistently observed and provided in fig. 24. A list of diffraction peaks expressed in degrees 2 θ above and having a relative intensity of ≧ 3.0% is provided in Table X1.

TABLE X1

One embodiment provides a crystalline form of the anhydrous tris (hydroxymethyl) aminomethane salt of 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid. In other embodiments, a crystalline form of the anhydrous (anhydrous) tris (hydroxymethyl) aminomethane salt of 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid is designated as "form I" (form I), which is characterized by its unique solid state properties (such as PXRD substantially as depicted in figure 24) associated with, for example, powder X-ray diffraction (PXRD) as described herein. In some embodiments, performance comprises at least two signals selected from at 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 8.5 +/-0.2 degrees; 10.1 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of 16.9. + -. 0.2 ℃ characteristic peaks expressed in 2. theta. In some embodiments, form I expression comprises at least three amino acid residues selected from at 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 8.5 +/-0.2 degrees; 10.1 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of 16.9. + -. 0.2 ℃ characteristic peaks expressed in 2. theta. In some embodiments, form I expression comprises at least four amino acid residues selected from at 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 8.5 +/-0.2 degrees; 10.1 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of 16.9. + -. 0.2 ℃ characteristic peaks expressed in 2. theta. In some embodiments, form I expression comprises at least five amino acid residues selected from at 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 8.5 +/-0.2 degrees; 10.1 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of 16.9. + -. 0.2 ℃ characteristic peaks expressed in 2. theta.

In some embodiments, form I exhibits a powder X-ray diffraction pattern comprising characteristic peaks expressed in 2 Θ at 3.7 ± 0.2 ° and 7.3 ± 0.2 °.

In some embodiments, form I expression comprises an angle of 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; and 14.7 ± 0.2 ° peaks expressed in 2 θ. In other embodiments, form I exhibits an amino acid sequence comprising at least one amino acid selected from the group consisting of 8.5 ± 0.2 °; 10.1 +/-0.2 degrees; and a powder X-ray diffraction pattern of peaks expressed in 2 θ at 16.9. + -. 0.2 ℃.

In some embodiments, form I expression comprises an angle of 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of peaks expressed in 2 θ at 16.9. + -. 0.2 ℃.

In some embodiments, form I expression comprises an angle of 3.7 ± 0.2 °; 7.3 +/-0.2 degrees; 8.5 +/-0.2 degrees; 10.1 +/-0.2 degrees; 14.7 +/-0.2 degrees; and a powder X-ray diffraction pattern of peaks expressed in 2 θ at 16.9. + -. 0.2 ℃.

In some embodiments, form I exhibits a powder X-ray diffraction pattern substantially as shown in figure 24. The diffraction peaks expressed in degrees 2 θ above and a list of relative intensities with relative intensities ≧ 3.0% are provided in Table X1.

Examples 8 and 9

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X1(8) [ from C56 ]; and 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2(9) [ from C57]

Step 1.3 Synthesis of 3-fluoro-4- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] benzonitrile, p-toluenesulfonate (C54)

P-toluenesulfonic acid (158mg, 0.919mmol) was added to a solution of P4(161mg, 0.367mmol) in ethyl acetate (8mL) and the reaction mixture was stirred at 65 ℃ for 16 h. Removal of the solvent in vacuo afforded C54 as a dark yellow gum; this material was taken directly to the next step.

Step 2.2 Synthesis of methyl 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate (C55)

Potassium carbonate (219mg, 1.58mmol) was added to a solution of C54 (from the previous step;. ltoreq.0.367 mmol) in acetonitrile (3.7mL), followed by P15(115mg, 0.390 mmol). The reaction mixture was stirred at 50 ℃ for 20 hours, whereupon it was diluted with ethyl acetate (10mL) and filtered. The filter cake was washed with ethyl acetate (3 × 10mL) and the combined filtrates were concentrated in vacuo. Chromatography on silica gel (gradient: 0% to 100% ethyl acetate in petroleum ether) afforded C55 as a dark yellow gum. Yield: 191.0mg, 0.320mmol, in 2 steps gave 87%. LCMS M/z 619.1[ M + Na ] +]。

Step 3.2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester, ENT-1(C56) and 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid methyl ester, isolation of ENT-2(C57)

C55(191mg, 0.320mmol) separated into its component stereoisomers at dioxolane via SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 3:2 carbon dioxide/2-propanol]The process is carried out. The first eluting isomer obtained as a white gum is designated ENT-1 (C56). Yield: 114 mg; this material contained residual ethanol. LCMS M/z 597.1[ M + H ]]+. Retention time 4.40 min (column: Chiral Technologies Chiral cel OD-3, 4.6 x 100mm, 3 μm; mobile phase a: carbon dioxide; mobile phase B: 2-propanol with 0.05% diethylamine; gradient: 5% to 40% B over 4.5 min, then held at 40% B over 2.5 min; flow rate: 2.8 ml/min).

The second eluted isomer was purified using SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 55:45 carbon dioxide/(2-propanol containing 0.1% ammonium hydroxide) ]Purification again yielded a colorless gel designated ENT-2 (C57). Yield: 50mg, 83.8. mu. mol, 26%. LCMS M/z 597.1[ M + H ]]+. Retention time 4.74 minutes (analysis conditions identical to those used for C56).

Step 4.Synthesis of 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X1(8) [ from C56]

A solution of C56(114mg, 0.191mmol) in acetonitrile (10mL) was reacted with 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a ]]Aqueous pyrimidine (0.97M, 394 μ L, 0.382mmol) and the reaction mixture was stirred at room temperature for 23 hours. More 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a ] is added]Aqueous pyrimidine solution (0.97M, 394 μ L, 0.382mmol) and stirring continued for 6 hours, whereupon the pH was carefully adjusted to 7 to 8 with the addition of 1M hydrochloric acid. After removal of volatiles in vacuo, purification was performed using reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase A: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 30% to 50% B) to give 8 as a white solid. Yield: 22.2mg, 38.1. mu. mol, 20%. LCMS M/z 583.3[ M + H ] ]+1H NMR (400MHz, methanol-d)4): δ 8.19(d, J ═ 1.4Hz,1H),7.94(dd, J ═ 8.4,1.5Hz,1H),7.77(dd, J ═ 7.7,7.7Hz,1H),7.64(dd, J ═ 10.6,1.6Hz,1H),7.58(d, J ═ 8.4Hz,1H),7.57(dd, J ═ 8.0,1.5Hz,1H),6.81-6.75(m,1H),6.75-6.68(m,2H),5.34-5.25(m,1 ddh), 4.73 (J ═ 15.3,3.0Hz,1H),4.67-4.59(m,1H), 4.9 (dt, J ═ 9.2,6.0, 1H), four-fold (J ═ 15.3,3, 3.0Hz,1H),4.67-4.59(m,1H),4.49(dt, J ═ 9.2,6.0, 1H), four-fold (J ═ 6.96, 1H), and four-fold (J ═ peak (1H)AB=13.7Hz,ΔνAB=41.2Hz,2H),3.06(br d,J=11Hz,1H),2.95(br d,J=11Hz,1H),2.87-2.76(m,1H),2.71(tt,J=12.0,3.9Hz,1H),2.61-2.50(m,1H),2.36-2.21(m,2H),2.06(s,3H),1.95-1.72(m,4H)。

Step 5.2 Synthesis of- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2(9) [ from C57]

A solution of C57(50mg, 84. mu. mol) in acetonitrile (10mL) was treated with 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a ]]Aqueous pyrimidine (0.97M; 173. mu.L, 0.168mmol) was treated. The reaction was stirred at room temperature (about 25 ℃) for 16 hours, followed by the addition of an additional amount of 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a]Aqueous pyrimidine (0.97M; 173. mu.L, 0.168mmol) and stirring was continued at 25 ℃ for 29 h. The reaction mixture was then carefully adjusted to pH 7 to 8 with the addition of 1M hydrochloric acid; the resulting mixture was concentrated in vacuo and subjected to reverse phase HPLC (column: Xtimate)TMC18,5 μm; mobile phase A: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 27% to 67% of B) to give 9 as a white solid. Yield: 18.0mg, 30.9. mu. mol, 37%. LCMS M/z 583.3[ M + H ] ]+1H NMR (400MHz, methanol-d)4) δ 8.36-8.33(m,1H),7.97(dd, J ═ 8.5,1.5Hz,1H),7.78(dd, J ═ 7.7,7.7Hz,1H),7.70-7.63(m,2H),7.57(dd, J ═ 8.0,1.5Hz,1H),6.83-6.76(m,1H),6.76-6.71(m,2H),5.34-5.25(m,1H),4.95-4.85(m,1H, assumed value; partially masked by water peak), 4.73(dd, fraction of ABX pattern, J ═ 15.3,2.7Hz,1H),4.68-4.60(m,1H),4.50(dt, J ═ 9.2,6.0Hz,1H),4.02(AB quartet, J), JAB=13.8Hz,ΔνAB=48.2Hz,2H),3.13(br d,J=11Hz,1H),3.01(br d,J=11.5Hz,1H),2.89-2.78(m,1H),2.78-2.68(m,1H),2.60-2.50(m,1H),2.45-2.30(m,2H),2.07(br s,3H),2.00-1.86(m,2H),1.83(m,2H)。

Example 10

2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2(10) [ from P9]

Step 1.Synthesis of 5-chloro-2- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] pyridine, ENT-X2, P-toluenesulfonate (C58) [ from P9]

A solution of P9(228mg, 0.529mmol) in ethyl acetate (2.7mL) was treated with P-toluenesulfonic acid monohydrate (116mg, 0.610mmol) and the reaction mixture was taken up at 50Heating at deg.C for 16 hr. It was then allowed to stir at room temperature overnight, whereupon the precipitate was collected by filtration and washed with a mixture of ethyl acetate and heptane (1:1, 2X 20mL) to give C58 as a white solid. Yield: 227mg, 0.451mmol, 85%. LCMS M/z 331.0 [. M + H ] ]+1H NMR(400MHz,DMSO-d6) δ 8.73(d, J ═ 2.4Hz,1H),8.61-8.46(br m,1H),8.35-8.18(br m,1H),8.02(dd, J ═ 8.5,2.5Hz,1H),7.64(d, J ═ 8.5Hz,1H),7.47(d, J ═ 7.8,2H),7.11(d, J ═ 7.8Hz,2H),6.89-6.81(m,2H),6.72(pentet, J ═ 4.0Hz,1H),3.45-3.27(m,2H, assumed values; partially masked by water peaks), 3.10-2.91(m,3H),2.28(s,3H),2.02(s,3H),1.97-1.80(m, 4H).

Step 2.Synthesis of methyl 2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate, DIAST-Y2(C59) [ from P9]

N, N-diisopropylethylamine (0.234mL, 1.34mmol) was added to a solution of C58(225mg, 0.447mmol) in acetonitrile (2.2 mL). After the mixture was stirred at 45 ℃ for 5 minutes, P15(120mg, 0.407mmol) was added and stirring continued at 45 ℃ for 16 hours, followed by the addition of P15(11mg, 37. mu. mol). After stirring for an additional 3 hours, the reaction mixture was treated with water (2.5mL) and allowed to cool to room temperature. More water (5mL) was added and the resulting slurry was stirred for 2 hours whereupon the solid was collected via filtration and washed with a mixture of acetonitrile and water (15:85, 3 x 5mL) to give C59(252mg) as an off-white solid. To be provided with 1This material, analyzed by H NMR, contained some N, N-diisopropylethylamine and was taken directly in the following step. LCMS M/z589.1 [. M + H ]]+1H NMR (400MHz, chloroform-d) 8.61(d, J ═ 2.3Hz,1H),8.18(d, J ═ 1.5Hz,1H),7.96(dd, J ═ 8.5,1.5Hz,1H),7.74(d, J ═ 8.5Hz,1H),7.67(dd, a component of the ABX pattern, J ═ 8.4,2.4Hz,1H),7.59-7.51(m,1H),6.82-6.75(m,1H),6.74-6.66(m,2H),5.28-5.19(m,1 ddh), 4.75 (a component of the ABX pattern, J ═ 15.3,6.0Hz,1H),4.68(dd, a component of the ABX pattern, J ═ 15.3, 4.3, 4.81H, 4.95H, 3, 9.4.4.4H, 9H, 1H, 9.53 (1H, 2H, 1H), 3-2H, 9.4.4.4.4.4.4H, 9H, 1H, 9.53 (1H, 2H, 1H, 9H, 2H, 1H, 2H, and 4H, 1H) 2.37-2.22(m,2H),2.05(s,3H),1.93-1.74(m,4H)。

Step 3.2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, Synthesis of DIAST-X2(10) [ from P9]

A suspension of C59 (from previous step; 250mg,. ltoreq.0.407 mmol) in methanol (2mL) was heated to 40 ℃ and aqueous sodium hydroxide (1M; 0.81mL, 0.81mmol) was then added. After 17 hours, the reaction mixture was allowed to cool to room temperature and the pH was adjusted to 5 to 6 with 1M aqueous citric acid. The resulting mixture was diluted with water (2mL), stirred for 2 hours and extracted with ethyl acetate (3 × 5 mL); the combined organic layers were washed with saturated aqueous sodium chloride (5mL), dried over sodium sulfate, filtered and concentrated in vacuo to afford a foamy solid. This material was dissolved in a mixture of ethyl acetate and heptane (1:1, 4mL), heated to 50 ℃, then allowed to cool and stir overnight. To give 10 as a white solid. Yield: 179mg, 0.311mmol, in 2 steps to yield 76%. LCMS M/z 575.1 [. M + H ] ]+1H NMR(400MHz,DMSO-d6) δ 12.73 (brs, 1H),8.71(d, J ═ 2.5Hz,1H),8.27(d, J ═ 1.5Hz,1H),8.00(dd, J ═ 8.5,2.5Hz,1H),7.80(dd, J ═ 8.4,1.6Hz,1H),7.64(d, J ═ 8.4Hz,1H),7.60(d, J ═ 8.5Hz,1H),6.83-6.72(m,3H),5.14-5.06(m,1H),4.77(dd, the component of ABX pattern, J ═ 15.2,7.2Hz,1H),4.63(dd, the component of ABX pattern, J ═ 15.2,2.8, 1H),4.50-4.42 (J ═ 15.5, 7.5 Hz,1H), 1H, 4.42 (dd, 5.5H), 7.5H, 1H), 7.5H, 1H, and 7.5HAB=13.6Hz,ΔνAB=71.5Hz,2H),3.01(br d,J=11.2Hz,1H),2.85(br d,J=11.2Hz,1H),2.74-2.57(m,2H),2.47-2.38(m,1H),2.29-2.10(m,2H),2.01(s,3H),1.81-1.64(m,4H)。

Example 10 Synthesis of a 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt

Synthesis of 2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt, DIAST-X2(10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt) [ from P9]

A mixture of 10(1.54g, 2.68mmol) in tetrahydrofuran (10mL) was treated with aqueous 2-amino-2- (hydroxymethyl) propane-1, 3-diol (tris (hydroxymethyl) aminomethane, 1.0M; 2.81mL, 2.81 mmol). After 24 h, the reaction mixture was concentrated in vacuo with ethanol (2 × 50 mL). The residue was treated with ethanol (15 mL). After stirring for 20 hours, the solid was collected via filtration and washed with cold ethanol (5mL) to give 10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt as a white solid. Yield: 1.41g, 2.03mmol, 76%. LCMS M/z 575.3 [. M + H ] ]+1H NMR(600MHz,DMSO-d6) δ 8.71(d, J ═ 2.5Hz,1H),8.21(br s,1H),8.00(dd, J ═ 8.5,2.5Hz,1H),7.79(br d, J ═ 8.4Hz,1H),7.60(d, J ═ 8.5Hz,1H),7.57(d, J ═ 8.4Hz,1H),6.82-6.73(m,3H),5.13-5.07(m,1H),4.74(dd, J ═ 15.3,7.2Hz,1H),4.61(dd, J ═ 15.3,2.9Hz,1H),4.49-4.43(m,1H),4.37(d, J ═ 9.0,5.9, 3.3, 2.9Hz,1H),4.49-4.43(m,1H),4.37(d, J ═ 9.0,5.9, 3.3H, 2.9Hz,1H), 13.3H, 13-5H, 13 (d, 3H), 3H, 1H, 13-5H, 3H, 1H, 3H, 1H, 3H, 1H, 3H, 1H, 3H, and 1H, 1H) 2.01(s,3H),1.82-1.66(m, 4H). mp 184 ℃ to 190 ℃.

Example 10 acquisition of powder X-ray diffraction (PXRD) data for type A of the 1, 3-dihydroxy-2- (hydroxymethyl) propane-2-aminium salt (also known as type A of the anhydrous tris (hydroxymethyl) aminomethane salt of the compound of example 10)

The white solid of tris (hydroxymethyl) aminomethane salt of example 10 was submitted for PXRD analysis and found to be a crystalline material (designated as form a). Powder X-ray diffraction analysis was performed using a Bruker AXS D8 Endeavor diffractometer equipped with a Cu radiation source. The divergent slit is set at 15mm continuous illumination. The diffracted radiation was detected with a PSD-Lynx Eye detector with the detector PSD set at 2.99 degrees on. The X-ray tube voltage and current amount were set to 40kV and 40mA, respectively. Using a step size of 0.01 degrees and a step time of 1.0 second in a theta-theta goniometer of 3.0 to 40.02 theta degrees at a Cu wavelength Data is collected. The anti-scatter screen is set to a fixed distance of 1.5 mm. The sample was rotated during data collection. The sample was prepared by placing it in a silicon low background sample holder and rotated during collection. Data were collected using Bruker DIFFRAC Plus software and analysis was performed with EVA DIFFRAC Plus software. The PXRD data file is not processed until peak search. Preliminary peak assignment was performed using a peak search algorithm in the EVA software, using peaks selected to have a threshold value of 1. In order to ensure the effectiveness, the adjustment is performed manually; to visually inspect the output of the automated dispense and adjust the peak position to the maximum peak value. Peaks with a relative intensity of 3% or more are generally selected. Peaks that are unresolved or consistent with noise are generally not selected. Typical errors associated with peak positions of PXRD are specified in USP as at most +/-0.2 deg. 2 theta (USP-941). One diffraction pattern was consistently observed and provided in fig. 25. A list of diffraction peaks expressed in degrees 2 θ above and having a relative intensity of ≧ 3.0% is provided in Table X2.

TABLE X2

One embodiment provides a crystalline form of the anhydrous tris (hydroxymethyl) aminomethane salt of 2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2. In other embodiments, the crystalline form of the anhydrous (anhydrous) tris (hydroxymethyl) aminomethane salt of 2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid, DIAST-X2, is designated "form a" and is characterized by its unique solid state properties (such as substantially as depicted in fig. 25) associated with powder X-ray diffraction (PXRD) as described, for example, herein. In some embodiments, the type a phenotype comprises at least two phenotype selected from the group consisting of at 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; 15.7 +/-0.2 degrees; and 17.6 ± 0.2 ° characteristic peaks expressed in 2 θ. In some embodiments, the type a phenotype comprises at least three phenotype selected from the group consisting of at 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; 15.7 +/-0.2 degrees; and 17.6 ± 0.2 ° characteristic peaks expressed in 2 θ. In some embodiments, the type a phenotype comprises a phenotype selected from the group consisting of 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; 15.7 +/-0.2 degrees; and 17.6 ± 0.2 ° characteristic peaks expressed in 2 θ.

In some embodiments, form I exhibits a powder X-ray diffraction pattern comprising characteristic peaks expressed in 2 Θ at 7.7 ± 0.2 ° and 17.6 ± 0.2 °.

In some embodiments, the type a phenotype is comprised at 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; and 17.6 ± 0.2 ° peaks expressed in 2 θ.

In some embodiments, form I expression comprises at 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; and a powder X-ray diffraction pattern of peaks expressed in 2. theta. at 15.7. + -. 0.2 ℃.

In some embodiments, form I expression comprises at 7.7 ± 0.2 °; 15.2 +/-0.2 degrees; 15.7 +/-0.2 degrees; and 17.6 ± 0.2 ° peaks expressed in 2 θ.

In some embodiments, form a exhibits a powder X-ray diffraction pattern substantially as shown in figure 25. A list of diffraction peaks expressed in degrees 2 θ above and having a relative intensity of ≧ 3.0% is provided in Table X2.

As is well known in the art of powder diffraction, the relative intensities (reflections) of the peaks may vary depending on the sample preparation technique, the sample installation procedure, and the particular instrument used. Furthermore, instrumental variations and other factors may affect the 2 θ value. Thus, the XRPD peak assignments may vary by about + or-0.2 °.

Example 11

1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1H-benzimidazole-6-carboxylic acid, formate (11)

This entire synthesis procedure was performed in library form.

Step 1.1 Synthesis of methyl 1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1H-benzimidazole-6-carboxylate (C60)

A mixture of P14(44mg, 100. mu. mol) and 3-ethynylpyridine (21mg, 200. mu. mol) in toluene (800. mu.L) was treated with sodium bicarbonate (100. mu. mol), followed by the addition of triruthenium dodecacarbonyl (6mg, 9. mu. mol). The reaction vial was then capped and shaken at 120 ℃ for 16 hours. Use ofThe concentrator removed the solvent to afford C60, which was taken directly to the next step.

Step 2.1 Synthesis of 1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1H-benzimidazole-6-carboxylic acid, Formate (11)

Aqueous sodium hydroxide (1.0M; 200. mu.L, 200. mu. mol) was added to a solution of C60 (from the previous step,. ltoreq.100. mu. mol) in a mixture of methanol (400. mu.L) and tetrahydrofuran (400. mu.L). The reaction vial was capped and shaken at 80 ℃ for 16 hours, whereupon the reaction mixture was used The concentrator was evaporated and purified using reverse phase HPLC (column: Agela Durashell C18, 5 μm; mobile phase A: 0.225% formic acid in water; mobile phase B: acetonitrile; gradient: 12% to 52% B) to give 11. Yield: 2.2mg, 4.2. mu. mol, 4% in 2 steps. LCMS M/z 529[ M + H]+. Retention time: 2.47 minutes (column: Waters Xbridge C18, 2.1X 50mm, 5 μm; mobile phase A: 0.0375% trifluoroacetic acid in water; mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; gradient: 1% to 5% B over 0.6 minutes;5% to 100% of B over 3.4 minutes; flow rate: 0.8 ml/min).

Example 12

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [2- (dimethylamino) ethyl ] -1H-benzimidazole-6-carboxylic acid (12)

This entire synthesis procedure was performed in library form.

Step 1.3 Synthesis of methyl 3- { [2- (dimethylamino) ethyl ] amino } -4-nitrobenzoate (C61)

Methyl 3-fluoro-4-nitrobenzoate (0.2M solution in N, N-dimethylformamide; 1mL, 200. mu. mol) was treated with N, N-dimethylethane-1, 2-diamine (18mg, 200. mu. mol) and N, N-diisopropylethylamine (78mg, 600. mu. mol). The reaction vial was then capped and shaken at 50 ℃ for 16 hours, whereupon the reaction mixture was used The concentrator was evaporated to give C61. This material was taken directly to the next step.

Step 2.4 Synthesis of methyl amino-3- { [2- (dimethylamino) ethyl ] amino } benzoate (C62)

The zinc powder was activated with dilute hydrochloric acid. Methanol (2mL) was added to C61 (from previous step,. ltoreq.200. mu. mol), followed by aqueous calcium chloride (1.0M; 200. mu.L, 200. mu. mol) and activated zinc powder (130mg, 2.0 mmol). The reaction vial was capped and shaken at 70 ℃ for 16 hours, whereupon the reaction mixture was filtered. Using the filtrateThe concentrator was concentrated, and the residue was dissolved in water (2mL) and then extracted with ethyl acetate (2X 3 mL). The combined organic layers were usedThe concentrator was evaporated to give C62 (estimated 150. mu. mol), which was used directly in the next step.

Step 3.4 Synthesis of methyl- [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetyl) amino ] -3- { [2- (dimethylamino) ethyl ] amino } benzoate (C63)

Compound P10(41mg, 100. mu. mol) was added to C62 (from previous step, ca. 150. mu. mol) and the mixture was treated with 2-hydroxypyridine 1-oxide and 1- [3- (dimethylamino) propyl]Solutions of 3-ethylcarbodiimide hydrochloride in N, N-dimethylacetamide (0.1M each; 1mL each, 100. mu. mol) were treated. N, N-diisopropylethylamine (39mg, 300. mu. mol) was then added, the reaction vial was capped and shaken at 50 ℃ for 16 h. Then the reaction mixture was used The concentrator was concentrated and purified using preparative thin layer chromatography to afford C63, which proceeded directly to the following step.

Step 4.2 Synthesis of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [2- (dimethylamino) ethyl ] -1H-benzimidazole-6-carboxylate (C64)

A mixture of acetic acid (500. mu.L) and C63 (. ltoreq.100. mu. mol from the previous step) was shaken in a closed vial at 150 ℃ for 2 hours, whereupon the reaction mixture was usedThe concentrator evaporates. The resulting C64 proceeded directly to the following step.

Step 5.2 Synthesis of 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [2- (dimethylamino) ethyl ] -1H-benzimidazole-6-carboxylic acid (12)

A solution of C64 (from previous step,. ltoreq.100. mu. mol) in ethanol (500. mu.L) was treated with aqueous lithium hydroxide (2.0M; 500. mu.L, 1mmol) and the reaction mixture was shaken for 2 hours at 50 ℃ in a sealed vial. After the pH of the mixture was adjusted to 7 by addition of 1.0M hydrochloric acid, the mixture was concentratedObtaining a mixture for useThe concentrator was concentrated and then purified via reverse phase HPLC [ column: agela Durashell C18, 5 μm; mobile phase A: ammonium hydroxide in water (pH 10); mobile phase B: acetonitrile; gradient: 25% to 65% of B ]To obtain 12. Yield: 7.0mg, 12. mu. mol, 12% in 3 steps. LCMS M/z 593[ M + H ]]+. Retention time: 2.45 minutes (column: Waters Xbridge C18, 2.1X 50mm, 5 μm; mobile phase A: 0.0375% trifluoroacetic acid in water; mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; gradient: 10% to 100% B over 4.0 minutes; flow rate: 0.8 ml/min).

Example 13

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid (13)

Step 1.6- [ (1, 3-oxazol-2-ylmethyl) amino ] -5-nitropyridine-2-carboxylic acid methyl ester (C65) Synthesis

Triethylamine (532mg, 5.26mmol) was added to a suspension of 1- (1, 3-oxazol-2-yl) methylamine, hydrochloride salt (236mg, 1.75mmol) and methyl 6-chloro-5-nitropyridine-2-carboxylate (386mg, 1.78mmol) in tetrahydrofuran (5 mL). After the reaction mixture was stirred at 25 ℃ for 14 hours, it was poured into water (30mL) and extracted with dichloromethane (2X 50 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo; chromatography on silica gel (gradient: 0% to 5% methanol in dichloromethane) afforded C65 as a yellow solid. Yield: 310mg, 1.11mmol, 63%. LCMS M/z 278.7[ M + H ] ]+1H NMR (400MHz, chloroform-d) delta 8.69-8.61(br m,1H),8.58(d,J=8.4Hz,1H),7.65(d,J=0.8Hz,1H),7.46(d,J=8.4Hz,1H),7.11(d,J=1.0Hz,1H),5.07(d,J=5.3Hz,2H),3.97(s,3H)。

The remainder of the synthesis procedure is performed in library form.

Step 2.5 Synthesis of amino-6- [ (1, 3-oxazol-2-ylmethyl) amino ] pyridine-2-carboxylic acid methyl ester (C66)

Aqueous ammonium chloride (5.0M; 400. mu.L, 2.0mmol) followed by activated zinc (131mg, 2.0mmol) was added to a solution of C65(56mg, 200. mu. mol) in methanol (2.0 mL). The reaction vial was then capped and shaken at 30 ℃ for 16 hours, whereupon the reaction mixture was filtered. Using the filtrateConcentrator concentrated, then mixed with water (1.0mL) and extracted with dichloromethane (3 × 1.0 mL); the combined organic layers were usedThe concentrator was evaporated to afford C66, which was taken directly to the next step.

Step 3.5 Synthesis of methyl [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetyl) amino ] -6- [ (1, 3-oxazol-2-ylmethyl) amino ] pyridine-2-carboxylate (C67)

A mixture of P10(81mg, 200. mu. mol) and C66 (from the previous step,. ltoreq.200. mu. mol) was mixed with N, N-dimethylacetamide and then treated with N, N-diisopropylethylamine (100. mu.L, 600. mu. mol). Adding 1- [3- (dimethylamino) propyl group contained in N, N-dimethylacetamide ](1.0mL) of a solution of (0.24M) 3-ethylcarbodiimide hydrochloride and (0.1M) 2-hydroxypyridine 1-oxide (240. mu. mol 1- [3- (dimethylamino) propyl ] amine]3-ethylcarbodiimide hydrochloride and 100. mu. mol 2-hydroxypyridine 1-oxide), the reaction vial was capped and shaken at 50 ℃ for 16 hours. Then useThe concentrator removed volatiles and the residue was subjected to preparative thin layer chromatography to give C67,it proceeds directly to the next step.

Step 4.2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid methyl ester (C68)

A mixture of acetic acid (1.0mL) and C67 (from the previous step,. ltoreq.200. mu. mol) was shaken at 150 ℃ for 2 hours, whereupon the reaction mixture was usedThe concentrator evaporates. The resulting C68 was used directly in the following step.

Step 5.2 Synthesis of- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3H-imidazo [4,5-b ] pyridine-5-carboxylic acid (13)

Aqueous lithium hydroxide (2.0M; 1.0mL, 2.0mmol) was added to a mixture of C68 (from the previous step,. ltoreq.200. mu. mol) in tetrahydrofuran (1.0 mL). After addition of methanol (500 μ L), the reaction vial was capped and shaken at 50 ℃ for 16 hours. In use After the concentrator removed volatiles, dimethyl sulfoxide (1.0mL) was added and the pH was adjusted to 7 to 8 with concentrated hydrochloric acid. The resulting mixture was purified using reverse phase HPLC [ column: agela Durashell C18, 5 μm; mobile phase A: ammonium hydroxide in water (pH 10); mobile phase B: acetonitrile; gradient: 24% to 64% of B]To obtain 13. Yield: 3.9mg, 6.5. mu. mol, 3% in 4 steps. LCMS M/z 604[ M + H ]]+. Retention time: 3.14 minutes (column: Waters Xbridge C18, 2.1X 50mm, 5 μm; mobile phase A: 0.0375% trifluoroacetic acid in water; mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; gradient: 1% to 5% B over 0.6 minutes; 5% to 100% B over 3.4 minutes; flow rate: 0.8 ml/min).

Example 14

2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1-methyl-1H-benzimidazole-6-carboxylic acid (14)

Step 1.2 Synthesis of methyl 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1-methyl-1H-benzimidazole-6-carboxylate (C69)

N, N-diisopropylethylamine (683. mu.L, 3.92mmol) was added to a mixture of P3(680mg, 1.31mmol) in acetonitrile (5.2 mL); this was allowed to stir at 45 ℃ for 5 minutes, followed by the addition of P16(319mg, 1.34 mmol). Stirring was continued for 2.75 hours at 45 ℃ then water (6mL) was added and the reaction mixture was allowed to cool to room temperature and stirred for 30 minutes. The solid was collected via filtration and washed with a mixture of acetonitrile and water (1:4, 3 × 5mL) to give C69 as a white solid. Yield: 635mg, 1.15mmol, 88%. LCMS M/z 550.1 [. sup. + H ] ]+1H NMR (400MHz, chloroform-d) δ 8.15-8.12(m,1H),7.97(dd, J ═ 8.5,1.6Hz,1H),7.74(d, J ═ 8.5Hz,1H),7.50(dd, J ═ 8.2,8.2Hz,1H),7.16-7.07(m,2H),6.79-6.73(m,1H),6.72-6.65(m,2H),3.98(s,3H),3.96(s,3H),3.88(s,2H),3.04-2.93(m,2H),2.76-2.66(m,1H),2.37-2.25(m,2H),2.04(br, 3H),1.89-1.78(m, 4H).

Step 2.2 Synthesis of 2- ({4- [ (2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1-methyl-1H-benzimidazole-6-carboxylic acid (14)

A mixture of C69(600mg, 1.09mmol) in methanol (11mL) was heated to 45 deg.C and then treated with aqueous sodium hydroxide (1M; 2.2mL, 2.2 mmol). After 24 hours, the reaction mixture was adjusted to pH 5 to 6 via addition of aqueous citric acid (1M; 1.1mL) and then diluted with water (10 mL). The resulting mixture was allowed to cool to room temperature and stirred for 1 hour, whereupon the precipitated solid was collected by filtration and washed with a mixture of methanol and water (1: 4; 3X 5 mL). This gave 14 as a white solid. Yield: 535mg, 0.998mmol, 92%. LCMS M/z 536.1 [. multidot.M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.16(d,J=1.5Hz,1H),7.81(dd,J=8.4,1.6Hz,1H),7.64(d,J=8.4Hz,1H),7.59-7.52(m,2H),7.33(dd,J=8.3,2.1Hz,1H),6.81-6.70(m,3H),3.94(s,3H),3.84(s,2H),3.01-2.91(m,2H),2.70-2.59(m,1H),2.28-2.16(m,2H),2.02(s,3H),1.73(m,4H)。

Examples 15 and 16

2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate (15) [ from P18, via C71 ]; and 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, DIAST-X2, trifluoroacetate (16) [ from P18, via C72]

Step 1.2 Synthesis of methyl 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylate (C70) [ from P18]

A mixture of P18(240mg, 0.699mmol), C4(275mg, 0.800mmol), cesium carbonate (455mg, 1.40mmol), tris (dibenzylideneacetone) dipalladium (0) (40.0mg, 43.7. mu. mol) and 1,1 '-binaphthyl-2, 2' -diylbis (diphenylphospholane) (BINAP; 52.2mg, 83.8. mu. mol) in toluene (5mL) was degassed with nitrogen for 5 minutes and then stirred at 90 ℃ for 16 hours. The reaction mixture was filtered and the filtrate was concentrated in vacuo; preparative thin-layer chromatography (eluent: 1:1 petroleum ether/ethyl acetate) gave C70, a mixture of diastereomers as a yellow oil. Yield: 165mg, 0.272mmol, 39%. LCMS M/z 628.1 [. multidot.M + Na ]+]。

Step 2.2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, DIAST-Y1(C71) [ from P18 ]; and isolation of 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, DIAST-Y2(C72) [ from P18]

Separation of stereoisomers on C70(165mg, 0.272mmol) dioxolane using SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65:35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide). The first eluting isomer was designated DIAST-Y1(C71) and the second eluting isomer was designated DIAST-Y2 (C72); both separated were white solids.

Yield of C71: 55.0mg, 90.7. mu. mol, 33%. LCMS M/z 605.9 [. M + H ]]+. Retention time 4.47 min (column: Chiral Technologies Chiralpak AD-3, 4.6 x 100mm, 3 μm; mobile phase a: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% to 40% B over 4.5 min, then held at 40% B over 2.5 min; flow rate: 2.8 ml/min).

Yield of C72: 58.0mg, 95.7. mu. mol, 35%. LCMS M/z 628.0 [. M + Na ]+]. Retention time 4.88 minutes (analytical conditions were the same as those for C71).

Step 3.2 Synthesis of- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate (15) [ from P18, Via C71]

Aqueous sodium hydroxide (3M; 1.0mL, 3.0mmol) was added to a solution of C71(55.0mg, 90.7. mu. mol) in a mixture of methanol (2.0mL) and tetrahydrofuran (1.0 mL). After the reaction mixture was stirred at 20 ℃ for 2 hours, the pH was adjusted to 7 by addition of 1M hydrochloric acid and the resulting mixture was extracted with a mixture of dichloromethane and methanol (10:1, 3 × 30 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo. Reverse phase HPLC (column: Boston Green ODS, 5 μm; mobile phase A: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile; gradient: 10% to 95% B) gave 15 as a white solid. Yield: 35.8mg, 50.7. mu. mol, 56%. LCMS M/z 592.3 [. M + H ]]+1H NMR (400MHz, methanol-d)4)δ8.46(s,1H),8.21(d,J=8.6Hz,1H),7.78(d,J=8.6Hz,1H),7.54(dd, J ═ 8.3,8.3Hz,1H),7.16-7.08(m,2H),6.76(dd, J ═ 8.2,8.1Hz,1H),6.55-6.47(m,2H),4.9-4.70(m,2H, assumed values; partially obscured by a water peak), 3.82(t, J ═ 4.9Hz,2H),3.66-3.56(m,1H),3.50-3.41(m,1H),3.19-3.09(m,1H),3.15(s,3H),3.08-2.99(m,1H),2.63-2.57(m,1H),2.27-2.17(m,1H),2.01(s,3H),1.76-1.66(m,2H),1.62-1.50(m,2H),1.35-1.26(m, 1H).

Step 4.Synthesis of 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] -6-azaspiro [2.5] oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid, DIAST-X2, trifluoroacetate (16) [ from P18, Via C72]

Aqueous sodium hydroxide (3M; 1.0mL, 3.0mmol) was added to a solution of C72(58.0mg, 95.7. mu. mol) in a mixture of methanol (2.0mL) and tetrahydrofuran (1.0 mL). After the reaction mixture was stirred at 20 ℃ for 2 hours, the pH was adjusted to 7 by addition of 1M hydrochloric acid and the resulting mixture was extracted with a mixture of dichloromethane and methanol (10:1, 3 × 30 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo. Reverse phase HPLC (column: Boston Green ODS,5 μm; mobile phase A: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile; gradient: 35% to 95% B) gave 16 as a white solid. Yield: 33.4mg, 47.3. mu. mol, 49%. LCMS M/z 592.2 [. M + H ]]+1H NMR (400MHz, methanol-d)4) δ 8.53-8.50(m,1H),8.25(dd, J ═ 8.6,1.4Hz,1H),7.80(br d, J ═ 8.6Hz,1H),7.57(dd, J ═ 8.4,8.2Hz,1H),7.25(dd, J ═ 10.8,2.0Hz,1H),7.19(br dd, J ═ 8.4,2.1Hz,1H),6.77(dd, J ═ 8.2,8.1Hz,1H),6.55-6.50(m,2H),4.9-4.72(m,2H, assumed values; partially masked by water peaks), 3.93-3.80(m,2H),3.68-3.58(m,1H),3.41-3.3(m,1H, assumed values; partially obscured by solvent peaks), 3.25(s,3H),3.22-3.12(m,1H),3.07-2.97(m,1H),2.67(dd, J ═ 8.3,5.8Hz,1H),2.28-2.17(m,1H),2.01(d, J ═ 1.0Hz,3H),1.86-1.71(m,2H),1.69-1.56(m,2H),1.36-1.26(m, 1H).

Examples 17 and 18

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid ammonium salt, ENT-1(17) and 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid Ammonium, ENT-2(18)

Step 1.Synthesis of methyl 4- [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } acetyl) amino ] -3- { [ (1-ethyl-1H-imidazol-5-yl) methyl ] amino } benzoate (C73)

O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethylurea hexafluorophosphate (566mg, 1.49mmol) was added to a mixture of P19(340mg, 1.24mmol) in N, N-dimethylformamide (10mL), and the mixture was stirred at 25 ℃ for 10 minutes. A solution of P10(503mg, 1.24mmol) and N, N-diisopropylethylamine (615. mu.L, 3.53mmol) in N, N-dimethylformamide (7.7mL) was then added and the reaction mixture was stirred at 25 ℃ for 16 h, whereupon it was poured into water (10mL) and extracted with ethyl acetate (3X 50 mL). The combined organic layers were washed sequentially with aqueous ammonium chloride (3 × 20mL) and saturated aqueous sodium chloride (2 × 20mL), dried over sodium sulfate, filtered and concentrated in vacuo. When purified using silica gel chromatography (gradient: 0% to 5% methanol in ethyl acetate), C73 was obtained as a light brown gum. Yield: 316mg, 0.477mmol, 38%. LCMS M/z 662.2 [. M + H ] ]+

Step 2.2 Synthesis of methyl 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylate (C74)

A solution of C73(316mg, 0.477mmol) in acetic acid (14mL) was stirred at 55 ℃ for 16 h. The solvent was removed under high vacuum and the residue was purified using preparative thin layer chromatography (eluent: 10:1 dichloromethane/methanol) to give C74 as a colorless oil. Yield: 200mg, 0.310mmol, 65%. LCMS M/z 644.3 [. sup. + H ]]+

Step 3.2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6-carboxylic acid ammonium salt, ENT-1(17) and 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl ] -1H-benzimidazole-6- Synthesis of ammonium carboxylates, ENT-2(18)

A mixture of C74(150mg, 0.233mmol) and aqueous sodium hydroxide (2M; 233. mu.L, 0.466mmol) in a mixture of methanol (3mL) and tetrahydrofuran (3mL) was stirred at 45 ℃ for 16 h. After the reaction mixture was adjusted to pH 7 with the addition of 1M hydrochloric acid, it was concentrated in vacuo to give a mixture of 17 and 18. The enantiomers were separated by passage through SFC [ column: chiral Technologies Chiral cel OD, 10 μm; mobile phase: 1:1 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ]. Designating the first eluted enantiomer as ENT-1(17) and the second eluted enantiomer as ENT-2 (18); both separated were white solids.

Yield of 17: 45.0mg, 69.5. mu. mol, 30%. LCMS M/z 630.3 [. M + H ]]+1H NMR (400MHz, methanol-d)4) δ 8.15(br s,1H),8.00(br d, J ═ 8.4Hz,1H),7.81(s,1H),7.72(d, J ═ 8.2Hz,1H),7.56(dd, J ═ 8.3,8.3Hz,1H),7.28(dd, J ═ 10.9,2.0Hz,1H),7.21(dd, J ═ 8.3,2.1Hz,1H),6.77(dd, components of ABC patterns, J ═ 8.0,7.7Hz,1H),6.69(dd, components of ABC patterns, J ═ 7.8,1.2Hz,1H),6.67-6.60(m,2H),5.82(s,2H),4.12(q, J ═ 7.2Hz, 1H), 89.89 (q, 2H), four peaks (89, 89H), and four peaksAB=14.3Hz,ΔνAB6.9Hz,2H),3.00-2.90(m,2H),2.74-2.64(m,1H),2.32-2.21(m,2H),2.02(s,3H),1.82-1.61(m,4H),1.29(t, J ═ 7.3Hz, 3H). Retention time 5.66 min (column: Chiral Technologies Chiralpak AD-3, 4.6 x 150mm, 3 μm; mobile phase a: carbon dioxide; mobile phase B: methanol with 0.05% diethylamine; gradient: 5% to 40% B over 5.5 min, then held at 40% B over 3.0 min; flow rate: 2.5 ml/min).

Yield of 18: 32.8mg, 50.7. mu. mol, 22%. LCMS M/z 630.3 [. M + H ]]+1H NMR (400MHz, methanol-d)4)δ8.15(s,1H) 8.00(d, J ═ 8.5Hz,1H),7.81(s,1H),7.72(d, J ═ 8.5Hz,1H),7.56(dd, J ═ 8.3,8.3Hz,1H),7.28(dd, J ═ 10.9,2.0Hz,1H),7.21(dd, J ═ 8.3,2.0Hz,1H),6.77(dd, components of ABC patterns, J ═ 7.8,7.8Hz,1H),6.69(dd, components of ABC patterns, J ═ 7.9,1.2Hz,1H),6.67-6.60(m,2H),5.82(s,2H),4.12(q, J ═ 7.3Hz,2H),3.89 (J, four-fold peak, J ═ 7.3, 2H),3.89 (J ═ 7.7.7, 1H),7.81 (AB, 1H),7.28(d, J ═ 10.9, 2H), and (c, 1H), and (c, b AB=14.1Hz,ΔνAB7.4Hz,2H),3.01-2.90(m,2H),2.74-2.63(m,1H),2.31-2.21(m,2H),2.02(s,3H),1.82-1.60(m,4H),1.29(t, J ═ 7.3Hz, 3H). Retention time 5.34 minutes (conditions for analytical SFC are the same as for 17).

The compounds listed in table 1 were prepared using procedures analogous to the examples identified in table 2 and using the appropriate intermediates identified in table 2. The compounds were purified using the methods discussed in this application. The final compound can be isolated as a neutral or acid or base salt.

TABLE 1 Structure and IUPAC name of examples 19 to 102

TABLE 2 preparation of examples 19 to 102 and physicochemical data

The area is an estimated value, and the peak is partially shielded by the water peak

The area is an estimate, the peak is partially masked by the solvent peak

Observed chlorine isotope pattern

1. Racemic methyl ester [2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester ] was purified by SFC [ column: chiral Technologies Chiral cel OD-H, 5 μm; mobile phase: 7:3 carbon dioxide/(2-propanol containing 0.1% ammonium hydroxide) ] into its component enantiomers. The first eluting enantiomer, ENT-1(C76), was used in the synthesis of example 21, and the second eluting enantiomer, ENT-2(C77), was converted to example 20. Retention time of C76: 5.72 minutes (column: Chiral Technologies Chiralpak OD-3, 4.6X 150mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol containing 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then held at 40% B over 3.0 minutes; flow rate: 2.5 ml/min). Retention time of C77: 6.01 minutes (conditions for analytical SFC were the same as for C76).

2. Methyl ester derived from the coupling of C4 with P17 (2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]-6-azaspiro [2.5]Oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester) was purified by SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65:35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]Separated into its component stereoisomers at the dioxolanes. The first eluted isomer, DIAST-1(C78) was converted to example 26; warp beam1Upon examination of the H NMR data, this material was the enantiomer of example 15. The second eluting isomer, DIAST-2(C79), was used for the synthesis of example 25; warp beam1Upon examination of the H NMR data, this material was the enantiomer of example 16. Retention time of C78: 3.60 minutes(column: Chiral Technologies Chiralpak AD-3, 4.6X 100mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% to 40% B over 4.5 minutes, then held at 40% B over 2.5 minutes; flow rate: 2.8 ml/min). Retention time of C79: 3.82 minutes (conditions for analytical SFC were the same as for C78).

4- (4-bromo-1, 3-benzodioxol-2-yl) -3-fluorobenzonitrile is prepared by: 3-fluoro-4-formylbenzonitrile and 3-bromobenzene-1, 2-diol were treated with p-toluenesulfonic acid in toluene and water was removed using a dean-Stark apparatus. This material was then reacted with [1- (tert-butoxycarbonyl) piperidin-4-yl ] (iodo) zinc in the presence of [1, 1' -bis (diphenylphosphino) ferrocene ] -dichloropalladium (II) and copper (I) iodide followed by ester cleavage using p-toluenesulfonic acid to give the requisite 3-fluoro-4- [4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] benzonitrile.

4. Conditions for analytical HPLC. Column: waters Xbridge C18, 2.1X 50mm, 5 μm; mobile phase A: 0.0375% trifluoroacetic acid in water; mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; gradient: 10% to 100% of B over 4.0 minutes; flow rate: 0.8 ml/min.

5. Conditions for analytical HPLC. Column: waters Xbridge C18, 2.1X 50mm, 5 μm; mobile phase A: 0.0375% trifluoroacetic acid in water; mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; gradient: 1% to 5% of B over 0.6 min; 5% to 100% of B over 3.4 minutes; flow rate: 0.8 ml/min.

4- [ 2-methyl-2- (pyridin-2-yl) -1, 3-benzodioxol-4-yl ] -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester was synthesized from 3-bromobenzene-1, 2-diol and 2-ethynylpyridine using the procedure described for the synthesis of C12 in preparation P7. Subsequent treatment with palladium on carbon hydride followed by hydrogen chloride in ethyl acetate gave the requisite 2- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] pyridine, hydrochloride salt.

7. Racemic methyl ester [1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-2-yl) -1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1H-benzimidazole-6-carboxylic acid methyl ester ] was purified by SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65:3 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ] into its component enantiomers. The first eluting enantiomer, ENT-1(C80), was used for the synthesis of example 90, and the second eluting enantiomer, ENT-2(C81), was converted to example 89. Retention time of C80: 4.11 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 100mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% to 40% B over 4.5 minutes, then held at 40% B over 2.5 minutes; flow rate: 2.8 ml/min). Retention time of C81: 4.62 minutes (conditions for analytical SFC were the same as for C80).

The conversion of P8 and P9 to the corresponding cyano-substituted derivatives was carried out using the method described in preparation P4 for the synthesis of P4 from P2.

9. Treatment of 1- (4-chloro-2-fluorophenyl) ethanone with trimethyl orthoformate and p-toluenesulfonic acid affords 4-chloro-1- (1, 1-dimethoxyethyl) -2-fluorobenzene, which is reacted with 3-bromo-6-fluorobenzene-1, 2-diol in the presence of p-toluenesulfonic acid to give 4-bromo-2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxole. This material was converted to the requisite tert-butyl 4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate using the procedure described for the synthesis of P1 from C2 in preparation P1.

10. Separation of stereoisomers at dioxolanes of 96 and 97 using SFC [ column: 5 μm for Chiral Technologies Chiralpak IG; mobile phase: 3:1 carbon dioxide/(2-propanol containing 0.2% ammonium hydroxide). The first eluting isoform is designated DIAST-1(96) and the second eluting isoform is designated DIAST-2 (97).

11. Conditions for analytical SFC. Column: chiral Technologies Chiral IG, 4.6 × 100mm, 5 μm; mobile phase: 7:3 carbon dioxide/(2-propanol containing 0.2% ammonium hydroxide); flow rate: 1.5 ml/min; back pressure: 150 bar.

2- (chloromethyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid tert-butyl ester was synthesized from tert-butyl 3-fluoro-4-nitrobenzoate and 1- (1, 3-oxazol-2-yl) methylamine using the method described for the synthesis of P11. Subsequent reaction with C54 was carried out using triethylamine to give 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ] piperidin-1-yl } methyl) -1- (1, 3-oxazol-2-ylmethyl) -1H-benzimidazole-6-carboxylic acid tert-butyl ester using SFC [ column: chiral Technologies Chiral cel OD-H, 5 μm; mobile phase: 55:45 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ] separated it into its component enantiomers. The first eluting enantiomer, ENT-1(C82), was used for the synthesis of 99, and the second eluting enantiomer, ENT-2(C83), was converted to 98. Retention time of C82: 1.47 min (column: Chiral Technologies Chiralpak OD-3, 4.6X 50mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: methanol with 0.05% diethylamine; gradient: 5% B over 0.2 min, followed by 5% to 40% B over 1.4 min, then held at 40% B over 1.05 min; flow rate: 4 ml/min). Retention time of C83: 1.85 minutes (conditions for analytical SFC were the same as for C82).

13. Reaction of 1-bromo-2, 3-difluoro-4-nitrobenzene in 1-methylpyrrolidin-2-one with copper (I) cyanide at elevated temperature affords 2, 3-difluoro-4-nitrobenzonitrile, which is subjected to thionyl chloride and methanol to give methyl 2, 3-difluoro-4-nitrobenzoate. This material was converted to the necessary methyl 2- (chloromethyl) -7-fluoro-1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylate by the method described in preparation P11 for the synthesis of P11 from methyl 3-fluoro-4-nitrobenzoate by using C29.

14. The requisite [ 2-phenyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] methanol was synthesized from 2-oxo-2-phenylethylacetate in a manner analogous to that described for the synthesis of C13.

Example 103

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid

Intermediate 103-0: 3-fluoro-4- (((6- (piperidin-4-yl) pyridin-2-yl) oxy) methyl) benzonitrile bis (4-methylbenzenesulfonate)

Step 1

N-butyllithium in heptane (2.6M, 250mL, 650mmol) was added over 15min to a solution of diisopropylamine (92mL, 656mmol) in THF (350mL) at-26 ℃. The mixture was cooled to-30 ℃ and a solution of piperidine-1, 4-dicarboxylic acid 1- (tert-butyl) ester 4-methyl ester (156g, 641mmol) in THF (150mL) was added over 25 min. After 10min, a solution of 2, 6-dichloropyridine (94g, 635mmol) in THF (150mL) was added over 2 min. The mixture was warmed to 25 ℃ for 2.5h, then cooled to 8 ℃ and treated with 6M HCl (125mL) for 20min to bring the pH of the mixture to-7 to 8. The mixture was diluted with water (100mL) and MTBE (150mL) and the layers were separated. The aqueous layer was extracted with MTBE (150mL) and the combined organic layers were washed with brine (150mL), MgSO 4And (5) drying. The solvent was removed under reduced pressure to give crude 4- (6-chloropyridin-2-yl) piperidine-1, 4-dicarboxylic acid 1- (tert-butyl) ester 4-methyl ester (241g) as a yellow oil, which was used in the next step without purification. Of purified samples1H NMR(400MHz,CDCl3)δ:7.62(t,1H),7.21(d,2H),3.83(br s,2H),3.71(s,3H),3.14(br s,2H),2.41(d,2H),2.08(ddd,2H),1.45(s,9H)。

Step 2

Crude 4- (6-Chloropyridin-2-yl) piperidine-1, 4-dicarboxylic acid 1- (tert-butyl) ester 4-methyl ester (241g, assumed 635mmol) was dissolved in MeOH (400mL) at 43 deg.C and treated with 4M aq NaOH (300mL) for 20 min. The mixture was warmed to 50 ℃ and stirred for 35 min. The mixture was then cooled to 11 ℃ and the pH was adjusted to-2 with the addition of 6M HCl (200mL) over 25min while continuing to cool to 5 ℃, whereupon a solid precipitate formed. The slurry was diluted with water (300mL) and stirred for 40min, then the solid was collected by filtration, washed with water, then dried under vacuum at 50 ℃ to give a white solid (224 g). The solid was wet milled in heptane (750mL) at 45 ℃ for 45 min. The mixture was cooled to 16 ℃ and the solid collected by filtration, washed with heptane and dried to give 1- (tert-butoxycarbonyl) -4- (6-chloropyridin-2-yl) piperidine-4-carboxylic acid (187g, 549mmol, 86% over two steps) as a white solid.

Step 3

A solution of 1- (tert-butoxycarbonyl) -4- (6-chloropyridin-2-yl) piperidine-4-carboxylic acid (187g, 549mmol) in DCE (900mL) was heated at 82 deg.C overnight and then cooled to 20 deg.C. Mixing the mixture with (30g) The treatment is carried out for 40 min. Passing the slurry through(30g) The pad was filtered and the solid was washed with 1:1 MTBE: heptane (300 mL). The filtrate was concentrated under reduced pressure to give a pale yellow solid, which was wet-milled in heptane (250mL) at 50 ℃. The mixture was cooled to 12 ℃ and the solid was collected by filtration, washed with heptane and dried under vacuum at 45 ℃ to give tert-butyl 4- (6-chloropyridin-2-yl) piperidine-1-carboxylate (143g, 481mmol, 88%) as a solid.1H NMR(600MHz,CDCl3)δ:7.58(t,1H),7.17(d,1H),7.06(d,1H),4.25(br s,2H),2.66-2.93(m,3H),1.91(d,2H),1.69(qd,2H),1.47(s,9H)。

Step 4

Tert-butyl 4- (6-chloropyridin-2-yl) piperidine-1-carboxylate (100g, 337mmol), 3-fluoro-4- (hydroxymethyl) benzonitrile (53.9g, 357mmol) in dioxane (900mL) with Cs2CO3The mixture (170g, 522mmol) was degassed with 5 vacuum/nitrogen fill cycles. John Phos ([1, 1' -biphenyl) was added]2-yl-di-tert-butylphosphine, 2.02g, 6.77mmol) and Pd2(dba)3(3.10g, 3.39mmol) and again 2 vacuum/nitrogen fill cycles were applied. The mixture was then heated at 95 ℃ for 3 h. Additional John Phos (660mg, 2.21mmol) and Pd were added2(dba)3(990mg, 1.08mmol) and heating was continued overnight. The mixture was cooled to 20 ℃ and washed by MTBE (250mL)The pad is filtered. The filtrate was concentrated under reduced pressure to give a red-orange oil (174 g). This material was dissolved in 30% MTBE/hexane (600mL) and reacted with (20g) Andg-60(10G) was stirred for 70min and then filtered through a pad of silica (100G) rinsed with 50% MTBE/hexanes (600 mL). The filtrate was concentrated under reduced pressure and azeotroped with EtOAc (100mL) to give tert-butyl 4- (6- ((4-cyano-2-fluorobenzyl) oxy) pyridin-2-yl) piperidine-1-carboxylate (147g) as an oil, which was used without further purification. Of purified samples1H NMR(600MHz,CDCl3)δ:7.62(t,1H),7.53(t,1H),7.44(d,1H),7.37(d,1H),6.75(d,1H),6.65(d,1H),5.49(s,2H),4.20(br s,2H),2.81(br s,2H),2.70(tt,1H),1.82(d,2H),1.67(d,2H),1.49(s,9H)。

Step 5

Mixing pTSA. H2O (161g, 846mmol) was added to a stirred solution of tert-butyl 4- (6- ((4-cyano-2-fluorophenylmethyl) oxy) pyridin-2-yl) piperidine-1-carboxylate (147g, assumed value 337mmol) in EtOAc (1.8L) at RT. The mixture was heated to 60 ℃ resulting in gas evolution and solid formation. The mixture was stirred for 1.5H, followed by addition of pTSA. H2O (12g, 63mmol) and stirring was continued for 45 min. The slurry was cooled to 17 ℃ and the solid was collected by filtration, washed with EtOAc (200mL) and dried to provide 205g of solid. This material was dissolved in MeOH (500mL) at 55 deg.C and diluted with EtOAc (1L). The resulting slurry was cooled to 20 ℃ and the solid was collected by filtration, washed with 9:1 EtOAc: MeOH (100mL) and EtOAc (250mL) and dried to give intermediate 103-0 as a white solid (176.6g, 269mmol, 80% over two steps). 1H NMR(600MHz,DMSO-d6)δ:8.53(br s,1H),8.26(br s,1H),7.89(d,1H),7.67-7.78(m,3H),7.48(d,4H),7.11(d,4H),6.90(d,1H),6.79(d,1H),5.48(s,2H),3.35(d,2H),2.96-3.09(m,2H),2.79-2.96(m,1H),2.29(s,6H),1.93-2.03(m,2H),1.77-1.90(m,2H)。

Intermediate 103-1: methanesulfonic acid (S) -oxetan-2-yl methyl ester

Step 1

Trimethyl sulphoxide iodide (1.32kg, 5.98mol) was added to a solution of potassium tert-butoxide (670g, 5.98mol) in t-BuOH (5L) at 25 ℃. The mixture was heated to 60 ℃ and stirred for 30min, followed by addition of (S) -2- ((benzyloxy) methyl) ethylene oxide (500g, 2.99 mol). The mixture was heated to 80 ℃ for 2 h. The mixture was cooled to 25 ℃ and passedAnd (5) filtering. The solid was washed with PE (3X 2L). The filtrate was treated with water (10L) and extracted with PE (2X 5L). The organic layer was washed with brine, dried, filtered and concentrated in vacuo. The crude product was purified by column chromatography (PE/EtOAc gradient from 15:1 to 10: 1) to give (S) -2- ((benzyloxy) methyl) oxetane (280g, 52.6%) as a clear oil.1H NMR(400MHz,CDCl3)δ7.15-7.34(m,5H),4.90(tdd,1H),4.44-4.67(m,4H),3.49-3.63(m,2H),2.44-2.66(m,2H)。

Step 2

The reaction was carried out in two parallel batches; example batches were as follows: pd (OH)2(14g) To a solution of (S) -2- ((benzyloxy) methyl) oxetane (140g, 780mmol) in THF (1.4L) under a nitrogen blanket was added. Heating the mixture to 45 ℃ and in H2Stirred (50psi) for 16 h. The mixture was cooled to 25 ℃ and passedFiltration to provide the desired compound (S) -oxetan-2-ylcarbinol as a solution in THF. A small portion is aliquoted to 1H NMR check and the remaining solution was used directly in the next step.1H NMR(400MHz,DMSO-d6)δ4.76-4.90(m,1H),4.66(tdd,1H),4.46(ddd,1H),4.37(td,1H),3.47(dd,2H),2.32-2.58(m,2H)。

Step 3

The reaction was carried out in two parallel batches; example batches were as follows: adding Et3N (197g, 1.95mol) was added to a solution of (S) -oxetan-2-ylcarbinol (from step 2, assumed value 69g, 780mmol) in THF (1.4L) at 0 ℃. Methanesulfonic anhydride (204g, 1.17mol) was added dropwise, maintaining the internal temperature below 10 ℃. The mixture was stirred at 25 ℃ for 2 h. The two batches were combined and the mixture was treated with water (1L) and the layers were separated. The aqueous phase was extracted with DCM (3X 2L). The combined organic solutions were dried, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (gradient of EtOAc/PE 50 to 100%) to give intermediate 20 as a yellow oil (250g, 96% over two steps).1H NMR(400MHz,CDCl3)δ4.98-5.09(m,1H),4.69(ddd,1H),4.59(td,1H),4.37(d,2H),3.11(s,3H),2.72-2.82(m,1H),2.64(tdd,1H)。

Intermediate 103-2: (S) -4-Nitro-3- ((oxetan-2-ylmethyl) amino) benzoic acid methyl ester

Step 1

Sodium azide (105g, 1.62mol) was added to a solution of (S) -oxetan-2-ylmethyl methanesulfonate (180g, 1.08mol) in DMF (1.2L). The mixture was heated to 80 ℃ and stirred for 16 h. The mixture was cooled to 0 ℃, treated with diethyl ether (1.5L) and the resulting suspension was stirred for 30 min. The solid was removed by filtration and the filter cake was washed with diethyl ether (2X 200 mL). The diethyl ether was removed under vacuum at 25 ℃ to give a solution of (S) -2- (azidomethyl) oxetane in DMF (. about.1.2L) which was used directly in the next step.

Step 2

The reaction was carried out in three parallel batches; example batches were as follows: (S) -2- (azidomethyl) oxetane (assumed value 41g, 36 g) in DMF (. about.400 mL) and THF (1L) was added 10% Pd/C (50 wt% wet, 13g) under a nitrogen blanket0mmol) of the solution. The mixture was heated at 25 ℃ in H2Stirred (50psi) for 16 h. Passing the solution throughFiltration, addition of 10% Pd/C (dry, 4.0g) and mixing at 40 ℃ in H2Stirring was carried out for 3h (50psi) and subsequent TLC analysis showed complete reaction. The mixture was cooled to 0 ℃ and all three batches were combined. Passing the mixture throughFiltration to obtain a solution of (S) -2- (aminomethyl) oxetane in DMF (. about.1.4L) and THF (. about.2.6L) which was used directly in the next step.

Step 3

Adding Et3N (327g, 3.24mol) and methyl 3-fluoro-4-nitrobenzoate (200g, 1.0mol) were added to a solution of (S) -2- (aminomethyl) oxetane (assumed value 94g, 1.08mol) in DMF (. about.1.4L) and THF (. about.2.6L) at 25 ℃. The mixture was stirred at 25 ℃ for 16 h. The mixture was concentrated under reduced pressure to remove THF and the remaining solution was diluted with water (1L). The mixture was extracted with EtOAc (2X 1.5L). The combined organic extracts were washed with brine (2 ×), dried and concentrated under reduced pressure. The crude product was purified by column chromatography (gradient of EtOAc/PE from 10 to 50%) to afford intermediate 103-2 as a yellow solid (158g, 55%). 1H NMR(600MHz,CDCl3)δ8.38(br s,1H),8.25(d,1H),7.64(s,1H),7.27(d,1H),5.13-5.20(m,1H),4.70-4.82(m,1H),4.64(td,1H),3.95(s,3H),3.57-3.71(m,2H),2.71-2.86(m,1H),2.55-2.70(m,1H);MS(ES+)=266.7。

Intermediate 103-3: (S) -4-amino-3- ((oxetan-2-ylmethyl) amino) benzoic acid methyl ester

Intermediate 103-2(15g, 56mmol) was dissolved inIn THF (100mL) in the reactor. Pd/C (10% w/w, 1.5g) was added to the reactor and the mixture was H at 50psi at RT2Shake for 4 h. Passing the mixture throughFiltration and concentration of the filtrate under reduced pressure gave intermediate 103-3(12.3g, 92%) as a tan solid.1H NMR(600MHz,CDCl3)δ7.49(dd,1H),7.39(d,1H),6.70(d,1H),5.05-5.18(m,1H),4.76(ddd,1H),4.62(dt,1H),3.87(s,3H),3.42-3.50(m,1H),3.34-3.40(m,1H),2.71-2.82(m,1H),2.60(ddt,1H)。

Preparation of 2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid (example 103)

Step 1

2-chloro-1, 1, 1-trimethoxyethane (20.1mL, 149mmol) was added to a stirred solution of intermediate 103-3(33.6g, 142mmol) in MeCN (285mL), followed by pTSA. H2O (1.35g, 7.1 mmol). After 2h at 50 ℃ MeCN (280mL), K were added2CO3(79g, 570mmol) and intermediate 103-0(93.2g, 142 mmol). After 2h, the solution was treated with water (800mL), allowed to cool to RT and stirred for 2 h. The resulting precipitate was collected by filtration, washed with 10% MeCN in water (150mL), water (2 × 200mL) and then dried under reduced pressure to give (S) -2- ((4- (6- ((4-cyano-2-fluorophenylmethyl) oxy) pyridin-2-yl) piperidin-1-yl) methyl) -1- (oxetan-2-ylmethyl) -1H-benzo [ d ] as a colorless solid ]Imidazole-6-carboxylic acid methyl ester (77g, 95%).1H NMR(600MHz,DMSO-d6)δ8.28(s,1H),7.87(d,1H),7.80(d,1H),7.55-7.73(m,4H),6.87(d,1H),6.70(d,1H),5.45(s,2H),5.04-5.19(m,1H),4.81(dd,1H),4.66(dd,1H),4.41-4.54(m,1H),4.36(dt,1H),3.94(d,1H),3.86(s,3H),3.76(d,1H),2.97(d,1H),2.82(d,1H),2.63-2.77(m,1H),2.49-2.63(m,1H),2.37-2.46(m,1H),2.18-2.29(m,1H),2.05-2.18(m,1H),1.47-1.82(m,4H)。

Step 2

1,5, 7-triazabicyclo [4.4.0 ] in water]Deca-5-Alkene solution (0.97M, 14.7mL) added to (S) -2- ((4- (6- ((4-cyano-2-fluorophenylmethyl) oxy) pyridin-2-yl) piperidin-1-yl) methyl) -1- (oxetan-2-ylmethyl) -1H-benzo [ d ] in MeCN (70mL)]Imidazole-6-carboxylic acid methyl ester (4g, 7mmol) in a stirred solution. After 20h, the solution was acidified to pH-6 with citric acid in water (2M, 7mL) and diluted with water (50 mL). The aqueous phase was extracted with EtOAc (2X 75mL) and the combined organic layers were dried over anhydrous Na2SO4Drying, filtration and removal of the solvent under reduced pressure gave an off-white solid. The crude material was purified using column chromatography eluting with MeOH/DCM (0:100 to 8:92) to give example 103(3.65g, 90%) as a solid.1H NMR(400MHz,DMSO-d6)δ12.75(br s,1H),8.27(s,1H),7.89(d,1H),7.80(d,1H),7.68-7.72(m,2H),7.60-7.67(m,2H),6.89(d,1H),6.72(d,1H),5.47(s,2H),5.11(d,1H),4.74-4.86(m,1H),4.62-4.72(m,1H),4.43-4.53(m,1H),4.35-4.42(m,1H),3.95(d,1H),3.77(d,1H),2.98(d,1H),2.84(d,1H),2.65-2.77(m,1H),2.53-2.64(m,1H),2.37-2.45(m,1H),2.10-2.28(m,2H),1.57-1.84(m,4H)。LC-MS(ES+):556.6(M+H)。

Tris (hydroxymethyl) aminomethane salt of example 103

2- [ (4- {6- [ (4-cyano-2-fluorophenylmethyl) oxy ] pyridin-2-yl } piperidin-1-yl) methyl ] -1- [ (2S) -oxetan-2-ylmethyl ] -1H-benzimidazole-6-carboxylic acid tris (hydroxymethyl) aminomethane salt

Aqueous tris (hydroxymethyl) aminomethane (2.0M, 6.1mL, 12.2mmol) was added dropwise to a stirred solution of example 103(6.5g, 11.7mmol) in 1-propanol (275mL) at 70 deg.C, during which time a homogeneous solution was maintained. After stirring for 5min, seed crystals were added and the mixture was allowed to cool to RT over 2 h. After stirring overnight at RT, a solid formed. The solid was collected by filtration, washed with 1-propanol (2 × 30mL) and dried first under a stream of nitrogen and then in a vacuum oven at 45 ℃ for 15h to give the tris (hydroxymethyl) aminomethane salt of example 3 (6.95g, 88%) as a crystalline solid. 1H NMR(600MHz,DMSO-d6)δ:8.20(s,1H),7.89(d,1H),7.79(d,1H),7.70(br s,2H),7.64(t,1H),7.56(d,1H),6.89(d,1H),6.72(d,1H),5.47(s,2H),5.11(qd,1H),4.77(dd,1H),4.64(dd,1H),4.44-4.53(m,1H),4.38(dt,1H),3.93(d,1H),3.76(d,1H),3.35(br s,9H),2.98(d,1H),2.85(d,1H),2.64-2.75(m,1H),2.54-2.64(m,1H),2.40-2.49(m,1H),2.08-2.26(m,2H),1.56-1.83(m,4H)。mp=194℃。

CHO GLP-1R clone H6-assay 1

GLP-1R-mediated agonist activity was determined in a cell-based functional Assay using the HTRF (homogeneous time resolved fluorescence) cAMP detection Kit (cAMP HI Range Assay Kit; CisBio cat #62AM6PEJ) that measures cAMP levels in cells. The method is a competitive immunoassay between native cAMP produced by the cell and exogenous cAMP labeled with dye d 2. Tracer binding was visualized with the anti-cAMP mAb labelled with cryptates. The specific signal (i.e., energy transfer) is inversely proportional to the concentration of cAMP in the standard or experimental sample.

The human GLP-1R coding sequence (NCBI reference sequence NP _002053.3, including the naturally occurring variant Gly 168Ser) was subcloned into pcDNA3(Invitrogen) and a cell line stably expressing the receptor (designated clone H6) was isolated. Use of125I-GLP-17-36Saturation binding analysis (Filter assay procedure) of (Perkin Elmer) showed that plasma membranes derived from this cell line express high GLP-1R Density (K)d:0.4nM,Bmax: 1900fmol/mg protein).

Cells were removed from cryopreservation, resuspended in 40mL Dulbecco's phosphate buffered saline (DPBS-Lonza Cat #17-512Q) and centrifuged at 800 Xg for 5 min at 22 ℃. The cell pellet was then resuspended in 10mL growth medium [ DMEM/F12 1: 1 mixture with HEPES, L-Gln, 500mL (DMEM/F12 Lonza Cat #12-719F), 10% heat-inactivated fetal bovine serum (Gibco Cat # 16140-. Cell viability and cell count per mL were determined by counting 1mL samples of cell suspension in growth medium on a Becton Dickinson ViCell. The remaining cell suspension was then conditioned with growth medium, 2000 viable cells per well were delivered using a Matrix Combi multistrop reagent dispenser, and the cells were dispensed into a tissue culture treated white 384-well assay plate (Corning 3570). The assay plates were then incubated at 37 ℃ in a humidified environment of 5% carbon dioxide for 48 hours.

Various concentrations of each compound to be tested (in DMSO) were diluted in assay buffer (HBSS with calcium/magnesium (Lonza/BioWhittaker cat # 10-527F)/0.1% BSA (Sigma Aldrich cat # A7409-1L)/20mM HEPES (Lonza/BioWhittaker cat #17-737E) containing 100. mu.M 3-isobutyl-1-methylxanthine (IBMX; Sigma cat # I5879) with a final DMSO concentration of 1%.

After 48 hours, the growth medium was removed from the assay disc wells and the cells were treated with 20 μ L of serially diluted compounds in assay buffer for 30 minutes at 37 ℃ in a humidified environment of 5% carbon dioxide. After a 30 minute incubation, 10 μ L of labeled d2cAMP and 10 μ L of anti-cAMP antibody (both diluted 1:20 in lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. The discs were then incubated at room temperature and after 60 minutes, changes in HTRF signals were read with an Envision2104 Multi-label (Multi-label) disc reader using excitation at 330nm and emission at 615 and 665 nm. The raw data were converted to nM cAMP by interpolation of the cAMP standard curve (as described in the manufacturer's assay protocol), and the full agonist GLP-1 was determined relative to the saturation concentration included for each disc 7-36(1. mu.M) percent Effect. EC (EC)50The determination was performed with agonist dose response curves analyzed using a curve fitting program of the 4-parameter logistic dose response equation.

CHO GLP-1R clone C6-assay 2

GLP-1R-mediated agonist activity was determined in a cell-based functional Assay using the HTRF (homogeneous time resolved fluorescence) cAMP detection Kit (cAMP HI Range Assay Kit; CisBio cat #62AM6PEJ) that measures cAMP levels in cells. The method is a competitive immunoassay between native cAMP produced by cells and exogenous cAMP labeled with the dye d 2. Tracer binding was visualized with the anti-cAMP mAb labelled with cryptates. The specific signal (i.e., energy transfer) is inversely proportional to the concentration of cAMP in the standard or experimental sample.

A human GLP-1R coding sequence (NCBI reference sequence NP-002053.3, including the naturally occurring variant Leu260Phe) was subcloned into pcDNA5-FRT-TO and Flp-In was usedTM T-RexTMThe system isolated a clonal CHO cell line stably expressing low receptor density as described by the manufacturer (ThermoFisher). Use of125Saturation binding analysis (Filter assay procedure) of I-GLP-1(Perkin Elmer) showed that plasma membranes derived from this cell line (designated clone C6) expressed low GLP-1R density (K) d:0.3nM,Bmax: 240fmol/mg protein) compared to clone H6 cell line.

Cells were removed from cryopreservation, resuspended in 40mL Dulbecco's phosphate buffered saline (DPBS-Lonza Cat #17-512Q) and centrifuged at 800 Xg for 5 min at 22 ℃. The DPBS was aspirated and the cell pellet resuspended in 10mL of complete growth medium (1: 1 mixture of DMEM/F12 with HEPES, L-Gln, 500mL (DMEM/F12 Lonza Cat #12-719F), 10% heat-inactivated fetal bovine serum (Gibco Cat #16140-071), 5mL100X Pen-Strep (Gibco Cat #15140-122), 5mL100X L-glutamic acid (Gibco Cat #25030-081), 700. mu.g/mL hygromycin (Invitrogen Cat #10687010) and 15. mu.g/mL blasticidin (Gibco Cat # R21001)). Cell viability and cell count per mL were determined by counting 1mL samples of cell suspension in growth medium on a Becton Dickinson ViCell. The remaining cell suspension was then conditioned with growth medium, 1600 viable cells per well were delivered using a Matrix Combi multistrop reagent dispenser, and the cells were dispensed into a tissue culture treated white 384-well assay plate (Corning 3570). The assay plates were then placed in a humid environment (95% O) at 37 deg.C 25% of CO2) Incubation was carried out for 48 hours.

Various concentrations of each compound to be tested (in DMSO) were diluted in assay buffer [ HBSS with calcium/magnesium (Lonza/BioWhittaker cat # 10-527F)/0.1% BSA (Sigma Aldrich cat # A7409-1L)/20mM HEPES (Lonza/BioWhittaker cat #17-737E) containing 100. mu.M 3-isobutyl-1-methylxanthine (IBMX; Sigma cat # I5879) ]. The final DMSO concentration in the compound/assay buffer mixture was 1%.

After 48 hours, the growth medium was self-assayedThe wells were removed and the cells were placed in a humid environment (95% O) at 37 deg.C25% of CO2) Was treated with 20 μ L of serially diluted compounds in assay buffer for 30 minutes. After a 30 minute incubation, 10 μ L of labeled d2cAMP and 10 μ L of anti-cAMP antibody (both diluted 1:20 in lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. The disks were then incubated at room temperature and after 60 minutes, changes in HTRF signals were read with an Envision 2104 multi-marker flat disk reader using excitation at 330nm and emission at 615 and 665 nm. The raw data were converted to nM cAMP by interpolation of the cAMP standard curve (as described in the manufacturer's assay protocol) and the percent effect of the full agonist GLP-1(1 μ M) was determined relative to the saturation concentration included for each disc. EC (EC) 50The determination was performed with agonist dose response curves analyzed using a curve fitting program of the 4-parameter logistic dose response equation.

In Table 3, the assay data are provided as two (2) significant figures, geometric means (EC) based on the number of repetitions listed50) And an arithmetic mean (Emax). A blank space means that this embodiment has no data or no Emax calculated.

TABLE 3 biological Activity of examples 1-102

Test with ammonium salt and trifluoroacetate salt

Test with ammonium salt and 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-amine (Tris) salt and free acid

Test with ammonium salt and free acid

Test with formate and free acid

Example ACCi: preparation of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, (ACCi compound):

in the preparation of the compounds of example ACCi, it should be noted that some of the preparation methods described herein may require protection of remote functional groups (e.g., primary amine, secondary amine, carboxyl groups in the precursors of formula I). The necessity of such protection varies depending on the nature of the remote functional group and the conditions of the preparation process. The necessity of such protection is readily determined by one skilled in the art. The use of such protection/deprotection methods is also within the skill of the art. For a general description of protecting Groups and their use, see t.w. greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991. Furthermore, the present invention is not limited to the particular synthetic methods provided in this application, which may vary.

Intermediate a 1: 1-isopropyl-4, 6-dihydrospiro [ indazole-5, 4' -piperidin ] -7(1H) -one, hydrochloride salt

Step 1.9-oxo-3-azaspiro [5.5] undec-7-ene-3-carboxylic acid tert-butyl ester

4-formylpiperidine-1-carboxylic acid tert-butyl ester (108Kg), cyclohexane (1080L) and pyrrolidine (64.8Kg) were charged to the anhydrous reactor at 25 to 30 ℃. The mixture was stirred for 5 to 10min and then heated to reflux for 12 to 16h while collecting the water using a dean-Stark trap. The reaction mixture was then cooled to 50 to 60 ℃, at which temperature vacuum was applied to distill excess pyrrolidine and cyclohexane. The reaction mixture was then cooled to 25 to 30 ℃ and charged with cyclohexane (648L), followed by methyl vinyl ketone (49.63 Kg). The mixture was stirred for 12 to 16h, then filtered and the filtrate was charged to a clean and dry reactor. The solution was cooled to 10 to 15 ℃ and then a solution of acetic acid (54.75Kg) in water (54L) was slowly added to maintain the temperature below 15 ℃. At the end of the addition, the mixture was warmed to 25 to 30 ℃ and stirred for 12 to 16 h. The layers were separated and the aqueous layer was extracted with ethyl acetate (324L). The combined organic layers were washed with a solution of sodium bicarbonate (32.34Kg) in water (324L) and then dried over sodium sulfate. The solid was washed with ethyl acetate (54L) and the combined filtrates were concentrated under reduced pressure below 40 ℃. N-heptane (216L) was charged to the reactor and distilled under reduced pressure below 40 ℃ until dry. The mixture was cooled to 25 to 30 ℃ and n-heptane (216L) was charged to the reactor. The mixture was stirred for 1 to 2h, then a solid was formed. The solid was then filtered, washed with n-heptane (54L) and dried at 40 to 50 ℃ for 10 to 12h to yield the desired material (90.1kg, 67% yield).

Step 2.10- ((dimethylamino) methylene) -9-oxo-3-azaspiro [5.5] undec-7-ene-3-carboxylic acid (E) -tert-butyl ester

Reacting 9-oxo-3-azaspiro [5.5]]Tert-butyl undec-7-ene-3-carboxylate (50Kg), N-dimethylformamide (500L) and N, N-dimethylformamide dimethyl acetal (135Kg) were charged into a clean and dry reactor at 25 to 30 ℃ under nitrogen. The reaction mixture was stirred for 5 to 10min, then heated to 120 to 130 ℃ for 20 h. The mixture is then cooled to 50 to 60 ℃ and the solvent is distilled off under high vacuum below 60 ℃. The mixture, xylene (200L), was charged below 45 ℃ and the solvent was distilled off below 60 ℃ under high vacuum. This was repeated with another batch of mixed xylene (200L). Toluene (200L) was then charged to the reactor and the solvent was distilled off under high vacuum below 60 ℃. Repeat with a second batch of toluene (200L)This operation. Methyl tert-butyl ether (100L) was then charged at below 30 ℃ and the solvent was distilled off under high vacuum below 40 ℃. The mixture was cooled to 15 to 20 ℃ and charged with methyl tertiary-butyl ether (100L) at below 20 ℃. The mixture was stirred for 20 to 30min and the solid was filtered, washed with methyl tert-butyl ether (50L) and dried under non-vacuum at 50 to 55 ℃ for 10h to provide the desired compound (52.1kg, 87% yield). 1H NMR(400MHz,CDCl3)δppm 7.48(s,1H),6.57(d,J=9.97Hz,1H),5.99(d,J=10.16Hz,1H),3.32-3.51(m,4H),3.06(s,6H),2.72(s,2H),1.57-1.66(m,2H),1.41-1.53(m,11H)。

Step 3.1-isopropyl-1, 4-dihydrospiro [ indazole-5, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester

Reacting 10- ((dimethylamino) methylene) -9-oxo-3-azaspiro [5.5]The (E) -tert-butyl undec-7-ene-3-carboxylate (80Kg), toluene (704L) and trimethylamine (16L) were charged into a clean and dry reactor at 25 to 30 ℃. The reaction mixture was warmed to 70 to 80 ℃ and added over 4 to 5h to a solution of isopropylhydrazine hydrochloride in methanol (1.25 eq, 141Kg total). The reaction mixture was then stirred at 70 to 80 ℃ for 8 to 10h and then cooled to 15 to 25 ℃. A solution of citric acid (48Kg) in water (480L) was then slowly added to maintain the internal temperature below 25 ℃. Ethyl acetate (208L) was added and the mixture was stirred for 10 min. The layers were separated and the organic layer was washed successively with a solution of citric acid (48Kg) in water (480L) followed by water (320L) only. The combined aqueous layers were extracted with ethyl acetate (320L). The combined organic layers were then dried over sodium sulfate (8Kg) and the solvent was evaporated to dryness under reduced pressure at below 40 ℃. Dichloromethane (240L) was charged to the reactor and the mixture was stirred at 25 to 30 ℃ until clear. Activated carbon (1.84Kg), magnesium silicate (1.84Kg) and silica gel (32Kg, 100-mesh) were charged continuously at 25 to 30 ℃ and the heterogeneous mixture was stirred for 1 h. The slurry was then filtered over a Hyflow bed prepared by mixing Hyflow super monomer (8Kg) with dichloromethane (40L). The filter cake is treated with dichloromethane Alkane (three 120L) washes. The combined filtrates were charged back to the reactor and the solvent was evaporated under reduced pressure below 40 ℃. Then n-heptane (160L) was charged and evaporated under reduced pressure below 40 ℃. N-heptane (200L) was charged into the reactor and the mixture was cooled to 0 to 5 ℃. After stirring for 12 to 15h, the solid was filtered at 0 ℃, washed with quenched (0 to 5 ℃) n-heptane (160L) and dried under vacuum at 40 to 50 ℃ to provide the title compound (82.4kg, 75%).1H NMR(400MHz,CDCl3)δppm 7.25(s,1H),6.42(dd,J=10.05,0.49Hz,1H)5.84(d,J=9.95Hz,1H),4.42-4.52(m,1H),3.36-3.53(m,4H),2.62(s,2H)1.56-1.68(m,2H)1.45-1.55(m,17H)。

Step 4.1-isopropyl-4, 6-dihydrospiro [ indazole-5, 4' -piperidin ] -7(1H) -one, hydrochloride

1-isopropyl-1, 4-dihydrospiro [ indazole-5, 4 '-piperidine ] -1' -carboxylic acid tert-butyl ester (60Kg) and methanol (600L) were charged at 25 to 30 ℃ into a clean and dry reactor. N-bromosuccinimide (32.4Kg) was added in 5 portions over 30 to 40min at 25 to 30 ℃ and stirring was continued for 30 to 60 min. A solution of sodium thiosulfate pentahydrate (5.4Kg) in water (102L) was slowly added to maintain the internal temperature below 30 ℃. The mixture was stirred for 20 to 30min, followed by evaporation of the solvent under reduced pressure below 45 ℃. The residue was cooled to 25 to 30 ℃ and 2-methyltetrahydrofuran (420L) was charged to the reactor along with water (90L). The mixture was stirred for 15 to 20min, then the layers were separated and the aqueous layer was extracted with 2-methyltetrahydrofuran (120L). The combined organic extracts were treated with a solution of sodium hydroxide (4.8Kg) in water (120L) at 25 to 30 ℃ for 15 to 20 min. The layers were separated and the organic layer was washed with water (120L), then with a solution of sodium chloride (12Kg) in water (120L), then dried over sodium sulfate (6 Kg). After filtration, the filter cake was washed with 2-methyltetrahydrofuran (30L) and the combined filtrates were charged back to the reactor. The solvent was completely evaporated under reduced pressure below 45 ℃ and the residue was dissolved in tetrahydrofuran (201L). Potassium tert-butoxide (60.6Kg) and tetrahydrofuran (360L) were charged into another clean and dry reactor at 25 to 30 ℃. A solution of the residue in tetrahydrofuran was slowly added to the mixture, maintaining the temperature below 30 ℃. The reaction mixture is then warmed to 60 to 65 ℃ and held at this temperature for 1 to 2 h. On completion, the mixture was cooled to 0 to 10 ℃ and quenched slowly with hydrochloric acid solution (1N, 196L) maintaining the internal temperature below 10 ℃. The reaction mixture was allowed to warm to 25 to 30 ℃ and charged with ethyl acetate (798L). After stirring for 15 to 20min, the layers were separated and the aqueous layer was extracted again with ethyl acetate (160L). The combined organic layers were washed with water (160L), dried over sodium sulfate (8Kg), filtered and the filter cake was washed with ethyl acetate (300L). The solvent was completely evaporated under reduced pressure below 45 ℃ and ethyl acetate (540L) was charged to the reactor at 25 to 30 ℃ followed by methanol (156L). The mixture was cooled to 0 to 5 ℃, at which time acetyl chloride (79.8Kg) was added slowly, maintaining the temperature within the specified range. The mixture is then allowed to warm to 20 to 25 ℃ and kept at this temperature with stirring for 4 to 5 h. The resulting slurry was filtered and the solid was washed with ethyl acetate (120L) then dried at 40 to 45 ℃ for 8 to 10h to provide the desired crude product (33.5kg, 65%).

The final purification step was carried out by dissolving this crude solid (56.8Kg) in methanol (454.4L) in a clean dry reactor at 25 to 30 ℃. The solution was stirred for 30 to 45min, then passed through a 0.2 micron cartridge into a clean and dry reactor at 25 to 30 ℃. Methanol was distilled off under reduced pressure below 50 ℃ until-1 vol solvent remained. The reaction mixture was cooled to 25 to 30 ℃ and charged with fresh acetonitrile (113.6L) through a 0.2 micron cartridge. The solvent was distilled off under reduced pressure below 50 ℃ until-1 vol solvent remained. The reaction mixture was cooled to 25 to 30 ℃ and charged to the reactor through a 0.2 micron cartridge with fresh acetonitrile (190L). The mixture was warmed to 65 to 70 ℃ and stirred for 45min, then cooled to 25 to 30 ℃ and stirred for 1 h. The resulting slurry was filtered and the filter cake was washed with quenched (15 ℃) acetonitrile (56.8L). The solid was dried under reduced pressure at 40 to 50 ℃ for 8h to give intermediate a1(36.4kg, 64%).1H NMR(400MHz,CD3OD)δppm 7.43(s,1H),5.32-5.42(m,1H),3.15-3.25(m,4H),2.89(s,2H),2.64(s,2H),1.69-1.90(m,4H),1.37-1.45(m,6H);ESI[M+H]+=248。

Intermediate a 2: 2- (4- (tert-butoxycarbonyl) phenyl) -6-methoxyisonicotinic acid

2, 6-dichloroisonicotinic acid (30Kg) and methanol (120L) were charged into a clean and dry reactor at 20 to 25 ℃. The slurry was stirred for 5min, then heated to 65 ℃ (reflux). Then slowly charged via addition funnel with a solution of sodium methoxide in methanol (30%, 87.2Kg) over at least 4 h. The funnel was rinsed with methanol (15L) and stirred at 65 ℃ for at least 15 h. The mixture was then cooled to 45 ℃ and distilled under reduced pressure until a residual volume of-90L. A solution of sodium bicarbonate (28.2Kg) and potassium carbonate (21.6Kg) in water (180L) was then charged to the reactor at 40 to 45 ℃. The reactor containing the aqueous solution was rinsed with water (21L) and the rinse was charged to the reaction mixture. The mixture was distilled under reduced pressure below 80 ℃ until a residual volume of 240L was reached, followed by cooling to 20 to 25 ℃.

Tert-butyl 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) benzoate (52.3Kg) and dioxane (340Kg) were charged to another clean and dry reactor and stirred at 2 to 25 ℃ until completely dissolved. The contents of the pre-reactor were then heated at 40 ℃ to ensure complete dissolution and transfer to this new reactor. The reaction mixture was cooled to 20 to 25 ℃ and the deoxygenation step was performed via vacuum/nitrogen recycle. The mixture was cooled again to 0 to 10 ℃ and palladium acetate (0.65Kg) was charged to the reactor, followed by triphenylphosphine (2.46Kg) under a stream of nitrogen. The mixture was warmed to 20 to 25 ℃ and another deoxygenation step was performed via vacuum/nitrogen recycle. The mixture was then heated to 80 ℃ and maintained at this temperature for at least 18 h. The mixture was cooled to 20 to 25 ℃ and methyl t-butyl ether (133.2Kg) and water (30L) were continuously charged into the reactor. The layers were separated and the aqueous layer was diluted with water (110L) followed by extraction with methyl tert-butyl ether (110L). To be combined withThe organic extract was washed with a solution of citric acid (52Kg) in water (84L) and the layers were separated. The aqueous layer was extracted with additional methyl tert-butyl ether (88.8Kg) and the organic layers were combined and washed three times with one third of a solution of sodium chloride (43Kg) in water (80L). After the final layer separation, the organic layer was filtered through a pall filter containing a charcoal cartridge and the filter cake was washed with methyl tert-butyl ether (11.2 Kg). The filtrate was distilled under reduced pressure below 50 ℃ to-90L and then continuously co-distilled with heptane (120L) below 50 ℃ to-120L. The mixture was then cooled to 20 to 25 ℃ over 1h, followed by stirring at this temperature for a further 1 h. The slurry was filtered and the filter cake was washed three times with heptane (3 × 18L) followed by three times with acetonitrile (3 × 18L). The resulting wet solid was dried under vacuum under a stream of nitrogen at less than 45 ℃ for at least 15h to give intermediate a2(44.6kg, 87% yield). 1H NMR(400MHz,CDCl3)δppm 8.13(s,2H),8.09(s,2H),7.97(d,J=1.17Hz,1H),7.34(d,J=0.98Hz,1H),4.08(s,3H),1.61(s,9H);ESI[M+H]+=330。

Intermediate a 3: 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid tert-butyl ester

2- (4- (tert-Butoxycarbonyl) phenyl) -6-methoxyisonicotinic acid (intermediate A2, 15.2g, 46.2mmol) and ethyl acetate (140mL) were charged into a round-bottomed flask. 1, 1' -carbonyldiimidazole (8.98g, 55.4mmol) was added in one portion and stirred at rt for 1 h. 1-isopropyl-4, 6-dihydrospiro [ indazole-5, 4' -piperidin ] -7(1H) -one hydrochloride (intermediate a1, 14.8g, 52.2mmol) was added followed by N, N-diisopropylethylamine (9.1mL, 52.2mL) and the reaction stirred at rt for 18H. Aqueous 2M HCl (40mL) was added followed by 1M potassium bisulfate (40mL) and 50mL heptane. The resulting mixture was stirred at rt for 1 h. The mixture was transferred to a separatory funnel. The organic phase was separated, washed successively with water (20mL), saturated sodium bicarbonate (30mL), water (20mL), brine (20mL), dried over 20g magnesium sulfate and 10g silica gel, filtered and concentrated in vacuo. Solids began to form towards the end of the concentration. The residue was stirred in 40mL ethyl acetate at 80 ℃ and heptane (120mL) was slowly added dropwise. The mixture was stirred at 80 ℃ for 1h, then slowly cooled to room temperature over 1h with stirring and stirred at rt for 18 h. The solid was collected via filtration, washed with water and ethyl acetate-heptane (1:3) and dried under vacuum at 50 ℃ for 18h to afford intermediate a3(19.64g, 76% yield).

Alternative preparation of intermediate a 3:

acetonitrile (219Kg) and 2- (4- (tert-butoxycarbonyl) phenyl) -6-methoxyisonicotinic acid (intermediate a2, 34.8Kg) were charged to a clean and dry reactor at 20 to 25 ℃. The mixture was stirred for 5min and then charged 1, 1-carbonyldiimidazole (18.9Kg) in three portions. The slurry was stirred at 20 to 25 ℃ for at least 1H, then 1-isopropyl-4, 6-dihydrospiro [ indazole-5, 4' -piperidin ] -7(1H) -one hydrochloride (intermediate a1, 33.0Kg) was charged to the reactor, followed by N, N-diisopropylethylamine (20.5Kg) via a pump. The reagent pump and the reactor walls were rinsed with acetonitrile (13.7Kg) and stirred at 20 to 25 ℃ for at least 2 h. Upon completion, the mixture was seeded with tert-butyl 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoate (intermediate a3,209g) and stirred for at least 30 min. After confirming the start of crystallization, a solution of citric acid monohydrate (58.5Kg) in water (257L) was charged over 1 h. The resulting slurry was stirred at 20 to 25 ℃ for a further 2h, then filtered and the filter cake washed with a mixture of acetonitrile (68.4Kg) and water (87L). The reactor is also flushed with this wash. The solid was dried under reduced pressure below 55 ℃ to give intermediate a3(43.44kg, 73% yield).

Compounds of example ACCi (as free acid): 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid

Tert-butyl 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoate (3.7g, 6.6mmol) and toluene (25mL) were charged to a round bottom flask. 85% phosphoric acid (3.0mL) was added dropwise with stirring and the reaction was heated to 60 ℃ for 4 hours. A colorless thick gum formed. The reaction was cooled to rt and water was added. A white solid was observed. The toluene organic layer was discarded, and the aqueous layer and solid were retained. Ethyl acetate (60mL) was added and 4N NaOH solution was added to adjust the pH to-7. The layers were separated and the aqueous layer was extracted with ethyl acetate (50 mL). The combined ethyl acetate organic layers were dried over sodium sulfate, filtered and concentrated in vacuo to afford a white solid. The solid was dissolved in ethyl acetate (80mL) at 50 ℃ and heptane (90mL) was slowly added. The heat was removed, the mixture was cooled to rt and stirred for 16 h. The resulting solid was collected by filtration, washed with mother liquor and dried to give the title compound (4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid as a white solid, free form, 2.15g, 65% yield).

Alternative preparation of the compound of example ACCi (as the free acid):

acetonitrile (130.4Kg) and tert-butyl 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoate (intermediate a3, 20.72Kg) were charged to a clean and dry reactor at 20 to 25 ℃. The mixture was stirred for 5min, then charged with p-toluenesulfonic acid (8.5Kg) under a gentle nitrogen purge. The reaction mixture was warmed to 70 ℃ and maintained at this temperature for at least 6.5 h. On completion, the mixture was cooled to 40 ℃, seeded with the compound of example ACCi (104g) and slowly charged with water (83L) over at least 1 h. The mixture was stirred at 40 ℃ for a minimum of 4h and then cooled to 20 to 25 ℃ over 2 h. Stirring was continued for at least 2h, followed by filtration and the filter cake was rinsed with a solution of acetonitrile (33Kg) and water (41L). The reactor is also flushed with this wash. The resulting solid was dried under reduced pressure below 55 ℃ to give 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (16.5kg, 89% yield).

Type 1-

Preparation of anhydrous mono-tris (hydroxymethyl) aminomethane for example ACCi:

A vial was charged with 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (151mg, 0.300mmol) and 3mL of ethanol. The mixture was heated to 80 ℃ for 5 minutes to dissolve the solid, then cooled to rt. Tris (hydroxymethyl) aminomethane (39mg, 0.32mmol) was added and the mixture was stirred at rt overnight. Heptane (2.25mL) was added dropwise to create a slurry, which was heated to 50 ℃ to create a clear solution. The mixture was cooled to rt overnight with stirring. A white solid was observed and the mixture was stirred for an additional 3 days. The material was filtered and dried in a vacuum oven at 50 ℃ overnight to give form 1 (151mg, 0.242mmol, 81% yield).

Example 1 type AcCi-

Example alternate preparation of anhydrous mono-tris (hydroxymethyl) aminomethane of ACCi:

ethanol (83L) was charged to a clean and dry reactor followed by the compound of example ACCi (9.43Kg) and tris (hydroxymethyl) aminomethane (2.55Kg) while the mixture was maintained at a temperature of 20 to 25 ℃. The walls of the tank were rinsed with ethanol (2L) and the resulting mixture was heated at 65 to 70 ℃ for at least 30 minutes at this temperature until all solids were dissolved, then cooled to 45 to 50 ℃. Warm filtration was performed through a 10 μm in-line polypropylene filter and the reactor and filter were rinsed with ethanol (9L). N-heptane (24L) was charged through the same in-line filter into the warm solution and the mixture was seeded with 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid anhydrous tris (hydroxymethyl) aminomethane salt (100g) in ethanol (0.5L) at 45 to 50 ℃. The temperature is maintained for at least 2h and then cooled to 20 to 25 ℃ over at least 2 h. Stirring was carried out for at least 5 days. The slurry was then filtered and the filter cake washed with a mixture of ethanol (13L) and n-heptane (6L). The solid was dried under reduced pressure below 45 ℃ for at least 12h to give example ACCi (11.7kg, 77%).

Example 2 type AcCi-

Example preparation of trihydrate of the mono tris (hydroxymethyl) aminomethane salt of ACCi:

form 2 of the compound of example ACCi was obtained from conversion of form 1 of the compound of example ACCi. Form 1 (1.7214g, 2.760mmol), isopropanol (16.50mL, 215.8mmol) and water (688 μ L, 38.190mmol) were added to a 50mL EasyMax reactor. The mixture was stirred (300rpm) at a reactor jacket temperature of 25 ℃ for about 72 hr. The reaction mixture was then warmed to 40 ℃ over 15min and held at 40 ℃ for about 24 hours, once cooled to 20 ℃ to remove the sample for testing. Mixture form observed as PXRD; additional water (688. mu.L, 38.190mmol) was therefore added. The stirring rate was increased to 400rpm, and the slurry was allowed to stir for 6 hours and then cooled to 15 ℃. The solid was separated on a 60mL/40M filter and washed with 96/4 isopropanol/water. The resulting material was identified by PXRD as being consistent with form 2 of the compound of example ACCi.

Type 2 of the Compounds of example ACCi-

Example alternate preparation of trihydrate of the mono tris (hydroxymethyl) aminomethane salt of ACCi:

isopropanol (60.4Kg) was charged to a clean and dry reactor and the compound of example ACCi (16.68Kg) and tris (hydroxymethyl) aminomethane (4.42Kg) were added while the mixture was maintained at a temperature of 20 to 25 ℃. The mixture was stirred for 5min, then charged with water (6.7Kg) and the slurry warmed to 55 ℃. The now clear solution was filtered through a 10 μm in-line polypropylene filter into a pre-warmed, clean and dry reactor (50 to 55 ℃). The solution was then seeded with the mono tris (hydroxymethyl) aminomethane salt of the compound of example ACCi (as trihydrate) (167 g). After verifying that the seeds were present continuously, the mixture was cooled to 15 ℃ over at least 2h, followed by a minimum of 16h at 15 ℃. The slurry was filtered and the filter cake was washed with quenched isopropanol (13.1 Kg). The solid was then dried under reduced pressure below 25 ℃ to afford form 2 of example ACCi only (22.1kg, 98% yield).

Form 1 of example ACCi is anhydrous and thermodynamically stable at water activity (20% RH) below about 0.2 at ambient temperature. Type 1 of example ACCi has a PXRD pattern substantially the same as the pattern shown in fig. 3 of example ACCi. The characteristic PXRD peaks for form 1 of example ACCi, expressed in 2 theta +0.2 deg. 2 theta, are at 9.6, 10.7 and 11.3. The peak positions and intensities of the PXRD patterns in fig. 3 are provided in table 1.

TABLE 1 PXRD Peak and relative intensity for form 1 of example ACCI

Form 1 of example ACCi has a raman spectrum substantially the same as the spectrum shown in fig. 4. Type 1 of example ACCI has a +2cm width at 568, 698, 989, 1218, 1511, 1561 and 1615-1In cm-1Characteristic raman peak shifts are indicated. The peak position (+2 cm) of type 1 of the embodiment ACCi in FIG. 4-1) And normalized intensities (W ═ weak, M ═ medium, S ═ strong) are listed in table 2.

TABLE 2 Raman peaks and normalized intensities of form 1 of example ACCI

Form 1 of example ACCI has a spectrum substantially the same as that shown in FIG. 513C ssNMR spectrum. Form 1 of example ACCI has characteristics in ppm expressed at 22.9, 146.2, 157.9, 161.9 and 172.9+0.2ppm13C ssNMR chemical shift. Type 1 of embodiment ACCi shown in FIG. 5 13The C chemical shifts (+0.2ppm) are shown in Table 3.

TABLE 3 form 1 of example ACCi13Chemical shift and intensity of C

Form 2 of example ACCi is a trihydrate and is thermodynamically stable at water activity above about 0.2 at ambient temperature and 20% RH. Type 2 of example ACCi has a PXRD pattern substantially the same as the pattern shown in fig. 6. The characteristic PXRD peaks for form 2 of example ACCi, expressed in 2 theta +0.2 deg. 2 theta, are at 8.4, 9.0, 10.5, 15.0 and 24.7. The peak positions and intensities of the PXRD patterns in fig. 6 are provided in table 4.

TABLE 4 PXRD peaks and relative intensities for form 2 of example ACCI

Form 2 of example ACCi has a raman spectrum substantially the same as the spectrum shown in fig. 7. Type 2 of the embodiment ACCi has values in 562, 692, 984, 1225, 1507, 1557 and 1610+2cm-1In cm-1Characteristic raman peak shifts are indicated. The peak position (+2 cm) of type 2 of example ACCi in FIG. 7-1) And normalized intensities (W ═ weak, M ═ medium, S ═ strong) are listed in table 5.

TABLE 5 Raman Peak and normalized intensity for form 2 of example ACCi

Form 2 of example ACCi has a spectrum substantially the same as that shown in FIG. 813C ssNMR spectrum. Form 2 of example ACCI has characteristics in ppm of 19.2, 149.5, 155.6, 163.8 and 188.3+0.2ppm 13C ssNMR chemical shift. Type 2 of embodiment ACCi shown in FIG. 813Chemical shift of C(+0.2ppm) is listed in Table 6.

TABLE 6 forms 2 of example ACCi13Chemical shift and intensity of C

Based on the disclosure provided herein, one of ordinary skill in the art will appreciate that each form 1 and form 2 of example ACCi can be uniquely identified with several different spectral peaks or patterns in different combinations. Illustrated below are exemplary combinations of characteristic peaks that can be used to identify form 1 and form 2, respectively, of compound a of the example ACCi, but in no way should be taken as limiting other peak combinations disclosed in this application.

To confirm the presence of three water molecules in form 2 of example ACCI, data were collected at room temperature using a Bruker D8 Venture diffractometer. See fig. 9. The structure is represented by monoclinic family space group P2 by using SHELX software package1Intrinsic phasing resolution of/c (5.1 edition, Bruker AXS, 1997). The structure is then refined with a full matrix least squares method. All non-hydrogen atoms were found and refined using anisotropic shift parameters.

The hydrogen atoms located on nitrogen and oxygen were found from the fourier difference plot and refined with the constrained distance. The remaining hydrogen atoms are positioned at the calculated positions and allowed to sit on their carrier atoms.

The final R index was 7.2%. The resulting difference fourier reveals the electron density without loss or dislocation.

Table 7 provides data collected on type 2 of example ACCi:

TABLE 7

Crystalline forms of 2-amino-2- (hydroxymethyl) propan-1, 3-dioi salt of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid. This crystalline salt is commonly referred to as tris (hydroxymethyl) aminomethane salt.

Example a crystalline form of tris (hydroxymethyl) aminomethane salt of ACCi wherein the ratio of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid to salt is 1: 1.

Crystalline tris (hydroxymethyl) aminomethane salt form ACCi of example wherein the crystalline salt is an anhydrous crystalline salt.

The anhydrous crystalline tris (hydroxymethyl) aminomethane salt of example ACCi wherein the anhydrous crystalline salt has a PXRD pattern comprising peaks at diffraction angles of 9.6, 10.7, and 11.32 theta, +0.2 ° 2 theta.

The anhydrous crystalline form of tris (hydroxymethyl) aminomethane salt of embodiment ACCi wherein the anhydrous crystalline salt has a crystalline structure comprised between 1511, 1561 and 1615cm-1+2cm-1Raman spectrum of the peak shift of (a).

The anhydrous crystalline form of tris (hydroxymethyl) aminomethane salt of embodiment ACCi wherein the anhydrous crystalline salt has chemical shifts comprised at 22.9, 146.2, and 161.9ppm +0.2ppm 13C ssNMR spectrum.

The anhydrous crystalline form of tris (hydroxymethyl) aminomethane salt of embodiment ACCi, wherein the anhydrous crystalline salt has analytical parameters selected from the group consisting of: included at 1511 and 1615cm-1+2cm-1And at least one chemical shift comprised at 22.9, 146.2 or 161.9ppm +0.2ppm13C ssNMR spectrum.

The anhydrous crystalline form of tris (hydroxymethyl) aminomethane salt of an embodiment, wherein the anhydrous crystalline salt is substantially pure.

Crystalline tris (hydroxymethyl) aminomethane salt form ACCi wherein the crystalline salt is a trihydrate crystalline salt.

The trihydrate crystalline tris (hydroxymethyl) aminomethane salt of example ACCi wherein said trihydrate crystalline salt has a PXRD pattern comprising peaks at diffraction angles of 8.4, 9.0, and 10.52 theta +0.2 DEG 2 theta.

The crystalline trihydrate tris (hydroxymethyl) aminomethane salt of example ACCi wherein said crystalline trihydrate salt has a composition comprised between 1507, 1557 and 1610cm-1+2cm-1Raman spectrum of the peak shift of (a).

The trihydrate crystalline tris (hydroxymethyl) aminomethane salt of example ACCi wherein said trihydrate crystalline salt has chemical shifts comprised at 19.2, 149.5 and 163.8ppm +0.2ppm13C ssNMR spectrum.

The trihydrate crystalline tris (hydroxymethyl) aminomethane salt of example ACCi, wherein said trihydrate crystalline salt has analytical parameters selected from the group consisting of:

a PXRD pattern containing peaks at diffraction angles of 8.4 and 9.02 theta +0.2 DEG 2 theta,

contained in 1557 and 1610cm-1+2cm-1Raman spectrum of the peak shift of (A), and

comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm +0.2ppm13C ssNMR spectrum.

The trihydrate crystalline tris (hydroxymethyl) aminomethane salt of example ACCi, wherein said trihydrate crystalline salt has analytical parameters selected from the group consisting of: PXRD pattern comprising peaks at diffraction angles of 8.4 and 9.02 theta +0.2 DEG 2 theta, and PXRD pattern comprising peaks at 1507, 1557 or 1610cm-1+2cm-1At least one peak shift of the raman spectrum.

The trihydrate crystalline tris (hydroxymethyl) aminomethane salt of example ACCi, wherein said trihydrate crystalline salt has analytical parameters selected from the group consisting of: PXRD pattern comprising peaks at diffraction angles of 8.4 and 9.02 theta +0.2 DEG 2 theta and a PXRD pattern comprising at least one chemical shift at 19.2, 149.5 or 163.8ppm +0.2ppm13C ssNMR spectrum.

Example DGAT2i (DGAT2i compound): (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide

Step 1: 3-ethoxypyridine

Cesium carbonate (12mol, 1.5 equivalents) and iodoethane (9.7mol, 1.2 equivalents) were added to a solution of 3-hydroxypyridine (8.10mol, 1.0 equivalents) in acetone (12L) at 15 ℃. The reaction mixture was stirred at room temperature for 24 hours. The reaction mixture was filtered and the organic layer was concentrated to give the crude product. Ethyl acetate (20L) was added and washed with water (3X 5L). The organic layer was dried over sodium sulfate, filtered, and concentrated to give 3-ethoxypyridine (620g, 62%) as an oil.1H NMR(400MHz,CDCl3)δ1.44(t,3H),4.07(q,2H),7.15-7.23(m,2H),8.20(dd,1H),8.30(d,1H)。

Step 2: 3-ethoxypyridine-1-oxide

M-chloroperoxybenzoic acid (6.5mol, 1.3 equivalents) was added to a solution of 3-ethoxypyridine (5.0mol, 1.0 equivalents) in dichloromethane (12L) at 10 ℃. The reaction mixture was stirred at room temperature for 24 hours. Sodium thiosulfate (4kg in 5L water) was added. The reaction mixture was stirred at 15 ℃ for 2 hours. Another portion of sodium thiosulfate (1.5kg in 5L of water) was added. The reaction mixture was stirred at 15 ℃ for 1 hour. The mixture was extracted with dichloromethane (16X 10L). The combined organic layers were concentrated to yield the crude product. The crude product was purified by silica gel column chromatography (dichloromethane: methanol; 100:1 to 10:1) to give the title compound (680g, 97%) as a brown oil. This was further purified by wet milling with petroleum ether (4L) for 24 hours at room temperature to give 3-ethoxypyridine-1-oxide (580g, 83%) as a yellow solid. 1H NMR(400MHz,CDCl3)δ1.41(t,3H),4.02(q,2H),6.84(dd,1H),7.12(dd,1H),7.85(d,1H),7.91-7.95(m,1H)。

And step 3: 2- ((5-bromopyridin-3-yl) oxy) -3-ethoxypyridine

The reaction was carried out in five parallel batches.

Diisopropylethylamine (2.69mol, 3.7 equivalents) and trispyrrolidinylphosphonium bromide hexafluorophosphate (0.93mol, 1.3 equivalents) were added toIn a stirred solution of 3-ethoxypyridine-1-oxide (0.72mol, 1.0 eq.) and 3-bromo-5-hydroxypyridine (0.72mol, 1.0 eq.) in tetrahydrofuran (2500mL) at room temperature. The reaction mixture was stirred at room temperature for 2 days, then the separate batches were combined into a single batch. The resulting suspension was concentrated to dryness and dissolved in dichloromethane (25L). The organic layer was washed with 1N sodium hydroxide (15L), water (3X 20L) and brine (20L). The organic layer was dried over sodium sulfate, filtered and concentrated to give an oil. The crude oil was purified by silica gel column chromatography (petroleum ether: ethyl acetate; 10:1 to 1:1) to give the crude product as a brown solid. The solid was purified as methyl tert-butyl ether: petroleum ether (1: 10; 11L) wet milling gave 2- ((5-bromopyridin-3-yl) oxy) -3-ethoxypyridine (730g, 69%) as a pale yellow solid.1H NMR(400MHz,CDCl3)δ1.49(t,3H),4.16(q,2H),7.04(dd,1H),7.25(dd,1H),7.68-7.73(m,2H),8.44(d,1H),8.49(d,1H)。MS(ES+)297.1(M+H)。

And 4, step 4: 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid ethyl ester

A solution of 2- ((5-bromopyridin-3-yl) oxy) -3-ethoxypyridine (300mmol, 1.0 eq) in tetrahydrofuran (1.3L) was degassed with nitrogen for 30 minutes. A strong effect Grignard (Turbo Grignard) (390mmol, 1.3 eq, 1.3M in tetrahydrofuran) was added at room temperature at a rate to maintain an internal temperature below 30 ℃. The reaction mixture was allowed to cool to room temperature and stirred for 3 hours. The reaction was cooled to 10 ℃ and zinc chloride (390mmol, 1.3 equivalents, 1.9M in 2-methyltetrahydrofuran) was added at a rate to maintain the temperature below 15 ℃. The resulting suspension was warmed to room temperature until all the precipitate dissolved and then cooled to 10 ℃. Solid ethyl 2-chloropyrimidine-5-carboxylate (360mmol, 1.2 equiv.) and dichloro [ bis (2- (diphenylphosphino) phenyl) ether were added]Palladium (II) (6.00mmol, 0.02 eq). The resulting suspension was degassed with nitrogen for 30 minutes and then heated to 50 ℃ for 16 hours. The reaction was worked up under aqueous conditions, followed by treatment sequentially with ethylenediaminetetraacetic acid disodium salt, thiosilica (thiosilica) and charcoal to remove metal impurities. The crude compound was recrystallized from methanol (450mL) to give 2- (5- ((3-ethoxypyridine-2-) -as a pale yellow solidYl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid ethyl ester (77g, 70%). 1H NMR(400MHz,CDCl3)δ1.44(t,3H),1.50(t,3H),4.19(q,2H),4.46(q,2H),7.00-7.04(m,1H),7.25(s,1H),7.71(d,1H),8.59(s,1H),8.66(d,1H),9.32(s,2H),9.55(s,1H)。

And 5: 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid (intermediate 1)

Sodium hydroxide (307mmol, 1.5 equiv., 4M aq.) and methanol (50mL) were added to a suspension of 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylate (205mmol, 1.0 equiv.) in tetrahydrofuran (300 mL). The resulting solution was stirred at room temperature for 3 hours. The reaction mixture was diluted with water (400mL) and extracted with 2:1 diethyl ether: heptane (2X 300 mL). The aqueous layer was acidified to pH 4 with 4M hydrochloric acid. The resulting suspension was stirred at room temperature for 1 hour. The solid was filtered, washed with water and dried to give 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid as a pale yellow solid (69g, 100%).1H NMR(400MHz,DMSO-d6)δ1.37(t,3H),4.18(q,2H),7.19(dd,1H),7.58(dd,1H),7.70(dd,1H),8.35-8.40(m,1H),8.66(d,1H),9.33(s,2H),9.41(d,1H),13.9(br.s,1H)。

Step 6: (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide [ example DGAT2i (DGAT2i Compound) ]

Oxalyl chloride (13.8mL, 160mmol, 1.2 equiv.) and dimethylformamide (0.510mL, 6.65mmol, 0.05 equiv.) are added to a suspension of 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid (45.0g, 133mmol, 1.0 equiv.) in dichloromethane (500 mL). When a solution was obtained, the suspension was stirred for 2 hours. The reaction mixture was concentrated to give crude acid chloride as a red solid. A solution of (S) -tetrahydrofuran-3-amine (12.2g, 140mmol, 1.05 equiv.) and diisopropylethylamine (51.0mL, 293mmol, 2.2 equiv.) in tetrahydrofuran (100mL) was added dropwise to a solution of the crude acid chloride in dichloromethane (200mL) at 0 ℃. The reaction was allowed to warm to room temperature and stirred for 16 hours. Water (1.0L) and ethyl acetate (600mL) were added and the organic layer was separated to saturate Sodium bicarbonate was washed, dried over magnesium sulfate and filtered. The filtrate was treated with activated charcoal (20g) and stirred at 65 ℃ for 20 minutes. The suspension was filtered warm and the filtrate was concentrated to a light yellow solid which was recrystallized from methanol in ethyl acetate (1:4, 1L) to give (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide (43.5g, 81%) as a colorless solid. The title compound was slurried with the previous batch (108.7g, 266.8mmol) prepared in the same manner and in ethyl acetate (1.0L) at 80 ℃ for 4 hours. The suspension was allowed to cool to room temperature and stirred for 4 days. The solid was filtered, washed with ethyl acetate (3 × 200mL) and dried under high vacuum at 50 ℃ for 24 hours to give (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide as a colorless solid (100.5g, 92%).1H NMR(300MHz,DMSO-d6) δ 1.38(t,3H),1.89-1.98(m,1H),2.15-2.26(m,1H),3.65(dd,1H),3.70-3.78(m,1H),3.85-3.92(m,2H),4.18(q,2H),4.46-4.55(m,1H),7.18(dd,1H),7.58(dd,1H),7.69(dd,1H),8.37(dd,1H),8.64(d,1H),8.95(d,1H),9.28(s,2H),9.39(d, 1H). MS (ES +)408.4(M + H). Melting point 177.5 ℃. C 21H21N5O4Elemental analysis of (2): calcd for C, 61.91; h, 5.20; n, 17.19; found C, 61.86; h, 5.18; and N, 17.30.

The solid form from this procedure was characterized by powder X-ray diffraction (PXRD) analysis and assigned form 1 of example DGAT2 i.

Alternative step 6 for the preparation of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide (example DGAT2i)

Acetonitrile (35mL), 2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) pyrimidine-5-carboxylic acid (5.0g, 15mmol), and (S) -tetrahydrofuran-3-amine hydrochloride (2.2g, 18mmol, 1.2 equiv.) were charged to a 100mL reactor. Diisopropylethylamine (18mL, 103mmol, 7.0 equiv.) was charged while maintaining the temperature at 20 ℃ to 30 ℃. A solution of propanephosphonic anhydride (T3P) (21mL, 30mmol, 2.0 equiv.) in acetonitrile was charged at a rate to maintain the temperature below 45 ℃. The reactor was heated to 40 ± 5 ℃ for 1 hour, followed by sampling to confirm completion of the reaction. The reaction was cooled to 20 ℃ to 25 ℃ and tetrahydrofuran (25mL) was added. Sodium bicarbonate solution (0.5M, 40mL) was charged and the mixture was stirred for 1 hour. The pH was checked and found to be 8.5. Ethyl acetate (40mL) was added and the mixture was stirred for 15 minutes. The mixture settled and the phases separated. The aqueous layer was transferred to a separatory funnel and back-extracted with ethyl acetate (100 mL). The organic phases were combined and washed with water (40 mL). The organic layer was partially transferred to a 100mL reactor and concentrated to low volume under vacuum. Methyl ethyl ketone (100mL) was added and the mixture was concentrated to a final volume of about 60 mL. The vacuum was removed and the slurry was heated to reflux and settled until the solids washed off the reactor walls. The slurry was cooled to 15 ℃ over 2 hours and granulated overnight. The solid was isolated by filtration and the reactor and filter cake were washed twice with methylethylketone (10 mL each). The solid was dried in a vacuum oven at 50 ℃ to give 4.86g (81%) of the desired product. The solid form from this program was characterized by PXRD analysis and assigned form 2 of example DGAT2 i.

Example conversion of form 2 to form 1 of DGAT2i

Form 2 (10.0g, 24.6mmol, 1.00 equiv.) of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide (example DGAT2i), methylethylketone (8.8mL/g, 88.0mL), and water (1.2mL/g, 12.0mL) were charged to a 100mL reactor. The reactor was heated to 50 ℃ over 30 minutes. Complete solution occurred at about 44 ℃. The reactor was cooled to 40 ℃ over 30 minutes, then seed crystal form 1 (0.050g, 0.123mmol, 0.0050 equiv.) of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide (example DGAT2i) was charged. After seeding, the cloudy slurry was stirred for 1 hour, then cooled to 5 ℃ over 2 hours, and then stirred at 5 ℃ for 12 hours. A control sample was sampled during the process and characterized by PXRD analysis to confirm that the solid was type 1. The slurry was filtered and the reactor and filter cake were washed with 0 ℃ methylethylketone (2.5mL/g, 25 mL). The solid was dried in a vacuum oven at 50 ℃ to give 8.15g (81.5%) of the desired product. The PXRD pattern of the desired product was consistent with form 1 of example DGAT2 i.

Powder X-ray diffraction:

powder X-ray diffraction analysis Using a Cu-equipped radiation Source (A) K α average wavelength) was performed with a Bruker AXS D8 Advance diffractometer equipped with a dual primary color (twin primary) using a gobel mirror. The diffracted radiation was detected with a PSD-Lynx Eye detector. Both the primary and secondary (secondary) primaries are equipped with a 2.5 soller slit. The X-ray tube voltage and current amount were set to 40kV and 40mA, respectively. Data was collected in a theta-theta goniometer using a scan speed of 6 seconds per step with 1000 steps from a locked conjugate scan (locked conjugate scan) of 3.0 to 40.0 degrees 2 theta. The samples were prepared by placing in a silicon low background sample holder (C79298a3244B 261). Data were collected using Bruker DIFFRAC Plus software. The analysis was performed with EVA Diffrac Plus software.

The PXRD data file is not processed until peak search. Using a peak search algorithm in the EVA software, a peak with a threshold value of 5 and a width value of 0.2 was selected. To visually check the output of the automated dispense to ensure effectiveness and to make manual adjustments if necessary. Peaks with a relative intensity of 3% or more are generally selected. Peaks that were not resolved or consistent with noise were also discarded. Typical errors associated with peak positions from PXRD as described by USP are in the range of +/-0.2 deg. (USP-941).

TABLE 9 characterization of crystalline materials example critical PXRD peaks for DGAT2i

Example DGAT2i form 1 Example type 2 of DGAT2i
2 Theta Angle (°) 2 Theta Angle (°)
5.3,7.7,15.4 6.5,9.3,13.6

FIG. 1 is a graph showing the characteristic x-ray powder diffraction pattern (vertical axis: intensity (CPS); horizontal axis: 2 θ (degrees)) for form 1 of the compound of example DGAT2 i.

FIG. 2 is a graph showing the characteristic x-ray powder diffraction pattern (vertical axis: intensity (CPS); horizontal axis: 2 θ (degrees)) for form 2 of the compound of example DGAT2 i.

Example FXRa ("tropiseto"):

intermediates

2- (trifluoromethoxy) benzaldoxime (I-1B). NH in water (120mL)2A stirred solution of oh.hcl (11.8g, 169.78mmol, 1.15 equiv.) is added to a solution of sodium hydroxide (7g, 175.00mmol, 1.19 equiv.) in water (120mL) at 0 ℃. The resulting solution was stirred at 0 ℃ for 10 min. A solution of 2- (trifluoromethoxy) benzaldehyde (28g, 147.29mmol, 1.00 eq) in ethanol (120mL) was then added. The resulting solution was allowed to stir at room temperature for an additional 1 h. The resulting solution was taken up in 500ml of H2Diluted O, extracted with 2 × 700mL ethyl acetate and the organic layers combined, washed with 2 × 300mL brine, dried over anhydrous sodium sulfate and concentrated under vacuum to give (E) -2- (trifluoromethoxy) benzaldehyde oxime as an off-white crystalline solid.

N-hydroxy-2- (trifluoromethoxy) imidobenzyl chloride (I-1C). NCS (22g, 166.04mmol, 1.12 equiv.) was slowly added to a stirred solution of (E) -2- (trifluoromethoxy) benzaldehyde oxime (30g, 146.27mmol, 1.00 equiv.) in N, N-dimethylformamide (300mL) to keep the internal temperature below 25 ℃. The reaction mixture was stirred at room temperature for 1 h. The resulting solution was diluted with water (300mL) and extracted with ethyl acetate (2X 500 mL). The organic layers were combined, washed with brine (5 × 300mL), dried over anhydrous sodium sulfate and concentrated in vacuo to give (Z) -2- (trifluoromethoxy) benzoyl chlorooxime as a pale yellow crystalline solid.

5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazole-4-carboxylic acid methyl ester (I-1D). Potassium carbonate (11g, 79.7mmol, 1.09 eq) was suspended in THF (100mL) and the mixture was stirred. A solution of methyl 3-cyclopropyl-3-oxopropanoate (11g, 77.5mmol, 1.06 eq.) in 50ml THF was added to the stirred mixture above and stirred at-10 deg.C for 30 min. A solution of (Z) -2- (trifluoromethoxy) benzoyl chlorooxime (17.6g, 73.3mmol, 1.00 eq.) in THF (50mL) was added to this reaction mixture at-5 deg.C, then allowed to stir at 35 deg.C for 6 h. The reaction mixture was taken up in 200mL of H2Diluted O and extracted with ethyl acetate (2X 300 mL). The organic layer was washed with brine (2 × 200mL), dried over anhydrous sodium sulfate, concentrated under vacuum, and then purified by silica gel column chromatography using ethyl acetate/petroleum ether (1:100 to 1:20) eluent to give methyl 5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazole-4-carboxylate as a white solid.

(5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) -methanol (I-1E). A250 mL round bottom flask was purged with nitrogen and LiAlH in tetrahydrofuran (50mL) was added4(2.5g, 65.8mmol, 2.87 equiv.) of the suspension. A solution of methyl 5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazole-4-carboxylate (7.5g, 22.9mmol, 1.00 equiv.) in tetrahydrofuran (50mL) was then added dropwise at-10 ℃. The resulting reaction mixture was stirred at-10 ℃ for 30 min. When the reaction was complete, it was treated with 3mL ethyl acetate followed by 3mL water and O 1mL 15% aqueous NaOH was quenched, all while maintaining vigorous stirring. Passing the obtained white precipitate throughFilter and wash the filter cake with 200mL ethyl acetate. The filtrate was washed with brine (2 × 100mL), dried over anhydrous sodium sulfate, and concentrated in vacuo. This gave 7g of (5-cyclopropyl-3-(2- (trifluoromethoxy) phenyl) isoxazol-4-yl) methanol. (1H-NM(300MHz,CDCl3)δ7.56(m,2H),7.41(m,2H),4.50(s,2H),2.20(m,1H),1.72(s,1H,--OH)1.11-1.28(m,4H)。

4- (bromomethyl) -5-cyclopropyl-3- (2- (trifluoromethoxy) -phenyl) isoxazole (I-1F). (5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) -methanol (4g, 13.3mmol), triphenylphosphine (5.6g, 20mmol, 1.5 equiv.) and dichloromethane (40mL) were placed in a 100mL round bottom flask. The mixture was stirred until completely dissolved and then slowly added dropwise via cannula to a stirred solution of carbon tetrabromide (6.6g, 20mmol, 1.5eq) in dichloromethane (20 ml). The mixture was stirred for 1 hour, then the solvent was evaporated in vacuo. The crude residue was purified by silica gel chromatography using a gradient of 0 to 50% ethyl acetate/hexane. The desired product was obtained as a colorless oil. MS M/z 361.9/363.9(M +1, Br)79/Br81Isotope patterns).

3- ((5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) methoxy) -8-azabicyclo [3.2.1 ]Octane-8-carboxylic acid tert-butyl ester (I-1G). A250 mL flask was purged with nitrogen and then charged with N-Boc-nortropine (2.9g, 12.8mmol), 18-crown-6 (3.4g, 12.8mmol) and anhydrous tetrahydrofuran (80 mL). Potassium tert-butoxide (2.9g, 25.6mmol) was added in small portions and the mixture was stirred vigorously under nitrogen for 1 h. 4- (bromomethyl) -5-cyclopropyl-3- (2- (trifluoromethoxy) -phenyl) isoxazole (4.18g, 11.6mmol) was dissolved in anhydrous tetrahydrofuran (20mL) and added dropwise, and the reaction mixture was stirred under positive nitrogen pressure overnight. The solvent was removed in vacuo and the mixture was diluted with water (100mL) and ethyl acetate (100 mL). The organic layer was separated and dried over anhydrous MgSO4Dried and evaporated in vacuo. The crude residue was purified by silica gel chromatography using a gradient of 0 to 100% ethyl acetate in hexanes to give the desired product as a yellow oil. MS M/z 509.2(M + 1).

4- ((8-azabicyclo [ 3.2.1)]Octyl-3-oxy) methyl) -5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazole (I-1H). Reacting 3- ((5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) -methoxy) -8-azabicyclo [3.2.1]Octane-8-carboxylic acidTert-butyl ester was dissolved in 30mL of a 20% solution of trifluoroacetic acid in dichloromethane. The solution was stirred at room temperature for 1h and the solvent was evaporated. The residue was dissolved in ethyl acetate (125mL), washed with a saturated solution of sodium bicarbonate (100mL), and the organic layer was washed with anhydrous MgSO 4Dried and evaporated in vacuo. The crude residue was purified by silica gel chromatography using a gradient of 0 to 20% ethanol/dichloromethane to give the desired product as a colorless oil. MS M/z 409.2(M + 1).

1H NMR(DMSOd6,400MHz);δ8.51(br s,1H,NH),7.72-7.68(m,1H),7.64(dd,J=7.6,1.8Hz,1H),7.58-7.52(m,2H),4.33(s,2H),3.81(bs,2H),3.55(t,J=4.5Hz,1H),2.36-2.33(m,1H),1.98(app dt,J=14.8,4.0Hz,2H),1.91-1.76(m,6H),1.14-1.07(m,4H)。

4- ((8-azabicyclo [ 3.2.1)]Oct-3-oxy) methyl) -5-cyclopropyl-3- (2- (trifluoromethyl) phenyl) isoxazole (I-1I) was prepared according to the same procedure. MS M/z 393.2(M + 1);1H NMR(DMSOd6,400MHz);δ8.51(br s,1H,NH),7.92(d,J=8.0,1.8Hz,1H),7.81(app t,J=7.1Hz,1H),7.78(app t,J=7.1Hz,1H),7.58(d,J=7.3Hz,1H),4.24(s,2H),3.81(bs,2H),3.52(t,J=3.7Hz,1H),2.36-2.33(m,1H),1.92(app dt,J=14.8,4.0Hz,2H),1.81-1.69(m,6H),1.14-1.09(m,4H)。

4- ((8-azabicyclo [ 3.2.1)]Oct-3-oxy) methyl) -5-cyclopropyl-3- (2- (difluoromethoxy) phenyl) isoxazole (I-1J) was prepared according to the same procedure. MS M/z 391.3(M + 1);1H NMR(CDCl3,400MHz);δ9.10(br s,1H,NH),7.32(app t,J=8.4Hz,2H),7.26(app d,J=8.4Hz,2H),6.44(t,J=74Hz,1H,CHF2),4.32(s,2H),3.82(bs,2H),3.56(t,J=4.0Hz,1H),2.32(app dt,J=15.2,4.6Hz,2H),2.08-2.04(m,1H),1.98-1.89(m,4H),1.78(app br d,J=15.9Hz,2H),1.26-1.20(m,2H),1.14-1.09(m,2H)。

4- ((8-azabicyclo [ 3.2.1)]Octyl-3-oxy) methyl) -5-cyclopropyl-3- (2, 6-difluorophenyl) isoxazole (I-1K) was prepared according to the same procedure. MS M/z 361.2(M + 1);1H NMR(CDCl3,400MHz);δ9.18(br s,1H,NH),7.48-7.40(m,1H),7.06-6.99(m,2H),4.31(s,2H),3.82(bs,2H),3.59(t,J=4.7Hz,1H),2.16(app dt,J=15.9,4.0Hz,2H),2.09-2.02(m,1H),1.98-1.92(m,4H),1.76(app br d,J=15.2Hz,2H),1.26-1.19(m,2H),1.15-1.09(m,2H)。

example FXRa: top-held halyard

Methyl 2- (3- ((5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) methoxy) -8-azabicyclo [3.2.1] oct-8-yl) -4-fluorobenzo [ d ] thiazole-6-carboxylate (I-1A). 4- ((8-azabicyclo [3.2.1] oct-3-oxy) methyl) -5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazole (I-1H) (0.525g, 1.29mmol), 3.6mL of N, N-dimethylacetamide, cesium carbonate (1.08g, 3.31mmol), and methyl 2-bromo-4-fluorobenzo [ d ] thiazole-6-carboxylate (1.12g, 3.87mmoles) were added sequentially to a 25mL round bottom flask equipped with a stir bar. After the resulting slurry was stirred at room temperature for 10 minutes, the mixture was then warmed to 60 ℃ and stirred for 1 h. The reaction slurry was allowed to cool to RT, diluted with 200mL ethyl acetate and washed with water (3 × 30 mL). The organic extracts were concentrated in vacuo and purified directly using normal phase silica gel chromatography (40g silica gel column) with a 15min gradient of 10% to 60% ethyl acetate/hexane. The desired fractions were concentrated in vacuo and the resulting residue crystallized on standing to give the desired product as a white crystalline solid.

2- [3- ({ 5-cyclopropyl ] -3- [2- (trifluoromethoxy) phenyl-1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid (I-1B). Methyl 2- (3- ((5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) methoxy) -8-azabicyclo [3.2.1] oct-8-yl) -4-fluorobenzo [ d ] thiazole-6-carboxylate (0.55g, 0.89mmol), 4.0mL THF, 2.0mL MeOH and 3N aqueous KOH (1mL, 3mmol) were added sequentially to a 25mL round bottom flask equipped with a stir bar. The resulting homogeneous solution was stirred at 70 ℃ for 1 hour, cooled to RT, and then quenched with AcOH (ca. 0.2mL glacial acetic acid, 3mmoles) until pH 6 was achieved (Whatman grade pH paper). At this point the reaction was diluted with ethyl acetate (40mL) and washed with water (3 times, 5 mL). The ethyl acetate fraction was concentrated in vacuo to give an oily residue. Then 6mL MeOH was added to the resulting oil. The oil dissolved rapidly and crystallization started immediately. After standing for 2.5hr, the mother liquor was withdrawn and the crystals were washed (3X 2mL ice-cold MeOH). The crystals were dried via vacuum (10mm Hg pressure, overnight at 45 ℃), then recrystallized from acetonitrile, filtered and dried under vacuum to give the desired product 2- (3- ((5-cyclopropyl-3- (2- (trifluoromethoxy) phenyl) isoxazol-4-yl) -methoxy) -8-azabicyclo [3.2.1] oct-8-yl) -4-fluorobenzo [ d ] thiazole-6-carboxylic acid.

2- [3- ({ 5-cyclopropyl-3- [2- (trifluoromethyl) phenyl ] -1, 2-oxazol-4-yl } methoxy) -8-azabicyclo [3.2.1] oct-8-yl ] -4-fluoro-1, 3-benzothiazole-6-carboxylic acid (I-2B). Examples 1-2A and the corresponding acids 1-2B can be prepared according to the same procedure from the reaction of intermediate 4- ((8-azabicyclo [3.2.1] oct-3-oxy) methyl) -5-cyclopropyl-3- (2- (trifluoromethyl) phenyl) isoxazole.

Physical data Ex MS (m/z),1H NMR

top-held halyard

Elemental analysis (C)30H29F4N3O6S): c56.69, H4.60, N6.61; measured value: c56.79, H4.61, N6.65. MS M/z 604.2(M + 1);1H NMR(MeOD,400MHz)δ8.03(d,J=1.6Hz,1H),7.57-7.53(m,2H),7.49(dd,J=8.1,1.8Hz,2H),7.41(app t,J=7.6,1H),4.31(s,2H),4.22(broad s,2H),3.50(t,J=4.4Hz,1H),2.22-2.15(m,1H),2.00(app dt,J=14.8,4.0Hz,2H),1.91-1.81(m,4H),1.75(d,J=14.4,2H),1.10-1.05(m,4H)。

example KHKi (KHKi compound):

[ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid

Step 1:

{ (1R,5S,6S) -3- [ 2-chloro-6- (trifluoromethyl) pyrimidin-4-yl ] -3-azabicyclo [3.1.0] hex-6-yl } acetic acid methyl ester

2, 4-dichloro-6- (trifluoromethyl) pyrimidine (145.7g, 671.5mmol) in DCM (50mL) was added dropwise to a solution of methyl (1R,5S,6S) -3-azabicyclo [3.1.0] hex-6-ylacetate hydrochloride (120.2g, 627.2mmol) in DCM (1250mL) at-72 ℃; the addition funnel was rinsed with DCM (50ml) and the rinse was added to the reaction flask. DIPEA (273mL, 1570mmol) was added over 10min to maintain the reaction temperature between-70 ℃ and-60 ℃. The mixture was stirred at-65 ℃ to-63 ℃ for 1h and then warmed to 25 ℃ over 3 h. The resulting clear solution was concentrated to an initial volume of-1/5. MTBE (700mL) and heptane (700mL) were added to the thick slurry obtained and the resulting slurry was stirred at 25 ℃ for 10min, then the solids were filtered off and washed with MTBE-heptane (4: 1). The combined mother liquors were concentrated in vacuo to an oil, which was combined with heptane (1200 mL). The heterogeneous mixture obtained was stirred at 25 ℃ for 2.5 days. A white solid formed. The liquid was decanted and the solid was washed with heptane (200mL) and dried under a stream of nitrogen. The title product obtained was used in the next step without additional purification.

1H NMR(400MHz,CDCl3)δ:6.47(s,1H),4.07(d,1H),3.71(s,3H),3.53-3.68(m,3H),2.36-2.49(m,1H),2.21-2.34(m,1H),1.60-1.73(m,2H),0.88-0.97(m,1H)。

Step 2

Reacting { (1R,5S,6S) -3- [ 2-chloro-6- (trifluoromethyl) pyrimidin-4-yl group from step 1]-3-azabicyclo [3.1.0]Methyl hex-6-yl } acetate was dissolved in acetonitrile (1500mL) and [ (1R,4S) -7, 7-dimethyl-2-oxobicyclo [2.2.1 ] was added]Hept-1-yl]Methanesulfonic acid (2S) -2-methylazetidinium salt (223.0g, 735 mmol). The mixture was stirred at 60 ℃ and DIPEA (77.0mL, 442mmol) was added over a period of 3 h. The mixture was stirred for 3h, then DIPEA (180mL, 1.03mol) was added over 3h and the mixture was stirred at 60 ℃ for 18 h. Addition of additional [ (1R,4S) -7, 7-dimethyl-2-oxobicyclo [2.2.1 ]]Hept-1-yl](2S) -2-methylazetidinium mesylate (18.0g, 59mmol) and the mixture stirred at 60 ℃ for a further 18 h. Concentrating the mixture1/4 in initial volume and the resulting yellow oil was partitioned between 500mL water, 400mL heptane, and 400mL MTBE. The aqueous phase was separated and extracted again with a mixture of MTBE-heptane (1:1) (2X 150 mL). The combined organic extracts were taken up in 120mL of saturated NaHCO3(120mL) and then with SiO2(70g) And anhydrous MgSO4(70g) And (4) stirring. The solid was filtered off and the clear solution was concentrated to give 216.6g of example FXRa as a colorless oil.

MS(ES+):371.1(M+H)。1H NMR(400MHz,CDCl3)δ:5.91(s,1H),4.37-4.48(m,1H),3.87-4.05(m,3H),3.70(s,3H),3.50-3.64(m,1H),3.41-3.50(m,2H),2.33-2.42(m,1H),2.31(d,2H),1.88-1.99(m,1H),1.52-1.59(m,2H),1.49(d,3H),0.88-0.96(m,1H)。

Step 3

A solution of sodium hydroxide (35.1g, 877mmol) in water (70mL) was added in small portions with stirring to unpurified [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] in methanol (650mL) at 5 ℃ to 15 ℃ ]-6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0]Hex-6-yl]A stirred solution of methyl acetate. The mixture became clear within 30 min. The clear solution was stirred at RT for 3h, then concentrated to an initial volume of-1/3, and the residue was diluted with water (750mL) and brine (250mL), then washed with a mixture of MTBE (260mL) and heptane (130 mL). The organic wash was discarded. The aqueous phase was washed with a mixture of MTBE-heptane (2:1) (2X 300mL) and the organic layer discarded. The aqueous layer was then combined with MTBE (250mL) and heptane (250mL) and cooled to 0 ℃. 6M aq.HCl (130mL) was added slowly with stirring at 0 ℃ to 4 ℃ followed by 1M aq.KHSO4(150mL) and the resulting mixture was stirred for 15 min. The organic phase was separated and the aqueous phase was additionally extracted with a mixture of MTBE (170mL) and heptane (170 mL). The combined organic extracts were washed with a water-brine (1:1) mixture (150mL) over anhydrous MgSO4(60g) And SiO2(60g) Dried, filtered and concentrated to give a colorless oil. It was combined with another batch prepared using the same conditions on the same scale (concentrated solution in MTBE). The combined MTBE solution was concentrated in vacuo, followed by the addition of heptane (2000mL) and re-concentration of the increasing volume suspension To obtain the desired product (406.0 g). A portion of this material (196g) was dissolved in slowly stirred MTBE (220mL) at 60 ℃ to 63 ℃ and heptane (1500mL) was added at 55 ℃ to 60 ℃. The mixture was seeded with the title compound in crystalline form (50 mg). The mixture was stirred at 60 ℃ for 30min, followed by addition of additional heptane (1700mL) over a 20min period. The heterogeneous mixture was stirred at 60 ℃ for 2h, then slowly cooled to 25 ℃ and stirred for 20 h. A small amount of solid adhered to the flask wall and was easily transferred to the liquid phase with a spatula and the mixture was stirred at 25 ℃ for a further 24 h. The solid was filtered off, washed with 5% MTBE in heptane and dried in vacuo at 50 ℃ over 48h to give example KHKi (178.2g, 73% over 3 steps) as a white crystalline solid. The crystalline solid of example KHKi was also obtained using similar purification conditions without seeding.

MP:122-123℃,[α]D+86.3°(CDCl3,c=1.37)。MS(ES+):357.3(M+H)。1H NMR(400MHz,CDCl3)δ:10.84(br.s,1H),5.92(s,1H),4.38-4.51(m,1H),3.89-4.10(m,3H),3.53-3.66(m,1H),3.41-3.53(m,2H),2.30-2.46(m,3H),1.94(ddt,1H),1.55-1.63(m,2H),1.50(d,3H),0.94(m,1H)。

Powder X-ray diffraction analysis was performed using a Bruker AXS D4Endeavor diffractometer equipped with a Cu radiation source. The divergence slit is set at 0.6mm and the secondary optics uses a variable slit. The diffracted radiation was detected with a PSD-Lynx Eye detector. The X-ray tube voltage and current amount were set to 40kV and 40mA, respectively. At Cu wavelength And 3.0 to 40.0 degrees 2 theta using a step size of 0.020 degrees and a step time of 0.3 seconds. The sample was prepared for placement in a silicon low background sample holder and rotated during collection. Data were collected using Bruker DIFFRAC Plus software and analysis was performed with EVA DIFFRAC Plus software.

The PXRD data file is not processed until peak search. Using a peak search algorithm in the EVA software, a peak with a threshold value of 1 is selected and a width value of 0.3 is used for preliminary peak assignment. To visually check the output of the automated dispense to ensure effectiveness and to make manual adjustments if necessary. Peaks with a relative intensity of 3% or more are generally selected. Peaks that were not resolved or consistent with noise were also discarded. Typical errors associated with peak positions from PXRD described in USP and JP are at most +/-0.2 °.

Characteristic peaks of the crystalline form free acid of example KHKi include 2 theta angle (°) values of about 9.0, 10.4, 15.0, and 21.4+/-0.2 °. Example yet another embodiment of the crystalline form of the free acid of KHKi is wherein the characteristic peaks comprise 2 theta angle (°) values of about 9.0, 15.019.6, 21.4, and 26.5+/-0.2 °. Yet another embodiment of the crystalline form of the free acid of example KHKi is wherein the characteristic peaks comprise 2 theta angle (°) values of about 9.0, 10.4, 11.5, 15.0, 16.5, 19.6,21.4, and 26.5+/-0.2 °. Yet another embodiment of the crystalline form of the free acid of example KHKi is wherein the characteristic peaks comprise 2 theta angle (°) values of about 10.4, 11.5, 15.0, 19.6, and 26.5+/-0.2 °. Table 10 provides a list of PXRD peaks for the crystalline free acid of example KHKi, +/-0.2 ° is applicable for the peaks. Figure 1 provides a PXRD pattern of the crystalline free acid of example KHKi.

TABLE 10 PXRD Peak List of crystalline free acids of example KHKi

2 Θ angle (°) ° Strength (%) 2 Θ angle (°) ° Strength (%) 2 Θ angle (°) ° Strength (%)
9.0 37 18.3 85 25.9 20
10.4 17 18.8 17 26.5 30
11.5 16 18.9 7 27.1 9
13.5 10 19.6 100 27.6 5
13.9 5 21.4 36 28.1 9
15.0 45 22.8 22 29.1 6
16.5 23 22.9 15 30.1 10
17.3 4 23.3 55 30.5 6
17.7 14 23.7 6 31.6 4
18.1 40 25.7 7

Additional pharmacology and pharmacology data

Pharmacology of 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid (example ACCi)

4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4' -piperidine)]-1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid is a reversible dual ACC1/2 inhibitor currently being developed for the treatment of NASH with liver fibrosis. The compounds were designed to have an asymmetric distribution in the liver, demonstrating a ≥ 100-fold asymmetric liver distribution in both rat and monkey, as would be expected to inhibit endogenous lipogenesis (DNL) in the liver and stimulate fatty acid oxidation to a greater extent than in peripheral tissues. Dose-response experiments for inhibition of human and rat ACC isozymes were performed at saturating concentrations of Adenosine Triphosphate (ATP), where the inhibitory concentration (IC50) approximates the equilibrium dissociation constant of the enzyme (Ki) for a non-competitive inhibitor. Example the Compound of ACCi inhibits [ 2 ] in a concentration-dependent manner14C]Incorporation of carbonate [ sic ], [ solution of a copolymer of (A) and (B)14C]-malonyl-CoA. In rats, ACC inhibitors cause dose-and free plasma concentration-dependent reduction of hepatic malonyl-CoA. In humans, 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4' -piperidine) ]Administration of-1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid was shown to inhibit liver DNL in phase 1 studies in healthy adult individuals (Bergman et al, 2017); in addition, the drug is expected to stimulate hepatic fatty acid oxidation and thus reduce fat accumulation in the liver. This inhibition of hepatic DNL is hypothesized to result in the reduction and normalization of excess DNL observed in non-alcoholic fatty liver disease (NAFLD). In addition, 4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4' -piperidine)]-1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid also has the potential for anti-inflammatory effects in nonalcoholic steatohepatitis (NASH).

Pharmacological (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide (example DGAT2i)

(S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is an oral small molecule DGAT2 inhibitor, hypothesized to reduce hepatic Triglyceride (TG) synthesis and hepatic lipid burden in NAFLD and NASH. The compounds inhibited in vitro as assessed biochemically. The selectivity of (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide is supported by an in vitro study that demonstrated, on a biochemical assessment, a > 2000-fold selectivity over related acyltransferases, including recombinant human DGAT1 (hggat 1), monoacylglycerol acyltransferase 2(MGAT2), and MGAT3, and mouse MGAT 1. (S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide demonstrated a robust dose-dependent reduction in plasma triacylglycerols in rats fed a sucrose diet. In a longer study in rats fed a western diet, the compound reduced both plasma triacylglycerol and liver lipid accumulation.

Pharmacology of tropiseto (example FXRa)

The pharmacological activity of tropiseto has been proposed as a target for the treatment of NASH (caroou, 2008, Porez et al, 2012). Clinical validation of FXR agonists for the treatment of NASH was demonstrated with obeticholic acid (semisynthetic bile acid) (Neuschwander-Tetri et al, 2015). The nuclear receptor FXR (also known as bile acid receptor (BAR; NR1H4)) is responsible for the regulation of bile acid production, conjugation and elimination. Tropisetol is a potent and selective non-steroidal FXR agonist. Both single and multiple doses of tropisetron resulted in robust, dose-dependent increases in circulating FGF19 levels.

Pharmacological [ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid (example KHKi)

[ (1R,5S,6R) -3- {2- [ (2S) -2-methylazetidin-1-yl ] -6- (trifluoromethyl) pyrimidin-4-yl } -3-azabicyclo [3.1.0] hex-6-yl ] acetic acid is a potent, reversible inhibitor of human KHK-C. The component of dietary sugar, fructose (non-glucose), is unique in its ability to promote the properties of metabolic syndrome, including steatosis, insulin resistance and obesity. This compound reduces the incidence of fructose-induced steatosis in a Sprague Dawley rat model fed dietary fructose.

Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application for all purposes.

It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

291页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:使用曲美替尼和标志物治疗神经退化性疾病的给药方法和剂量方案

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!