GLP-1 receptor agonists and uses thereof

文档序号:883453 发布日期:2021-03-19 浏览:14次 中文

阅读说明:本技术 Glp-1受体激动剂及其用途 (GLP-1 receptor agonists and uses thereof ) 是由 G·E·阿斯普内斯 S·W·巴利 J·M·库尔托 D·J·艾蒙兹 M·E·弗拉纳根 二木建 于 2019-06-11 设计创作,主要内容包括:本文提供了作为GLP-1R激动剂的苯并咪唑以及4-氮杂-、5-氮杂-、和7-氮杂-苯并咪唑的6-甲酸,制备所述化合物的方法,以及包括将所述化合物施用于需要其的哺乳动物的方法。(Provided herein are benzimidazoles that are GLP-1R agonists as well as 6-carboxylic acids of 4-aza-, 5-aza-, and 7-aza-benzimidazoles, methods of making the compounds, and methods comprising administering the compounds to a mammal in need thereof.)

1. A compound of the formula I, wherein,

or a pharmaceutically acceptable salt thereof, wherein

R is F, Cl, or-CN;

p is 0 or 1;

ring A is phenyl or 6-membered heteroaryl;

m is 0, 1, 2, or 3;

each R1Independently selected from: halogen, -CN, -C1-3Alkyl, or-OC1-3Alkyl radical, wherein C1-3Alkyl and OC 1-3The alkyl group of the alkyl group is substituted with 0 to 3F atoms;

R2is H or-C1-3Alkyl, wherein alkyl is substituted with 0 to 1 OH;

each R3Independently F, -OH, -CN, -C1-3Alkyl, -OC1-3Alkyl, or-C3-4Cycloalkyl, or 2R3Can be cyclized together to form-C3-4Spirocycloalkyl, wherein if valency allows, C1-3Alkyl and OC1-3The alkyl, cycloalkyl, or spirocycloalkyl groups of the alkyl group may be substituted with 0 to 3F atoms and with 0 to 1-OH;

q is 0, 1 or 2;

X-L is N-CH2、CHCH2Or cyclopropyl;

y is CH or N;

R4is-C1-3Alkyl, -C0-3alkylene-C3-6Cycloalkyl, -C0-3alkylene-R5or-C1-3alkylene-R6Wherein if allowed by valence, said alkyl group may be substituted with 0 to 3 substituents independently selected from 0 to 3F atoms and 0 to 1 substituent selected from-C0-1alkylene-CN, -C0-1alkylene-ORO、-SO2-N(RN)2、-C(O)-N(RN)2、-N(C=O)(RN) and-N (R)N)2Substituted with the substituent(s); and is

Wherein said alkylene and cycloalkyl groups may be independently substituted, if valency permits, with 0 to 2 substituents independently selected from 0 to 2F atoms and 0 to 1 substituent selected from-C0-1alkylene-CN, -C0-1alkylene-OROand-N (R)N)2Substituted with the substituent(s);

R5is a 4-to 6-membered heterocycloalkyl group, wherein if valency permits, said heterocycloalkyl group may be substituted with 0 to 2 substituents independently selected from the group consisting of:

0 to 1 oxo (= O),

0 to 1-CN group, and the N-CN group,

0 to 2F atoms, and

0 to 2 are independently selected from-C1-3Alkyl and-OC1-3Alkyl substituents, wherein if valency permits, C1-3Alkyl and OC1-3The alkyl group of the alkyl group may be substituted with 0 to 3 substituents independently selected from the group consisting of:

from 0 to 3 atoms of F,

0 to 1-CN, and

0 to 1-ORO

R6Is a 5-to 6-membered heteroaryl, wherein the heteroaryl, if valency permits, may be substituted with 0 to 2 substituents independently selected from:

0 to 2 halogen atoms in the molecule of a halogen,

0 to 1 is selected from-OROand-N (R)N)2A substituent of (a), and

0 to 2-C1-3Alkyl, wherein the alkyl may be substituted with 0 to 3 substituents independently selected from the group consisting of:

0 to 3F atoms, and

0 to 1-ORO

Each ROIndependently is H, or-C1-3Alkyl radical, wherein C1-3Alkyl groups may be substituted with 0 to 3F atoms;

each RNIndependently is H, or-C1-3An alkyl group;

Z1、Z2and Z3Each is-CRZOr is or

Z1、Z2And Z3One of which is N and the other two are-CRZ(ii) a And

each RZIndependently H, F,Cl, or-CH3

2. The compound of claim 1, wherein the compound is of formula II

Or a pharmaceutically acceptable salt thereof, wherein

R is F;

p is 0 or 1;

ring a is phenyl or pyridyl;

m is 0, 1, or 2;

each R1Independently selected from: halogen, -CN, -C1-3Alkyl, or-OC1-3Alkyl radical, wherein C1-3Alkyl and OC1-3The alkyl group of the alkyl group is substituted with 0 to 3F atoms;

R2is H or CH3

X-L is N-CH2Or cyclopropyl;

y is CH or N;

Z3is-CRZOr N; and

RZis H, F, Cl, or-CH3

3. A compound according to claim 1 or claim 2, wherein the compound is of formula III

Or a pharmaceutically acceptable salt thereof, wherein

Ring a is phenyl or pyridyl;

m is 0, 1, or 2;

each R1Independently selected from F, Cl, or-CN;

R2is H or CH3(ii) a And

y is CH or N.

4. The compound of any one of claims 1-3, wherein R4is-CH2-R5Wherein R is5Is a 4-to 5-membered heterocycloalkyl group, wherein if valency permits, said heterocycloalkyl group may be substituted with 0 to 2 substituents independently selected from the group consisting of:

0 to 2F atoms, and

0 to 1 is selected from-OCH3and-CH2OCH3A substituent of (1);

or a pharmaceutically acceptable salt thereof.

5. The compound of any one of claims 1-3, wherein R4is-CH2-R6Wherein R is6Is a 5-membered heteroaryl, wherein the heteroaryl, if valency permits, may be substituted with 0 to 2 substituents independently selected from:

0 to 2 halogens, wherein the halogens are independently selected from F and Cl,

0 to 1-OCH3And are and

0 to 1-CH3、-CH2CH3、-CF3or-CH2CH2OCH3

Or a pharmaceutically acceptable salt thereof.

6. The compound of any one of claims 1-5, wherein R2Is H, or a pharmaceutically acceptable salt thereof.

7. The compound of claim 6, wherein said compound is

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; or

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,

or a pharmaceutically acceptable salt thereof.

8. The compound of claim 6, wherein said compound is

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, or

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,

or a pharmaceutically acceptable salt thereof.

9. A compound which is 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, or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt.

10. A compound which is 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 as the free acid.

11. A compound which is

Or a pharmaceutically acceptable salt thereof.

12. The compound of any one of claims 1-5, wherein R2Is CH3Or a pharmaceutically acceptable salt thereof.

13. The compound of claim 12, wherein said compound is

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-1-yl)H-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- [ (2 S) -Oxetazedin-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; or

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;

or a pharmaceutically acceptable salt thereof.

14. The compound of claim 12, wherein said compound is

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; or

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;

or a pharmaceutically acceptable salt thereof.

15. A compound which is 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, or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt.

16. A compound which is 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 as the free acid.

17. A compound which is

Or a pharmaceutically acceptable salt thereof.

18. The compound of claim 12, wherein said compound is

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; or

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;

or a pharmaceutically acceptable salt thereof.

19. The compound of claim 12, wherein said compound is

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; or

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,

or a pharmaceutically acceptable salt thereof.

20. A compound which is the following compound as a free 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; or

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.

21. A compound which is

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; or

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;

or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt.

22. A compound which is 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.

23. A crystalline form of an anhydrous 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 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 (form I) having a powder X-ray diffraction pattern (CuK α radiation) comprising at least two characteristic peaks selected from 3.7 ± 0.2 ° in terms of 2 Θ; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and 16.9 ± 0.2.

24. A crystalline form according to claim 23, having a powder X-ray diffraction pattern comprising peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and at least three characteristic peaks at 16.9 ± 0.2.

25. A crystalline form according to claim 23, having a powder X-ray diffraction pattern comprising peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and at least four characteristic peaks at 16.9 ± 0.2.

26. A crystalline form according to claim 23, having a powder X-ray diffraction pattern comprising peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and at least five characteristic peaks at 16.9 ± 0.2.

27. A crystalline form according to claim 23, having an X-ray powder diffraction pattern comprising characteristic peaks, in terms of 2 Θ, at 3.7 ± 0.2 and 7.3 ± 0.2 degrees.

28. A crystalline form according to claim 23, having a refractive index that is between 3.7 ± 0.2 ° with respect to 2 Θ; an X-ray powder diffraction pattern for peaks at 7.3 + -0.2 and 14.7 + -0.2.

29. The crystalline form of claim 27 or 28, wherein the X-ray powder diffraction pattern further comprises at least one residue selected from 8.5 ± 0.2 ° in terms of 2 Θ; 10.1 ± 0.2; and peaks at 16.9 ± 0.2.

30. A crystalline form according to claim 29, having a refractive index that is between 3.7 ± 0.2 ° with respect to 2 Θ; 7.3 ± 0.2; 14.7 ± 0.2; and a powder X-ray diffraction pattern of peaks at 16.9 ± 0.2.

31. A crystalline form according to claim 30, having a refractive index that comprises at 3.7 ± 0.2 ° in terms of 2 Θ; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and a powder X-ray diffraction pattern of peaks at 16.9 ± 0.2.

32. The crystalline form of any one of claims 23 to 31, having a powder X-ray diffraction pattern substantially as shown in figure 1.

33. A crystalline form of an anhydrous 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 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 (form a) having a powder X-ray diffraction pattern (CuK α radiation) comprising at least two characteristic peaks selected from 7.7 ± 0.2 in terms of 2 Θ; 15.2 ± 0.2; (vii) 15.7 + 0.2 and 17.6 + 0.2.

34. The crystalline form of claim 33, having a powder X-ray diffraction pattern comprising at least three characteristic peaks, in terms of 2 Θ, selected from 7.7 ± 0.2; 15.2 ± 0.2; (vii) 15.7 + 0.2 and 17.6 + 0.2.

35. A crystalline form of claim 33, having an X-ray powder diffraction pattern comprising characteristic peaks, in terms of 2 Θ, at 7.7 ± 0.2 ° and 17.6 ± 0.2 ℃.

36. A crystalline form according to claim 33, having a refractive index that is between 7.7 ± 0.2 ° with respect to 2 Θ; 15.2 ± 0.2; and a powder X-ray diffraction pattern of peaks at 17.6 ± 0.2.

37. A crystalline form according to claim 33, having a refractive index that is between 7.7 ± 0.2 ° with respect to 2 Θ; 15.2 ± 0.2; and a powder X-ray diffraction pattern of peaks at 15.7 ± 0.2.

38. A crystalline form according to claim 33, having a refractive index that is between 7.7 ± 0.2 ° with respect to 2 Θ; 15.2 ± 0.2; a powder X-ray diffraction pattern for peaks at 15.7 + -0.2 and 17.6 + -0.2.

39. The crystalline form of any one of claims 33 to 38, having a powder X-ray diffraction pattern substantially as shown in figure 2.

40. A pharmaceutical composition comprising a compound of any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

41. A method of treating cardiovascular metabolism and related diseases or disorders, comprising administering to a mammal in need of such treatment a therapeutically effective amount of a compound of any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, wherein the disease or disorder is selected from: T1D, T2DM, pre-diabetes, idiopathic T1D, LADA, EOD, YOAD, MODY, malnutrition-related diabetes, gestational diabetes, hyperglycemia, insulin resistance, hepatic insulin resistance, impaired glucose tolerance, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, adipocyte dysfunction, visceral fat deposition, sleep apnea, obesity, eating disorders, weight gain due to use of other agents, excessive carbohydrate craving, dyslipidemia, hyperinsulinemia, NAFLD, NASH, fibrosis, cirrhosis, hepatocellular carcinoma, cardiovascular disease, arteriosclerosis, coronary artery disease, peripheral vascular disease, hypertension, endothelial dysfunction, impaired vascular compliance, congestive heart failure, myocardial infarction, stroke, hemorrhagic stroke, ischemic stroke, traumatic brain injury, cerebral ischemia, stroke, Pulmonary hypertension, restenosis following angioplasty, intermittent claudication, postprandial lipemia, metabolic acidosis, ketosis, arthritis, osteoporosis, Parkinson's disease, left ventricular hypertrophy, peripheral arterial disease, macular degeneration, cataracts, glomerulosclerosis, chronic renal failure, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, transient ischemic attack, vascular restenosis, impaired glucose metabolism, conditions of impaired fasting plasma glucose, hyperuricemia, gout, erectile dysfunction, skin and connective tissue disorders, psoriasis, foot ulcers, ulcerative colitis, hyperpao B lipoproteinemia, Alzheimer's disease, schizophrenia, impaired cognitive function, inflammatory bowel disease, short bowel syndrome, Crohn's disease, colitis, irritable bowel syndrome, polycystic ovary syndrome, prophylaxis or treatment of atherosclerosis, transient ischemic attack, vascular restenosis, impaired glucose metabolism, impaired fasting plasma glucose, impaired glucose metabolism, stroke, And treatment of addiction.

42. A pharmaceutical composition comprising the crystalline form of any one of claims 23 to 39 and a pharmaceutically acceptable excipient.

43. A method of treating cardiovascular metabolism and related diseases or disorders, comprising administering to a mammal in need of such treatment a therapeutically effective amount of the crystalline form of any one of claims 23 to 39, wherein the disease or disorder is selected from the group consisting of: T1D, T2DM, pre-diabetes, idiopathic T1D, LADA, EOD, YOAD, MODY, malnutrition-related diabetes, gestational diabetes, hyperglycemia, insulin resistance, hepatic insulin resistance, impaired glucose tolerance, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, adipocyte dysfunction, visceral fat deposition, sleep apnea, obesity, eating disorders, weight gain due to use of other agents, excessive carbohydrate craving, dyslipidemia, hyperinsulinemia, NAFLD, NASH, fibrosis, cirrhosis, hepatocellular carcinoma, cardiovascular disease, arteriosclerosis, coronary artery disease, peripheral vascular disease, hypertension, endothelial dysfunction, impaired vascular compliance, congestive heart failure, myocardial infarction, stroke, hemorrhagic stroke, ischemic stroke, traumatic brain injury, cerebral ischemia, stroke, Pulmonary hypertension, restenosis following angioplasty, intermittent claudication, postprandial lipemia, metabolic acidosis, ketosis, arthritis, osteoporosis, Parkinson's disease, left ventricular hypertrophy, peripheral arterial disease, macular degeneration, cataracts, glomerulosclerosis, chronic renal failure, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, transient ischemic attack, vascular restenosis, impaired glucose metabolism, conditions of impaired fasting plasma glucose, hyperuricemia, gout, erectile dysfunction, skin and connective tissue disorders, psoriasis, foot ulcers, ulcerative colitis, hyperpao B lipoproteinemia, Alzheimer's disease, schizophrenia, impaired cognitive function, inflammatory bowel disease, short bowel syndrome, Crohn's disease, colitis, irritable bowel syndrome, polycystic ovary syndrome, prophylaxis or treatment of atherosclerosis, transient ischemic attack, vascular restenosis, impaired glucose metabolism, impaired fasting plasma glucose, impaired glucose metabolism, stroke, And treatment of addiction.

Technical Field

Provided herein are benzimidazoles that are GLP-1R agonists as well as 6-carboxylic acids of 4-aza-, 5-aza-, and 7-aza-benzimidazoles, methods of making the compounds, and methods comprising administering the compounds to a mammal in need thereof.

Background

Diabetes is a significant public health problem due to its increasing prevalence and associated health risks. This disease is characterized by high levels of blood glucose resulting from defects in insulin production, insulin action, or both. Two major forms of diabetes, type 1 and type 2, have been identified. Type 1 diabetes (T1D) develops when the body's immune system destroys the pancreatic beta cells, the only cells in the body that make the hormone insulin (regulating blood glucose). In order to survive, people with type 1 diabetes must administer insulin by injection or pump. Type 2 diabetes (commonly referred to as T2DM) typically begins when insulin is resistant or when insulin is not sufficiently produced to maintain an acceptable blood glucose level.

Various pharmacological approaches are currently available for the treatment of hyperglycemia and, consequently, T2DM (Hampp, C. et al)Use of Antidiabetic Drugs in the U.S., 2003-2012, Diabetes Care2014, 37, 1367-1374). These can be grouped into six important classes, each acting through different major mechanisms: (A) insulinotropic agents including sulfonyl-ureas (e.g., glipizide (glipizide), glimepiride (glimepiride), glyburide (glyburide)), meglitinides (meglitinides) (e.g., nateglinide (nateglinide), repaglinide (repaglinide)), dipeptidyl peptidase IV (DPP-IV) inhibitors (e.g., sitagliptin (sitagliptin), vildagliptin (vildagliptin), alogliptin (alogliptin), dulagliptin (dutogliptin), linagliptin (linagliptin), saxagliptin (saxogliptin)), and glucagon-like peptide-1 receptor (GLP-1R) agonists (e.g., liraglutide (liraglutide), albiglutide (albuglutide), exenatide (exenatide), linagliptin (lixigliptin), glycopeptide (glycopeptide), degree of glycopeptide (dep-1R) (e.g., liraglutide (linagliptin), insulin-d, insulin-s, insulin-producing drugs, insulin-producing(dulaglutide), somaglutide), which enhances insulin secretion by acting on pancreatic beta cells. The efficacy and tolerability of sulfonyl-ureas and meglitinides is limited, causing weight gain and often inducing hypoglycemia. DPP-IV inhibitors have limited efficacy. GLP-1R agonists that have been marketed are peptides that are administered by subcutaneous injection. Liraglutide is additionally approved for the treatment of obesity. (B) Biguanides (e.g., metformin (metformin)) are believed to act primarily by reducing hepatic glucose production. Biguanides often cause gastrointestinal disturbances and lactic acidosis, further limiting their use. (C) Inhibitors of alpha-glucosidase (e.g., acarbose) reduce intestinal glucose absorption. These agents often cause gastrointestinal disturbances. (D) Thiazolidinediones [ e.g. pioglitazone (pioglitazone), rosiglitazone (rosiglitazone) ]Act on specific receptors in the liver, muscle and adipose tissue (peroxisome proliferator-activated receptor-gamma). They modulate lipid mechanisms, which in turn enhance the response of these tissues 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 aforementioned agents is used in more severe cases, and frequent use may also lead to weight gain and the attendant risk of hypoglycemia. (F) Sodium-glucose cotransporter 2 (SGLT2) inhibitors (e.g., dapagliflozin (dapagliflozin), empagliflozin (empagliflozin), canagliflozin (canagliflozin), eggliflozin (ertugliflozin)) inhibit reabsorption of glucose in the kidney and thereby reduce glucose levels in the blood. Such emerging drugs may be associated with ketoacidosis and urinary tract infections.

However, in addition to GLP-1R agonists and SGLT2 inhibitors, the efficacy of these drugs is limited and fails to address the most important problem, beta cell functional decline and associated obesity.

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

While T2DM is most commonly associated with hyperglycemia and insulin resistance, other diseases associated with T2DM also include: hepatic insulin resistance, impaired glucose tolerance, 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 conditions including steatosis, nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis and ultimately hepatocellular carcinoma. NAFLD and NASH are considered major fatty liver diseases because they account for the largest proportion of individuals with elevated liver lipids. The severity of NAFLD/NASH is based on the presence of lipids, inflammatory cell infiltration, hepatocyte ballooning, and the degree of fibrosis. Although not all individuals with steatosis progress to NASH, a significant proportion of individuals experience it.

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

In healthy individuals, GLP-1 regulates postprandial blood by stimulating glucose-dependent insulin secretion from the pancreas, leading to increased peripheral glucose uptakeThe sugar level plays an important role. GLP-1 also suppresses glucagon secretion, resulting in decreased hepatic glucose production. Moreover, GLP-1 delays gastric emptying and slows small bowel movement, thereby delaying food absorption. In humans with T2DM, a normal postprandial elevation of GLP-1 is absent or reduced (Vilsball T et al,Diabetes. 2001. 50; 609-613)。

Holst (Physiol. Rev.2007, 87, 1409) and Meier: (Nat. Rev. Endocrinol.2012, 8, 728) describe GLP-1 receptor agonists such as GLP-1, liraglutide and exenatide-4 (exendin-4) having three main pharmacological activities for improving glycemic control in T2DM patients by lowering fasting and postprandial glucose (FPG and PPG): (i) increase glucose-dependent insulin secretion (improved first and second phases), (ii) glucagon suppression activity in the case of hyperglycemia, (iii) delay the rate of gastric emptying, resulting in a meal-derived slowing of glucose absorption.

There remains a need for prevention and/or treatment of cardiovascular metabolism and related diseases that are easy to administer.

Brief Description of Drawings

FIG. 1 shows an observed powder X-ray diffraction pattern of the anhydrous (anhydrate) crystalline form (form 1) of the tris salt of compound example 7.

FIG. 2 shows an observed powder X-ray diffraction pattern of the anhydrous (anhydrate) crystalline form (form A) of the tris salt of compound example 10.

Detailed Description

The invention relates to compounds of formula I

Or a pharmaceutically acceptable salt thereof, wherein

R is F, Cl, or-CN;

p is 0 or 1;

ring A is phenyl or 6-membered heteroaryl;

m is 0, 1, 2, or 3;

each R1Independently selected from: halogen, -CN, -C1-3Alkyl, and-OC1-3Alkyl radical, wherein C1-3Alkyl and OC1-3The alkyl group of the alkyl group is substituted with 0 to 3F atoms;

R2is H or-C1-3Alkyl, wherein alkyl is substituted with 0 to 1 OH;

each R3Independently F, -OH, -CN, -C1-3Alkyl, -OC1-3Alkyl, and-C3-4Cycloalkyl, or 2R3Can be cyclized together to form-C3-4Spirocycloalkyl, wherein if valency allows, C1-3Alkyl and OC1-3The alkyl, cycloalkyl, or spirocycloalkyl groups of the alkyl group may be substituted with 0 to 3F atoms and with 0 to 1-OH;

q is 0, 1 or 2;

X-L is N-CH2、CHCH2Or cyclopropyl;

y is CH or N;

R4is-C1-3Alkyl, -C0-3alkylene-C3-6Cycloalkyl, -C0-3alkylene-R5or-C1-3alkylene-R6Wherein if allowed by valence, said alkyl group may be substituted with 0 to 3 substituents independently selected from 0 to 3F atoms and 0 to 1 substituent selected from-C0-1alkylene-CN, -C 0-1alkylene-ORO、-SO2-N(RN)2、-C(O)-N(RN)2、-N(C=O)(RN) and-N (R)N)2Substituted with the substituent(s); and is

Wherein said alkylene and cycloalkyl groups may be independently substituted, if valency permits, with 0 to 2 substituents independently selected from 0 to 2F atoms and 0 to 1 substituent selected from-C0-1alkylene-CN, -C0-1alkylene-OROand-N (R)N)2Substituted with the substituent(s);

R5is a 4-to 6-membered heterocycloalkyl group, wherein if valency permits, said heterocycloalkyl group may be substituted with 0 to 2 substituents independently selected from the group consisting of:

0 to 1 oxo (= O),

0 to 1-CN group, and the N-CN group,

0 to 2F atoms, and

0 to 2 are independently selected from-C1-3Alkyl and-OC1-3Alkyl substituents, wherein if valency permits, C1-3Alkyl and OC1-3The alkyl group of the alkyl group may be substituted with 0 to 3 substituents independently selected from the group consisting of:

from 0 to 3 atoms of F,

0 to 1-CN, and

0 to 1-ORO

R6Is a 5-to 6-membered heteroaryl, wherein the heteroaryl, if valency permits, may be substituted with 0 to 2 substituents independently selected from:

0 to 2 halogen atoms in the molecule of a halogen,

0 to 1 is selected from-OROand-N (R)N)2A substituent of (a), and

0 to 2-C1-3Alkyl, wherein the alkyl may be substituted with 0 to 3 substituents independently selected from the group consisting of:

0 to 3F atoms, and

0 to 1-OR O

Each ROIndependently is H, or-C1-3Alkyl radical, wherein C1-3Alkyl groups may be substituted with 0 to 3F atoms;

each RNIndependently is H, or-C1-3An alkyl group;

Z1、Z2and Z3Each is-CRZOr is or

Z1、Z2And Z3One of which is N and the other two are-CRZ(ii) a And is

Each RZIndependently H, F, Cl, or-CH3

Another embodiment relates to compounds of formula II

Or a pharmaceutically acceptable salt thereof, wherein

R is F;

p is 0 or 1;

ring a is phenyl or pyridyl;

m is 0, 1, or 2;

each R1Independently selected from: halogen, -CN, -C1-3Alkyl, and-OC1-3Alkyl radical, wherein C1-3Alkyl and OC1-3The alkyl group of the alkyl group is substituted with 0 to 3F atoms;

R2is H or CH3

X-L is N-CH2Or cyclopropyl;

y is CH or N;

Z3is-CRZOr N; and is

RZIs H, F, Cl, or-CH3

Another embodiment relates to compounds of formula III

Or a pharmaceutically acceptable salt thereof, wherein

Ring a is phenyl or pyridyl;

m is 0, 1, or 2;

each R1Independently selected from F, Cl, and-CN;

R2is H or CH3(ii) a And is

Y is CH or N.

Another embodiment relates to compounds of formula IV

Or a pharmaceutically acceptable salt thereof, wherein

m is 0, 1, or 2;

each R1Independently selected from F, Cl, and-CN;

R2is H or CH3(ii) a And is

Y is CH or N.

Another embodiment relates to compounds of formula V

Or a pharmaceutically acceptable salt thereof, wherein

m is 0 or 1;

R1is F, Cl, or-CN;

R2is H or CH3(ii) a And is

Y is CH or N.

Another embodiment relates to compounds of formula IV or formula V, wherein the phenyl or pyridyl group of ring A has one R1Substituted at a position para to the carbon to which the phenyl or pyridyl group is attached to the dioxolane to provide:

or a pharmaceutically acceptable salt thereof, wherein

Each R1Independently selected from F, Cl, and-CN;

R2is H or CH3(ii) a And is

Y is CH or N.

Another embodiment relates to compounds of the other embodiments herein, e.g., compounds of formula I or II, or pharmaceutically acceptable salts thereof, wherein X-L is N-CH2(ii) a And Y is CH or N. By the embodiments described herein, in such cases, X is N and L is CH2

Another embodiment relates to compounds of other embodiments herein, e.g., compounds of formula I or II, or pharmaceutically acceptable salts thereof, wherein X-L is CHCH2(ii) a And Y is N. By the embodiments described herein, in such cases, X is CH and L is CH2

Another embodiment relates to compounds of other embodiments herein, e.g., compounds of formula I or II, or pharmaceutically acceptable salts thereof, wherein X-L is CHCH 2(ii) a And Y is CH. By the embodiments described herein, in such cases, X is CH and L is CH2

Another embodiment relates to compounds of other embodiments herein, e.g., compounds of formula I or II, or pharmaceutically acceptable salts thereof, wherein X-L is cyclopropyl; and Y is N.

In embodiments wherein X-L is cyclopropyl, the compound of formula I or II provides:

another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R is4is-CH2CH2OCH3、C1-3alkylene-R5Or C1-3alkylene-R6

Another embodiment relates to compounds of formula II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R4As defined for the compounds of formula I.

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R is4is-C1-3Alkyl, wherein the alkyl may be substituted with 0 to 1 substituents selected from the group consisting of: -C0-1alkylene-OROand-N (R)N)2

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R is4Is- (CH)2)2OCH3Or is- (CH)2)2N(CH3)2

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R is 4is-CH2-R5Wherein R is5Is a 4-to 5-membered heterocycloalkyl group, wherein if valency permits, said heterocycloalkyl group may be substituted with 0 to 2 substituents independently selected from the group consisting of:

0 to 2F atoms, and

0 to 1 is selected from-OCH3and-CH2OCH3A substituent of (1).

Another embodiment relates to compounds of formula I, II, III, IV, or V wherein heterocycloalkyl is

Wherein, if valency permits, the heterocycloalkyl group can be substituted with 0 to 2 substituents independently selected from (e.g., substituted with hydrogen):

0 to 1 oxo (O =),

0 to 1-CN group, and the N-CN group,

0 to 2F atoms, and

0 to 2 are independently selected from-C1-3Alkyl and-OC1-3Alkyl substituents, wherein if valency permits, C1-3Alkyl and OC1-3The alkyl group of the alkyl group may be independently substituted with 0 to 3 substituents independently selected from the group consisting of:

from 0 to 3 atoms of F,

0 to 1-CN, and

0 to 1-ORO

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of formula I, II, III, IV, or V wherein heterocycloalkyl is

Wherein, if valency permits, the heterocycloalkyl group can be substituted with 0 to 2 substituents independently selected from (e.g., substituted with hydrogen):

0 to 1-CN group, and the N-CN group,

0 to 2F atoms, and

0 to 2 are independently selected from-C 1-3Alkyl and-OC1-3Alkyl substituents, wherein if valency permits, C1-3Alkyl and OC1-3The alkyl group of the alkyl group may be independently substituted with 0 to 3 substituents independently selected from the group consisting of:

from 0 to 3 atoms of F,

0 to 1-CN, and

0 to 1-ORO

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of formula I, II, III, IV, or V wherein heterocycloalkyl is

Wherein heterocycloalkyl, if valency permits, may be substituted with 0 to 1 substituent (e.g., hydrogen) selected from:

-CN,

f atom, and

0 to 1 are independently selected from-C1-3Alkyl and-OC1-3Alkyl substituents, wherein if valency permits, C1-3Alkyl and OC1-3The alkyl group of the alkyl group may be substituted with 0 to 3 substituents independently selected from the group consisting of:

from 0 to 3 atoms of F,

0 to 1-CN, and

0 to 1-ORO

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of formula I, II, III, IV, or V wherein heterocycloalkyl is

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein heterocycloalkyl is

Wherein, if valency permits, the heterocycloalkyl radical may be substituted by from 0 to 1 methyl group, wherein said methyl group may be substituted by from 0 to 3F atoms.

Another embodiment relates to compounds of formula I, II, III, IV, or V wherein heterocycloalkyl is

Wherein heterocycloalkyl is unsubstituted.

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein-CH2-R5And R4The attached nitrogen provides:

another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein R is4is-CH2-R6Wherein R is6Is a 5-membered heteroaryl, wherein the heteroaryl, if valency permits, may be substituted with 0 to 2 substituents independently selected from:

0 to 2 halogens, wherein the halogens are independently selected from F and Cl,

0 to 1-OCH3And are and

0 to 1-CH3、-CH2CH3、-CF3or-CH2CH2OCH3

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein heteroaryl is

Wherein the heteroaryl group may be substituted (e.g., by hydrogen) with 0 to 2 substituents independently selected from the group consisting of:

0 to 2 halogens, wherein the halogens are independently selected from F and Cl,

0 to 1 is selected from-OROand-N (R)N)2A substituent of, or

0 to 2-C1-3Alkyl radical, in which is alkylatedWhen allowed by valency, the alkyl group may be substituted with 0 to 3 substituents independently selected from:

0 to 3F atoms, and

0 to 1-ORO

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein heteroaryl is

Wherein the heteroaryl group may be substituted (e.g., by hydrogen) with 0 to 2 substituents independently selected from the group consisting of:

0 to 2 halogens, wherein the halogens are independently selected from F and Cl,

0 to 1 is selected from-OROand-N (R)N)2A substituent of, or

0 to 2-C1-3Alkyl, wherein alkyl, if valency permits, may be substituted with 0 to 3 substituents independently selected from:

0 to 3F atoms, and

0 to 1-ORO

Another embodiment relates to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein heteroaryl is

Wherein if allowed by valence, said heteroaryl may be substituted by 0 to 1-C1-2Alkyl substituents, wherein alkyl may be substituted, if valency permits, with 0 to 3 substituents independently selected from:

0 to 3F atoms, and

0 to 1-ORO(ii) a And is

Each ROIndependently is H, or-C1-3An alkyl group.

It is recognized that any substituent may be substituted for H on the carbon or nitrogen being substituted. Non-limiting examples of substituted heteroaryl groups are:

it is recognized that H is substituted, e.g., R 6s(R6Any of the heteroaryl groups of (a) or (b) to provide:

wherein R is6sis-C1-2Alkyl, wherein the alkyl may be substituted with 0 to 3 substituents independently selected from the group consisting of:

0 to 3F atoms, and

0 to 1-RO(ii) a And is

Each ROIndependently is H, or-C1-3An alkyl group;

or a pharmaceutically acceptable salt thereof.

Another embodiment is directed to compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein heteroaryl is:

another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein Z is1、Z2And Z3Each is CRZ

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein R isZIs H.

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein Z is1、Z2And Z3Each is CH.

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein R is 3is-CH3or-CF3(ii) a And q is 1.

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein each R is1Independently F, Cl, or-CN.

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein R is4is-CH2-R5

Another embodiment relates to compounds of the other embodiments herein, or pharmaceutically acceptable salts thereof, e.g., compounds of formula I, II, III, IV, or V, wherein R is4is-CH2-R6

Another embodiment relates to compounds of other embodiments herein, e.g., compounds of formula I, II, III, IV, or V, wherein the compound is a free acid.

Another embodiment relates to any embodiment of the compounds of formula I, II, III, IV, or V, or a pharmaceutically acceptable salt thereof, wherein ring A and R2Providing:

wherein:

r is F, Cl, or-CN;

p is 0 or 1;

m is 0, 1, or 2; and is

Each R1Independently selected from: halogen, -CN, -C1-3Alkyl, and-OC1-3Alkyl radical, wherein C1-3Alkyl and OC1-3The alkyl group of the alkyl group is substituted with 0 to 3F atoms.

Another embodiment relates to compounds of formula I, II, III, IV, or VOr a pharmaceutically acceptable salt thereof, wherein R2Is H.

Another embodiment relates to where R is2A compound of the invention that is H, or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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; or

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;

or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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; or

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;

or a pharmaceutically acceptable salt thereof.

Another embodiment relates to where R is2Is CH3Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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-1-yl)H-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) -Oxetazedin-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; or

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;

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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; or

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;

or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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; or

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;

Or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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; or

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;

or a pharmaceutically acceptable salt thereof.

Another embodiment relates to compounds of the invention, wherein the compounds are

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, wherein 2' S chirality is derived from C56;

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 wherein 2' S chirality is derived from P9;

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, wherein the chirality at 2 comes from 17;

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, wherein 2' S chirality is derived from 96; or

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, wherein the 2-chirality is derived from C82;

or a pharmaceutically acceptable salt thereof.

Another embodiment includes a compound that is 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, or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt.

Another embodiment includes compounds which are 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 as the free acid.

Another embodiment includes compounds which are

Or a pharmaceutically acceptable salt thereof.

Another embodiment includes the compound 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, or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt { a tris salt of this compound is also referred to as: 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 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 }.

In certain embodiments, the present invention provides crystalline forms of the anhydrous tris 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 certain further embodiments, a crystalline form of an anhydrous (anhydrate) tris 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" according to its unique solid state characteristics (e.g., substantially as depicted in fig. 1) with respect to, for example, powder X-ray diffraction (PXRD) as described herein. In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising at least two characteristic peaks, in terms of 2 Θ, selected from 3.7 ± 0.2; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and 16.9 ± 0.2. In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising at least three characteristic peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and 16.9 ± 0.2. In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising at least four characteristic peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and 16.9 ± 0.2. In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising at least five characteristic peaks, in terms of 2 Θ, selected from 3.7 ± 0.2 ℃; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and 16.9 ± 0.2.

In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising characteristic peaks, in terms of 2 Θ, at 3.7 ± 0.2 ° and 7.3 ± 0.2 ℃.

In certain embodiments, form I exhibits a powder X-ray diffraction pattern comprising peaks, in terms of 2 Θ, at 3.7 ± 0.2 °, 7.3 ± 0.2 ° and 14.7 ± 0.2 ℃. In certain further embodiments, form I exhibits a powder X-ray diffraction pattern further comprising at least one peak selected, in terms of 2 Θ, from 8.5 ± 0.2; 10.1 ± 0.2; and 16.9 ± 0.2.

In certain embodiments, form I appears to comprise a peak level at 3.7 ± 0.2 ° in terms of 2 Θ; 7.3 ± 0.2; 14.7 ± 0.2; and a powder X-ray diffraction pattern of peaks at 16.9 ± 0.2.

In certain embodiments, form I appears to comprise a peak level at 3.7 ± 0.2 ° in terms of 2 Θ; 7.3 ± 0.2; 8.5 ± 0.2; 10.1 ± 0.2; 14.7 ± 0.2; and a powder X-ray diffraction pattern of peaks at 16.9 ± 0.2.

In certain embodiments, form I exhibits a powder X-ray diffraction pattern substantially as shown in figure 1. A series of diffraction peaks expressed in terms of 2 θ degrees and relative intensity (relative intensity ≧ 3.0%) are provided in Table X1 above.

As is well known in the art of powder diffraction, the relative intensities of the peaks (reflections) may vary with the sample preparation technique, the sample installation procedure, and the particular instrument used. In addition, instrument variations and other factors can affect the 2-theta values. Thus, XRPD peak assignments may vary by ± about 0.2.

Another embodiment includes compounds which are 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 as the free acid.

Another embodiment includes compounds which are

Or a pharmaceutically acceptable salt thereof.

Another embodiment includes compounds which are the following as free acids:

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; or

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.

Another embodiment includes compounds: which is that

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; or

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;

or a pharmaceutically acceptable salt thereof, wherein the salt is a tris salt.

Another embodiment includes compounds that are 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 certain further embodiments, the present invention provides a compound which is 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 tris salt thereof [ i.e., 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-amine salt ]. The chiral centers on the left part of the compound structure are labeled "abs" to indicate that the chiral centers have only one stereoconfiguration (i.e., not the racemate relative to the chiral centers).

In certain embodiments, the present invention provides crystalline forms of the anhydrous tris 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 certain further embodiments, a crystalline form of the anhydrous (anhydrate) tris 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 as "form a" as characterized by its unique solid state characteristics with respect to, for example, powder X-ray diffraction (PXRD) as described herein (e.g., substantially as depicted in fig. 2). In certain embodiments, form a exhibits a powder X-ray diffraction pattern comprising at least two characteristic peaks, in terms of 2 Θ, selected from 7.7 ± 0.2 ℃; 15.2 ± 0.2; (vii) 15.7 + 0.2 and 17.6 + 0.2. In certain embodiments, form a exhibits a powder X-ray diffraction pattern comprising at least three characteristic peaks, in terms of 2 Θ, selected from 7.7 ± 0.2 ℃; 15.2 ± 0.2; (vii) 15.7 + 0.2 and 17.6 + 0.2. In certain embodiments, form a exhibits a powder X-ray diffraction pattern comprising a characteristic peak selected, in terms of 2 Θ, from 7.7 ± 0.2 ℃; 15.2 ± 0.2; (vii) 15.7 + 0.2 and 17.6 + 0.2.

In certain embodiments, form I appears to comprise a peak level at 7.7 ± 0.2 ° in terms of 2 Θ; and a powder X-ray diffraction pattern of characteristic peaks at 17.6 ± 0.2.

In certain embodiments, form a appears to comprise a peak level at 7.7 ± 0.2 ° in terms of 2 Θ; 15.2 ± 0.2; and a powder X-ray diffraction pattern of peaks at 17.6 ± 0.2.

In certain embodiments, form I appears to comprise a peak level at 7.7 ± 0.2 ° in terms of 2 Θ; 15.2 ± 0.2; and a powder X-ray diffraction pattern of peaks at 15.7 ± 0.2.

In certain embodiments, form I appears to comprise a peak level at 7.7 ± 0.2 ° in terms of 2 Θ; 15.2 ± 0.2; an X-ray powder diffraction pattern for peaks at 15.7 + -0.2 and 17.6 + -0.2.

In certain embodiments, form a exhibits a powder X-ray diffraction pattern substantially as shown in figure 2. A series of diffraction peaks expressed in terms of 2 θ degrees and relative intensity (relative intensity ≧ 3.0%) are provided in Table X2 above.

As is well known in the art of powder diffraction, the relative intensities of the peaks (reflections) may vary with the sample preparation technique, the sample installation procedure, and the particular instrument used. In addition, instrument variations and other factors can affect the 2-theta values. Thus, XRPD peak assignments may vary by ± about 0.2.

In a further embodiment, the present invention provides a pharmaceutical composition comprising a compound of formula I, II, III, IV, or V as defined in any of the embodiments described herein, or a pharmaceutically acceptable salt thereof, in admixture with at least one pharmaceutically acceptable excipient. This includes pharmaceutical compositions comprising: a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof, in admixture with at least one pharmaceutically acceptable excipient and one or more other therapeutic agents discussed herein.

The present invention also includes the following embodiments:

a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for use as a medicament;

a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for use in the prevention and/or treatment of cardiovascular metabolism and related diseases (including T2DM, pre-diabetes, NASH, and cardiovascular disease) as discussed herein;

a method of treating a disease for which a GLP-1R agonist is indicated in a subject in need of such prevention and/or treatment, comprising administering to the subject a therapeutically effective amount of a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof;

Use of a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease or condition for which a GLP-1R agonist is indicated;

a compound of formula I, II, III, IV, or V as defined in any of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or condition for which a GLP-1R agonist is indicated; or

A pharmaceutical composition for use in treating a disease or condition for which a GLP-1R agonist is indicated, comprising a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof.

Each example, or a pharmaceutically acceptable salt thereof, may be claimed in groups individually, or in any combination with any number of each of the embodiments described herein.

The present invention also relates to pharmaceutical compositions comprising a compound of formula I, II, III, IV, or V as defined in any one of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of cardiovascular metabolism and related diseases (including T2DM, pre-diabetes, NASH, and cardiovascular disease) as discussed herein.

Another embodiment of the present invention relates to compounds of formula I, II, III, IV, or V as defined in any of the embodiments described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of cardiovascular metabolism and related diseases including: diabetes (T1D and/or T2DM, including pre-diabetes), idiopathic T1D (type 1 b), latent autoimmune diabetes in adults (LADA), early onset T2DM (EOD), juvenile-onset atypical diabetes (YOAD), adult-onset diabetes in young adults (MODY), malnutrition-related diabetes, gestational diabetes, hyperglycemia, insulin resistance, hepatic insulin resistance, impaired glucose tolerance, diabetic neuropathy, diabetic nephropathy, nephropathy (e.g., acute kidney disorder, tubular dysfunction, proinflammatory changes in the proximal tubule), diabetic retinopathy, adipocyte dysfunction, visceral fat deposition, sleep apnea, obesity (including hypothalamic obesity and monogenic obesity) and related complications (e.g., osteoarthritis and urinary incontinence), eating disorders (including eating disorders, urinary incontinence), Bulimia nervosa, and the syndrome obesity, e.g., Prader-Willi and Bardet-Biedl syndrome), weight gain due to the use of other agents (e.g., due to the use of steroids and antipsychotics), excessive carbohydrate craving, dyslipidemia (including hyperlipidemia, hypertriglyceridemia, increased total cholesterol, high LDL cholesterol, and low HDL cholesterol), hyperinsulinemia, NAFLD (including related diseases, e.g., steatosis, NASH, fibrosis, cirrhosis, and hepatocellular carcinoma), cardiovascular disease, atherosclerosis (including coronary artery disease), peripheral vascular disease, hypertension, endothelial dysfunction, impaired vascular compliance, congestive heart failure, myocardial infarction (e.g., necrosis and apoptosis), stroke, hemorrhagic stroke, ischemic stroke, traumatic brain injury, peripheral vascular disease, vascular dementia, and other, Pulmonary hypertension, restenosis following angioplasty, intermittent claudication, postprandial lipemia, metabolic acidosis, ketosis, arthritis, osteoporosis, Parkinson's disease, left ventricular hypertrophy, peripheral arterial disease, macular degeneration, cataracts, glomerulosclerosis, chronic renal failure, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, transient ischemic attacks, vascular restenosis, impaired glucose metabolism, conditions of impaired fasting plasma glucose, hyperuricemia, gout, erectile dysfunction, skin and connective tissue disorders, psoriasis, foot ulcers, ulcerative colitis, hyperpao B lipoproteinemia, schizophrenia, impaired cognitive function, inflammatory bowel disease, short bowel syndrome, Crohn's disease, colitis, irritable bowel syndrome, Prevention or treatment of polycystic ovary syndrome, and treatment of addiction (e.g., alcohol and/or drug abuse).

Abbreviations used herein are as follows:

the term "alkyl" as used herein means a group of the formula-CnH(2n+1)A straight or branched monovalent hydrocarbon group of (1). Non-limiting examples include methyl, ethyl, propyl, butyl, 2-methyl-propyl, 1-dimethylethyl, pentyl, and hexyl.

The term "alkylene" as used herein means a compound of formula-CnH2nA linear or branched divalent hydrocarbon group of (a). Non-limiting examples include ethylene and propylene.

The term "cycloalkyl" as used herein means a compound of formula-C containing at least three carbon atomsnH(2n-1)A cyclic, monovalent hydrocarbon group of (a). Non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.

The term "halogen" as used herein refers to fluorine, chlorine, bromine, or iodine.

The term "heterocycloalkyl" as used herein refers to a compound in which one or more of the cyclomethylene groups (-CH) are present2-) a cycloalkyl group which has been replaced by a group selected from-O-, -S-, or a nitrogen, where the nitrogen may provide the point of attachment or may be substituted as provided within the various embodiments. Where a nitrogen provides the point of attachment, the structural diagram of the heterocycloalkyl group will have one hydrogen on the nitrogen. In general, where valency permits, the heterocycloalkyl group can be substituted with from 0 to 2 substituents independently selected from: oxo, -CN, halogen, alkyl and-Oalkyl, and the alkyl may be further substituted. It may be noted that when there are 0 substitutions, the heterocycloalkyl group is unsubstituted.

The term "heteroaryl" as used herein refers to monocyclic aromatic hydrocarbons containing 5 to 6 carbon atoms in which at least one ring carbon atom is replaced by a heteroatom selected from oxygen, nitrogen and sulfur. Such heteroaryl groups may be attached through a ring carbon atom or, where valency permits, through a ring nitrogen atomAnd (4) sub-connection. In general, heteroaryl groups may be substituted, if valency permits, with 0 to 2 substituents independently selected from: halogen, OH, alkyl, O-alkyl, and amino (e.g., NH)2NH alkyl, N (alkyl)2) And the alkyl group may be further substituted. It may be noted that when there are 0 substitutions, the heteroaryl group is unsubstituted.

Room temperature: RT (15 to 25 ℃ C.).

Methanol: MeOH.

Ethanol: EtOH.

Isopropyl alcohol: iPrOH.

Ethyl acetate: EtOAc.

Tetrahydrofuran: THF.

Toluene: PhCH3

Cesium carbonate: cs2CO3

Lithium bis (trimethylsilyl) amide: LiHMDS.

Sodium tert-butoxide: NaOtBu.

Potassium tert-butoxide: KOtBu.

Lithium diisopropylamide: and (4) LDA.

Triethylamine: et (Et)3N。

N, N-diisopropylethylamine: DIPEA.

Potassium carbonate: k2CO3

Dimethylformamide: DMF.

Dimethyl acetamide: DMAc.

Dimethyl sulfoxide: DMSO.

N-methyl-2-pyrrolidone: NMP.

Sodium hydride: NaH.

Trifluoroacetic acid: TFA.

Trifluoroacetic anhydride: TFAA.

Acetic anhydride: ac of2O。

Dichloromethane: DCM.

1, 2-dichloroethane: and (3) DCE.

Hydrochloric acid: HCl.

1, 8-diazabicyclo [5.4.0] undec-7-ene: and DBU.

Borane-dimethyl sulfide complex: BH3-DMS。

Borane-tetrahydrofuran complex: BH3-THF。

Lithium aluminum hydride: LAH.

Acetic acid: AcOH.

Acetonitrile: MeCN.

P-toluenesulfonic acid: pTSA.

Dibenzylidene acetone: DBA.

2,2 '-bis (diphenylphosphino) -1, 1' -binaphthyl: BINAP.

1, 1' -ferrocenediyl-bis (diphenylphosphine): dppf.

1, 3-bis (diphenylphosphino) propane: and (3) DPPP.

3-chloroperoxybenzoic acid: m-CPBA.

Tert-butyl methyl ether: MTBE.

Methane sulfonyl group: ms.

N-methylpyrrolidone: NMP.

Thin-layer chromatography: and (5) TLC.

Supercritical fluid chromatography: SFC.

4- (dimethylamino) pyridine: DMAP.

T-butoxycarbonyl group: boc.

1- [ bis (dimethylamino) methylene ] -1H-1,2, 3-triazolo [4,5-b ] pyridinium 3-oxide hexafluorophosphate: HATU.

Petroleum ether: and (3) PE.

2- (1H-benzotriazol-1-yl) -1,1,3, 3-tetramethyluronium hexafluorophosphate: an HBTU.

2-amino-2- (hydroxymethyl) propane-1, 3-diol: and (3) tris.

Tris (dibenzylideneacetone) dipalladium: pd 2(dba)3

1The H Nuclear Magnetic Resonance (NMR) spectrum was consistent with the proposed structure in all cases. Characteristic chemical shift (. delta.) relative to deuterated solvent (CHCl at 7.27 ppm)3(ii) a CD at 3.31 ppm2HOD; at 1.94 ppm MeCN; parts per million of residual proton signal in 2.50 ppm DMSO), and recorded using conventional abbreviations for the major peaks designated: for example, s, singlet; d, double peak; t: a triplet; q, quartet; m, multiplet; br, broad peak. Symbol ^ represents1The H NMR peak area is presumed, becauseThe peak is partially obscured by the water peak. Symbol ^ represents1The H NMR peak area is assumed because the peak is partially obscured by the solvent peak.

Wavy lines, as used herein ""denotes the point of attachment of a substituent to another group.

The compounds and intermediates described below are named using the naming convention provided by ACD/ChemSetch 2012, File Version C10H41, Build 69045 (Advanced Chemistry Development, Inc., Toronto, Ontario, Canada). The naming convention provided by ACD/ChemSketch 2012 is well known to those skilled in the art and the naming convention provided by ACD/ChemSketch 2012 is believed to generally conform to IUPAC (International Union for Pure and Applied chemistry) recommendations and CAS indexing rules of organic chemical nomenclature. It may be noted that chemical names may have only parentheses or may have parentheses and brackets. Depending on the naming convention, the descriptors for stereochemistry may also be placed at different positions within the name itself. One skilled in the art can recognize these format variations and understand that they provide the same chemical structure.

Pharmaceutically acceptable salts of compounds of formula I, II, III, IV, or V include acid addition and base salts.

Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate, camphorsulfonate, citrate, cyclamate, edisylate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, salicylate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulfate, naphthalenedicarboxylate, 2-naphthalenesulfonate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, citrate, dihydrogenphosphate, citrate, salicylate, fumarate, gluconate, glucuronate, gluconate, and mixtures thereof, Pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate, 1, 5-naphthalenedisulfonate, and xinofoate salts.

Suitable base salts are formed from bases which form non-toxic salts. Examples include aluminum, arginine, benzathine, calcium, choline, diethylamine, bis (2-hydroxyethyl) amine (diethanolamine), glycine, lysine, magnesium, meglumine, 2-aminoethanol (ethanolamine), potassium, sodium, 2-amino-2- (hydroxymethyl) propane-1, 3-diol (tris or tromethamine) and zinc.

Hemisalts of acids and bases, such as hemisulfate and hemicalcium salts, may also be formed. For a review of suitable Salts, see Stahl and Wermuth, Handbook of Pharmaceutical Salts: Properties, Selection, and Use (Wiley-VCH, 2002).

Pharmaceutically acceptable salts of the compounds of formula I can be prepared by one or more of the following three methods:

(i) reacting a compound of formula I with a desired acid or base;

(ii) removing acid or base labile protecting groups from a suitable precursor of a compound of formula I, or opening a suitable cyclic precursor (e.g., a lactone or lactam) using a desired acid or base; or

(iii) One salt of the compound of formula I is converted to another salt by reaction with an appropriate acid or base or by means of a suitable ion exchange column.

All three reactions are usually carried out in solution. The resulting salt may precipitate out and be collected by filtration, or may be recovered by evaporation of the solvent. The degree of ionization of the resulting salt can vary from fully ionized to almost unionized.

The compounds of formula I and pharmaceutically acceptable salts thereof may exist in unsolvated as well as solvated forms. The term "solvate" is used herein to describe a molecular complex comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable solvent molecules (e.g., ethanol). When the solvent is water, the term "hydrate" is used.

The currently accepted classification system for organic hydrates is that of defining separation sites, channels or metal-ion coordinated hydrates-see k.r. MorrisPolymorphism in Pharmaceutical Solids(ed. H.G. Brittain, Marcel Dekker, 1995). An isolated site hydrate is a hydrate in which water molecules are separated from each other by insertion of organic molecules, without direct contact. In channeled hydrates, water molecules are located in lattice channels alongside other water molecules. In the metal-ion coordination hydrate, water molecules are bonded to the metal ions.

When solvent or water is intimately bound, the complex may have a well-defined stoichiometry independent of humidity. However, when solvents or water are weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content may depend on humidity and drying conditions. In such cases, non-stoichiometry would be the standard.

Also included within the scope of the present invention are multi-component complexes (in addition to salts and solvates) in which the drug and at least one other component are present in stoichiometric or non-stoichiometric amounts. Complexes of this type include clathrates (drug-host includes complexes) and co-crystals. The latter is generally defined as a crystalline complex of neutral molecular components held together via non-covalent interactions, but may also be a complex of a neutral molecule and a salt. The co-crystals may be prepared by melt crystallization, by recrystallization from a solvent, or by physically milling the components together. See Chem Commun by O.Almarsson and M.J. Zaworkko,17, 1889-1896 (2004). For a general review of multicomponent complexes, see J Pharm Sci by Haleblian,64(8) 1269-.

The compounds of the present invention may exist in a continuous solid state ranging from completely amorphous to completely crystalline. The term "amorphous" refers to a state in which a material lacks long range order (long range order) at the molecular level and can exhibit the physical properties of a solid or a liquid depending on temperature. Such materials typically do not produce a unique X-ray diffraction pattern and, although exhibiting solid properties, are more formally described as liquids. Upon heating, a change in solid to liquid properties occurs, characterized by a change in state, usually a second order change ("glass transition"). The term "crystalline" refers to a solid phase in which the material has a regularly ordered internal structure at the molecular level and produces a unique X-ray diffraction pattern with well-defined peaks. Such materials will also exhibit the properties of a liquid when heated sufficiently, but the change from solid to liquid is characterized by a phase change, typically a first order phase change ("melting point").

The compounds of formula I may also exist in a mesomorphic state (mesophase or liquid crystal) when subjected to suitable conditions. The mesomorphic state is an intermediate state between the true crystalline state and the true liquid state (melt or solution). Mesogenicity due to temperature changes is described as "thermally denatured" and mesogenicity due to the addition of a second component (such as water or another solvent) is described as "readily soluble". Compounds having the potential to form lyotropic mesophases are described as "amphoteric" and consist of a compound having an ionic polar head group (e.g., -COO-Na+、-COO-K+or-SO3 -Na+) Or a non-ionic polar head group (e.g. -N)-N+(CH3)3) The molecular composition of (a). For more information see n.h. hartshorn and a. StuartCrystals and the Polarizing Microscope4 th edition (Edward Arnold, 1970).

The compounds of formula I may exhibit polymorphism and/or one or more isomerism phenomena (e.g. optical isomerism, geometric isomerism or tautomerism). The compounds of formula I may also be isotopically labelled. Such variations imply that the compounds of formula I, which are defined by reference to their structural features, are within the scope of the invention.

The compounds of formula I containing one or more asymmetric carbon atoms may exist as two or more stereoisomers. When the compounds of formula I contain an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Tautomerism ("tautomerism") may exist when structural isomers can interconvert via a low energy barrier. This may take the form of proton tautomerism in compounds of formula I containing, for example, imine, keto, or oxime groups, or so-called valence tautomerism in compounds containing aromatic moieties. As a result, a single compound may exhibit more than one type of isomerization.

Pharmaceutically acceptable salts of the compounds of formula I may also contain optically active (e.g. d-lactate or 1-lysine) or racemic (e.g. dl-tartrate or dl-arginine) counterions.

The cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, such as chromatography and fractional crystallization.

Conventional techniques for preparing/separating the individual enantiomers include chiral synthesis from suitable optically pure precursors or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral High Pressure Liquid Chromatography (HPLC). Alternatively, racemic precursors containing chiral esters can be separated by enzymatic resolution (see, e.g., Int J Mol Sci 29682 (2015) to a.c.l.m. Carvaho et al). In the case where the compound of formula I contains an acidic or basic moiety, salts can be formed using optically pure bases or acids (e.g., 1-phenylethylamine or tartaric acid). The resulting diastereomeric mixtures can be separated by fractional crystallization and one or both diastereomeric salts can be converted into the corresponding pure enantiomers by means well known to the skilled person. Alternatively, the racemate (or a racemic precursor) may be covalently reacted with a suitable optically active compound (e.g., an alcohol, amine, or benzyl chloride). The resulting diastereomeric mixtures can be separated by chromatography and/or fractional crystallization by means well known to the skilled person to give the separated diastereomers in the form of a single enantiomer with 2 or more chiral centers. Chiral compounds of formula I (and chiral precursors thereof) in enantiomerically enriched form can be obtained using chromatography (typically HPLC) on an asymmetric resin with a mobile phase consisting of a hydrocarbon (typically heptane or hexane) containing 0 to 50% by volume (typically 2% to 20% by volume) of isopropanol and 0 to 5% by volume of an alkylamine (typically 0.1% diethylamine). Concentrating the eluate to obtain an enriched mixture. Chiral chromatography using sub-and supercritical fluids may be employed. Can be used in some embodiments of the invention Methods of chiral chromatography are known in the art (see, e.g., Smith, Roger M., Lough Borough University, Lough Borough, UK; Chromographic Science Series (1998), 75 (SFC with Packed Columns), pp. 223-. In certain related examples herein, the column is obtained from Chiral Technologies, Inc, West Chester, Pennsylvania, USA, Daicel®Subsidiary of Chemical Industries, ltd, Tokyo, Japan.

When any racemate crystallizes, two different types of crystals are possible. The first type is the racemic compound mentioned above (true racemate), which results in a homogeneous crystalline form containing equimolar amounts of the two enantiomers. The second type is a racemic mixture or an agglomerate, in which equimolar amounts of the two crystal forms, each comprising a single enantiomer, are produced. Although two crystalline forms present in a racemic mixture may have the same physical properties, they may have different physical properties compared to the true racemate. The racemic mixture can be separated by conventional techniques known to those skilled in the art. See, e.g., E.L. Eliel and S.H. Wilen Stereochemistry of Organic Compounds (Wiley, 1994)。

It must be emphasized that, in the present context, the compounds of formula I are drawn in a single tautomeric form, all possible tautomeric forms being included within the scope of the invention.

The present invention includes all pharmaceutically acceptable isotopically-labeled compounds of formula I wherein 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 which predominates in nature.

Examples of isotopes suitable for inclusion in compounds of the invention include: isotopes of hydrogen, for example,2h and3h; isotopes of carbon, for example,11C、13c and14c; isotopes of chlorine, for example,36cl; isotopes of fluorine, for example,18f; isotopes of iodine, for example,123i and125i; isotopes of nitrogenThe amount of the solvent to be used is, for example,13n and15n; isotopes of oxygen, for example,15O,17o and18o; isotopes of phosphorus, for example,32p; and isotopes of sulfur, such as, for example,35S。

certain isotopically-labeled compounds of formula I, for example, those incorporating a radioisotope, are useful in drug and/or substrate tissue distribution studies. In view of the ease of incorporation and the ready means of detection, the radioactive isotope tritium, i.e.,3h, and carbon-14, i.e., 14C is particularly useful for this purpose.

Heavier isotopes, such as, for example, deuterium, i.e.,2h replacement, due to its higher metabolic stability, may offer certain therapeutic advantages, such as increased in vivo half-life or reduced dosage requirements.

The use of positron emitting isotopes, for example,11C、18F、15o and13n substitution can be used in Positron Emission Tomography (PET) studies for testing substrate receptor occupancy.

Isotopically-labeled compounds of formula I 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 an appropriate isotopically-labeled reagent in place of the unlabeled reagent previously employed.

Pharmaceutically acceptable solvates according to the invention include solvates in which the crystallization solvent may be isotopically substituted (e.g., D)2O、d6-acetone, d6-DMSO)。

One way of carrying out the invention is to administer a compound of formula I in prodrug form. Thus, derivatives of certain compounds of formula I, which are themselves pharmacologically less or not active, may be converted into compounds of formula I having the desired activity when administered in or on the body, for example by hydrolytic cleavage, in particular by esterases or peptidases. Such derivatives are referred to as "prodrugs". Further information on the use of prodrugs can be found in: 'Pro-drugs as Novel Delivery Systems', Vol. 14, ACS Symposium Series (T. Higuchi and W. Stella) and 'Bioreversible Carriers in Drug Delivery', Pergamon Press, 1987 (Ed. E.B. Roche, American Pharmaceutical Association). Reference may also be made to Nature Reviews/Drug Discovery, 2008, 7, 355 and Current Opinion in Drug Discovery and Development, 2007, 10, 550.

Prodrugs according to the invention may, for example, be generated by replacing appropriate functional groups present in the compounds of formula I with certain moieties known to those skilled in the art as "pro-moieties", e.g. 'Design of precursors' by and C.G. Wermuth, 'Design precursors and Bioprecursors', Practice of Medicinal Chemistry, (fourth edition), Chapter 28, 657-696 (Elsevier, 2015), as described in H. Bundgaard (Elsevier, 1985) and Y.M. Choi-Sledeski.

Thus, prodrugs according to the present invention are (a) ester or amide derivatives of carboxylic acids within compounds of formula I; (b) ester, carbonate, carbamate, phosphate, or ether derivatives of hydroxyl groups within the compounds of formula I; (c) amide, imine, carbamate, or amine derivatives of amino groups within compounds of formula I; (d) oxime or imine derivatives of the carbonyl group within the compounds of formula I; or (e) a methyl, primary alcohol or aldehyde group that can be metabolically oxidized to a carboxylic acid within the compound of formula I.

Some specific examples of prodrugs according to the invention include:

(i) in the case where the compound of formula I contains a carboxylic acid function (-COOH), an ester thereof, for example, wherein the hydrogen of the carboxylic acid function of the compound of formula I is replaced by C 1-C8Alkyl (e.g. ethyl) or (C)1-C8Alkyl) C (= O) OCH2-a step of (a) receiving (e.g.,tBuC(=O)OCH2-) substituted compounds;

(ii) in the case where the compound of formula I contains an alcohol function (-OH), an ester thereof, for example, wherein the alcohol function of the compound of formula I is hydrogenated by-CO (C)1-C8Alkyl) (e.g., methylcarbonyl) or an alcohol esterified with an amino group;

(iii) in case the compound of formula I contains an alcoholic function (-OH), an ether thereof, e.g. wherein the compound of formula IHydrogen of alcohol function of the compound is represented by (C)1-C8Alkyl) C (= O) OCH2-or-CH2OP(=O)(OH)2A substituted compound;

(iv) in case the compound of formula I contains an alcohol function (-OH), the phosphate thereof, e.g. wherein the alcohol function hydrogen of the compound of formula I is-P (= O) (OH)2or-P (= O) (ONa)2or-P (= O) (O)-)2Ca2+A substituted compound;

(v) containing primary or secondary amino functions (-NH) in the compounds of formula I2or-NHR, where R.noteq.H), amides thereof, for example, where one or two (as the case may be) hydrogens of the amino function of the compound of formula I are replaced by (C)1-C10) Alkanoyl, -COCH2NH2Compounds in which the amino group is substituted or derivatized with an amino acid;

(vi) containing primary or secondary amino functions (-NH) in the compounds of formula I2or-NHR, where R.noteq.H), an amine thereof, for example, where one or two (as the case may be) hydrogens of the amino function of the compound of formula I are replaced by-CH 2OP(=O)(OH)2A substituted compound;

(vii) the carboxylic acid groups in the compounds of the formula I are substituted by methyl groups, -CH2OH groups or aldehyde groups.

Certain compounds of formula I may themselves act as prodrugs of other compounds of formula I. It is also possible for two compounds of the formula I to be bound together in the form of a prodrug. In certain instances, prodrugs of compounds of formula I may be generated by internally linking two functional groups within the compounds of formula I (e.g., by forming a lactone).

Reference to a compound of formula I includes the compound itself and prodrugs thereof. The invention includes such compounds of formula I as well as pharmaceutically acceptable salts of such compounds and pharmaceutically acceptable solvates of such compounds and salts.

Administration and dosing

In general, the compounds of the invention are administered in an amount effective to treat the conditions described herein. The compounds of the invention may be administered as the compound itself, or alternatively, in the form of a pharmaceutically acceptable salt. For the purposes of administration and administration, the compounds themselves or pharmaceutically acceptable salts thereof will be referred to simply as the compounds of the invention.

The compounds of the invention may be administered by any suitable route, in the form of pharmaceutical compositions adapted to such route, and in dosages effective for the intended treatment. The compounds of the invention may be administered orally, rectally, vaginally, parenterally, or topically.

The compounds of the invention may be administered orally. Oral administration may involve swallowing, whereby the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed whereby the compound will enter the blood stream directly from the mouth.

In another embodiment, the compounds of the invention may also be administered directly to the bloodstream, muscle, or viscera. Suitable means for parenteral administration include: intravenous, intra-arterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, and subcutaneous. Suitable devices for parenteral administration include: needle (including microneedle) injectors, needleless injectors, and infusion techniques.

In another embodiment, the compounds of the present invention may also be administered topically, i.e., transdermally or transdermally, to the skin or mucosa. In another embodiment, the compounds of the invention may also be administered intranasally or by inhalation. In another embodiment, the compounds of the invention may be administered rectally or vaginally. In another embodiment, the compounds of the invention may also be administered directly to the eye or ear.

The dosage regimen for the compounds of the present invention and/or compositions containing the compounds is based on a variety of factors including: type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; as well as the activity of the particular compound employed. Thus, the dosage regimen may vary widely. In one embodiment, for the treatment of a given condition as discussed herein, the total daily dose of a compound of the invention is typically from about 0.001 to about 100 mg/kg (i.e., mg of a compound of the invention per kg body weight). In another embodiment, the total daily dose of a compound of the invention is from about 0.01 to about 30 mg/kg, and in another embodiment, from about 0.03 to about 10 mg/kg, and in yet another embodiment, from about 0.1 to about 3 mg/kg. It is not uncommon to repeat the administration of a compound of the invention multiple times (usually no more than four times) in a day. If desired, multiple daily doses may be used to increase the total daily dose.

For oral administration, the compositions may be provided in the form of tablets containing 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 30.0, 50.0, 75.0, 100, 125, 150, 175, 200, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient. The medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1 mg to about 100 mg of the active ingredient. When administered intravenously, the dosage may range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.

Suitable subjects according to the invention include mammalian subjects. In one embodiment, a human is a suitable subject. The human subject may be of any gender and any stage of development.

Pharmaceutical composition

In another embodiment, the invention comprises a pharmaceutical composition. Such pharmaceutical compositions comprise a compound of the present invention together with a pharmaceutically acceptable carrier. Other pharmacologically active substances may also be present. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of the following: water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, and combinations thereof, and isotonic agents, for example, sugars, sodium chloride, or polyols (e.g., mannitol or sorbitol) may be included in the compositions. Pharmaceutically acceptable substances, for example, wetting agents or minor amounts of auxiliary substances, such as wetting or emulsifying agents, preservatives or buffers which enhance the shelf-life or effectiveness of the antibody or antibody portion.

The compositions of the present invention may take various forms. These forms include, for example, liquid, semi-solid, and solid dosage forms, such as, for example, liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes, and suppositories. The dosage form depends on the intended mode of administration and therapeutic application.

Typical compositions are in the form of injectable or infusible solutions, e.g., compositions similar to those containing general antibodies for passive immunization of humans. One mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In another embodiment, the antibody is administered by intravenous infusion or injection. In yet another embodiment, the antibody is administered by intramuscular or subcutaneous injection.

Oral administration of solid dosage forms can be presented, for example, as discrete units, e.g., hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention. In another embodiment, oral administration may be in the form of a powder or granules. In another embodiment, the oral dosage form is sublingual, e.g., a lozenge. In such solid dosage forms, the compound of formula I is typically combined with one or more adjuvants. Such capsules or tablets may contain a controlled release formulation. In the case of capsules, tablets, and pills, the dosage forms may also contain buffering agents or may be prepared using enteric coatings.

In another embodiment, oral administration may be in the form of a liquid dose. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions may also contain adjuvants, such as wetting agents, emulsifying agents, suspending agents, flavoring agents (e.g., sweetening agents), and/or perfuming agents.

In another embodiment, the invention encompasses parenteral dosage forms. "parenteral administration" includes, for example, subcutaneous injection, intravenous injection, intraperitoneal injection, intramuscular injection, intrasternal injection, and infusion. Injectable formulations (i.e., sterile injectable aqueous or oleaginous suspensions) can be formulated according to the known art using suitable dispersing, wetting, and/or suspending agents.

In another embodiment, the present invention comprises a topical dosage form. "topical administration" includes, for example, transdermal administration (e.g., via a transdermal patch or iontophoretic device), intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. Topical formulations may include compounds that enhance the absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of the present invention are administered via a transdermal device, administration is accomplished using a patch of reservoir and either a porous membrane type or a solid matrix variant. Typical formulations for this purpose include: gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used. Typical vectors include: alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol, and propylene glycol. Penetration enhancers can be incorporated-see, e.g., b.c. Finnin and t.m. Morgan, j. pharm. sci., vol. 88, pp. 955-.

Formulations suitable for topical administration to the eye include, for example, eye drops wherein a compound of the invention is dissolved or suspended in a suitable carrier. Typical formulations suitable for ocular or otic administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and otic administration include: ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses, and particulate or vesicular systems (e.g., vesicles (niosomes) or liposomes). Polymers, for example, crosslinked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, cellulosic polymers (e.g., hydroxypropyl methylcellulose, hydroxyethyl cellulose, or methyl cellulose), or heteropolysaccharide polymers (e.g., gellan gum (gelan gum)), can be incorporated with preservatives (e.g., benzalkonium chloride). Such formulations may also be delivered by iontophoresis.

For intranasal administration or administration by inhalation, the compounds of the invention are conveniently delivered from pump spray containers (squeezed or pumped by the patient), in the form of solutions or suspensions, or from pressurised containers or nebulisers using suitable propellants, in the form of aerosol spray presentation. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (alone, as a mixture (e.g., with lactose in dry blends), or as a mixed component particle (e.g., with a phospholipid, e.g., lecithin)), from a dry powder inhaler; or as an aerosol spray, by a pressurized container, pump, spray, nebulizer (preferably one that uses electrohydrodynamics to produce a fine mist), or atomizer, with or without the use of a suitable propellant (e.g., 1,1,1, 2-tetrafluoroethane or 1,1,1,2,3,3, 3-heptafluoropropane). For intranasal use, the powder may comprise a bioadhesive, for example, chitosan or cyclodextrin.

In another embodiment, the invention comprises a rectal dosage form. Such rectal dosage forms may be in the form of, for example, suppositories. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.

Other carrier materials and modes of administration known in the pharmaceutical art may also be used. The pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as, for example, effective formulation and administration procedures. The foregoing considerations regarding effective formulation and administration procedures are well known in the art and have been described in standard texts. Formulations of the drugs are discussed, for example, in Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975, Liberman et al, eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe et al, eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.

Co-administration

The compounds of the present invention may be used alone or in combination with other therapeutic agents. The present invention provides any use, method or composition as defined herein, wherein a compound of any of the embodiments of formula I herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable solvate of said compound or salt, is used in combination with one or more other therapeutic agents as discussed herein. This would include a pharmaceutical composition for treating a disease or condition for which an agonist of GLP-1R is indicated, comprising a compound of formula I, II, III, IV, or V as defined in any of the embodiments described herein, or a pharmaceutically acceptable salt thereof, and one or more other therapeutic agents discussed herein.

By "administering two or more compounds in combination" is meant that all compounds are administered sufficiently close in time that each compound can produce a biological effect over the same time frame. The presence of one drug may alter the biological effects of other compounds. Two or more compounds may be administered simultaneously, concurrently or sequentially. In addition, simultaneous administration can be carried out by mixing the mixtures prior to administration or at the same time point in separate dosage forms at the same or different administration sites.

By "concurrently administering", "co-administering", "simultaneously administering", and "simultaneously administering" is meant that the compounds are administered in combination.

In another embodiment, the present invention provides a method of treatment comprising administering a compound of the present invention together with one or more other drugs, wherein the one or more other drugs may be selected from the agents discussed herein.

In one embodiment, the compounds of the present invention are administered with an antidiabetic agent, including but not limited to: biguanides (e.g., metformin (metformin)); sulfonylureas (e.g., tolbutamide, glibenclamide, gliclazide, chlorpropamide, tolazamide, acetophencyclamide, glimepiride, or glipizide); thiazolidinediones (e.g., pioglitazone (pioglitazone), rosiglitazone (rosiglitazone), or lobeglitazone (lobeglitazone)), glitazones (e.g., salograza (saroglitazar), aleglitazone (aleglitazozar), moglicate (muraglitazozar), or tegaser (tesaglitazar)), meglitinides (meglitinides) (e.g., nateglinide (nateglinide), repaglinide (repaglinide)); dipeptidyl peptidase 4(DPP-4) inhibitors (e.g., sitagliptin (sitagliptin), vildagliptin (vildagliptin), saxagliptin (saxagliptin), linagliptin (linagliptin), gitagliptin (gemagliptin), anegliptin (anagliptin), terliptin (teneligliptin), alogliptin (alogliptin), trelagliptin (trelagliptin), dulagliptin (dutogliptin), or alogliptin (omarigliptin)); glitazones (e.g., pioglitazone (pioglitazone), rosiglitazone (rosiglitazone), balaglitazone (balaglitazone), rivoglitazone (rivoglitazone), or lobeglitazone); sodium-glucose cotransporter 2 (SGLT2) inhibitors (e.g., engelizin (empagliflozin), canagliflozin (canagliflozin), dapagliflozin (dapagliflozin), Ipragliflozin (Ipragliflozin), tolagliflozin (tofogliflozin), seragliflozin (seragliflozin etabonate), remogliflozin (remogliflozin etabonate), or egagliflozin); SGLTL1 inhibitors; a GPR40 agonist (FFAR1/FFA1 agonist, e.g., furaglifloam); glucose-dependent insulinotropic polypeptide (GIP) and analogs thereof; alpha glucosidase inhibitors (e.g., voglibose, acarbose, or miglitol); insulin or an insulin analog; including the pharmaceutically acceptable salts of the agents specifically identified as well as the pharmaceutically acceptable solvates of the agents and salts.

In another embodiment, the compounds of the present invention are administered with anti-obesity agents, including but not limited to: peptide YY or analogs thereof, agonists of neuropeptide Y receptor type two (NPYR2), agonists of NPYR1 or NPYR5, agonists of cannabinoid receptor type one (CB1R), lipase inhibitors (e.g., orlistat), human proaxlet peptide (HIP), agonists of melanocortin receptor 4 (e.g., setmellanote), antagonists of melanin-aggregating hormone receptor 1, agonists of Farnesoid X Receptor (FXR) (e.g., obeticholic acid), zonisamide (zonisamide), phentamine (phentermine) (alone or in combination with topiramate), norepinephrine/dopamine reuptake inhibitors (e.g., bupropion), opioid receptor antagonists (e.g., norepinephrine/dopamine), opioid reuptake inhibitors in combination with dopamine inhibitors (e.g., opioid receptor antagonists such as opioid receptor antagonists, GDG-15 analogs, sibutramine (sibutramine), cholecystokinin agonists, amylin and analogs thereof (e.g., pramlintide (pramlintide)), leptin and analogs thereof (e.g., meterolin), serotonin drugs (e.g., lorcaserin (iorricin)), methionine aminopeptidase 2(MetAP2) inhibitors (e.g., beloran or ZGN-1061), phendimetrazine (phenmetrazine), diethylpropion (diethylpropiophenone), benzphetamine (benzphentermine), SGLT2 inhibitors (e.g., engrel, canagliflozin, dapagliflozin, Ipragliflozin (Ipragliflozin), rigagliflozin (Ipragliflozin), toragliflozin, iselezin, epregagliflozin or egagliflozin), inhibitors of dual activators of sgl receptor (SGLT) for cell growth factor (SGLT 36 ) modulators, or a glucagon receptor agonist (alone or in combination with another GLP-1R agonist, e.g., liraglutide, exenatide, dulaglutide, albiglutide, lixisenatide, or semaglutide); including the pharmaceutically acceptable salts of the agents specifically identified as well as the pharmaceutically acceptable solvates of the agents and salts.

In another embodiment, the compounds of the invention are administered in combination with one or more of the following: agents for treating NASH (including, but not limited to, PF-05221304), FXR agonists (e.g., obeticholic acid), PPAR α/δ agonists (e.g., elafibranor), synthetic fatty acid-cholic acid conjugates (e.g., aramchol), caspase inhibitors (e.g., enrichagan), anti-lysyl oxidase homolog 2(LOXL2) monoclonal antibodies (e.g., simtuzumab), galectin 3 inhibitors (e.g., GR-MD-02), MAPK5 inhibitors (e.g., GS-4997), dual antagonists of chemokine receptor 2(CCR2) and CCR5 (e.g., cenicriviroc), fibroblast growth factor 21(FGF21) agonists (e.g., BMS-986036), leukotriene D4(LTD4) receptor antagonists (e.g., tylukast (tipelikast analogs), nicotinic acid analogs (e.g., ARI 7MO), ASBT inhibitors (e.g., 303303), volixibat), acetyl-coa carboxylase (ACC) inhibitors (e.g., NDI 010976 or PF-05221304), ketohexokinase (KHK) inhibitors, diacylglycerol acyltransferase 2(DGAT2) inhibitors, CB1 receptor antagonists, anti-CB 1R antibodies, or apoptosis signal-modulating kinase 1(ASK1) inhibitors, including pharmaceutically acceptable salts of the agents specifically identified and pharmaceutically acceptable solvates of the agents and salts.

Certain specific compounds that may be used in combination with the compounds of the present invention for the treatment of the diseases or disorders described herein (e.g., NASH) include:

4- (4- (1-isopropyl-7-oxo-1, 4,6, 7-tetrahydrospiro [ indazole-5, 4 '-piperidine ] -1' -carbonyl) -6-methoxypyridin-2-yl) benzoic acid, which is an example of a selective ACC inhibitor and is prepared in the form of 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 form (form 1) and the trihydrate of the mono-tris salt (form 2), described in international PCT application No. PCT/IB2018/058966, the disclosure of which is hereby incorporated by reference in its entirety for all purposes;

(S) -2- (5- ((3-ethoxypyridin-2-yl) oxy) pyridin-3-yl) -N- (tetrahydrofuran-3-yl) pyrimidine-5-carboxamide, or a pharmaceutically acceptable salt thereof, and crystalline solid forms thereof (forms 1 and 2) are examples of DGAT2 inhibitors described in example 1 of U.S. patent No. 10,071,992, the disclosure of which is hereby incorporated by reference in its entirety for all purposes;

[ (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 (including a crystalline free acid form thereof), is an example of a ketohexokinase inhibitor and is described 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; and is

The FXR agonist tropifuexor 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.

These agents and compounds of the invention can be combined with a pharmaceutically acceptable vehicle (e.g., saline, ringer's solution, dextrose solution, and the like). The particular dosage regimen, i.e., dosage, timing and repetition, will depend on the particular individual and the individual's medical history.

Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations used, and may include: buffers (e.g., phosphates, citrates, and other organic acids); salts (e.g., sodium chloride); antioxidants (including ascorbic acid and methionine); preservatives (e.g., octadecyl dimethyl benzyl ammonium chloride; hexa hydroxy quaternary ammonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, e.g., methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins (e.g., serum albumin, gelatin, or Igs); hydrophilic polymers (e.g., polyvinylpyrrolidone); amino acids (e.g., glycine, glutamine, asparagine, histidine, arginine, or lysine); monosaccharides, disaccharides, and other carbohydrates (including glucose, mannose, or dextrins); chelating agents (e.g., EDTA); sugars (e.g., sucrose, mannitol, trehalose, or sorbitol); salt-forming counterions (e.g., sodium); metal complexes (e.g., zinc-protein complexes); and/or nonionic surfactants (e.g., TWEEN) TM、PLURONICSTMOr polyethylene glycol (PEG)).

Liposomes containing these agents and/or compounds of the invention are prepared by methods known in the art, for example, as described in U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with increased circulation time are disclosed in U.S. Pat. No. 5,013,556. Particularly useful liposomes can be produced by reverse phase evaporation methods using lipid compositions comprising lecithin, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). The liposomes are extruded through a filter having a defined pore size to obtain liposomes having the desired diameter.

These drugs and/or compounds of the invention can also be embedded in microcapsules prepared, for example, by coacervation techniques or interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively; embedded in colloidal drug delivery systems (e.g., liposomes, albumin microparticles, microemulsions, nanoparticles, and nanocapsules) or embedded in macroemulsions. This technique is disclosed in Remington, The Science and Practice of Pharmacy, 20th Ed., Mack Publishing (2000).

Sustained release formulations may be used. Suitable examples of sustained release formulations include semipermeable matrices of solid hydrophobic polymers containing a compound of formula I, II, III, IV, or V, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained release matrices include: polyesters, hydrogels (e.g., poly (2-hydroxyethyl-methacrylate), or poly (vinyl alcohol)), polylactic acid (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and 7-ethyl-L-glutamine, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers (e.g., for LUPRON DEPOT) TM(those in injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)), sucrose acetate isobutyrate, and poly-D- (-) -3-hydroxybutyric acid.

Formulations for intravenous administration must be sterile. This can be readily accomplished, for example, by filtration through sterile filtration membranes. The compounds of the invention are typically placed in a container having a sterile access port, e.g., an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.

Suitable emulsions are commercially available fat emulsions, e.g., IntralipidTM、LiposynTM、InfonutrolTM、LipofundinTMAnd LipiphysanTMAnd (4) preparation. The active ingredient may be dissolved in a pre-mixed emulsion composition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed by mixing a phospholipid (e.g., egg phospholipid, soy phospholipid or soy lecithin) and water. It will be appreciated that other ingredients, for example, glycerol or glucose, may be added to adjust the tonicity of the emulsion. Suitable emulsions typically contain up to 20% oil, for example, 5 to 20%. The fat emulsion may comprise fat droplets of 0.1 to 1.0 μm, in particular 0.1 to 0.5 μm, and have a pH in the range of 5.5 to 8.0.

The emulsion composition may be prepared by mixing a compound of the present invention with an IntralipidTMOr those prepared by mixing the components thereof (soybean oil, egg phospholipids, glycerin and water).

Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents or mixtures thereof, as well as powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out hereinbefore. In certain embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic efficacy. Compositions in a preferred sterile pharmaceutically acceptable solvent can be nebulized by the use of a gas. Nebulized solutions can be inhaled directly by the nebulizing device or the nebulizing device can be connected to a face mask, a curtain, or an intermittent positive pressure respirator. The solution, suspension or powder composition may be administered by a device that delivers the formulation in a suitable manner, preferably orally or nasally.

Reagent kit

Another aspect of the invention provides a kit comprising a compound of formula I, II, III, IV, or V or a pharmaceutical composition comprising a compound of formula I, II, III, IV, or V of the invention. In addition to the compounds of formula I, II, or III, or pharmaceutical compositions thereof, of the present invention, the kit can also include a diagnostic or therapeutic agent. The kit may also include instructions for use in a diagnostic or therapeutic method. In certain embodiments, the kit comprises a compound of formula I, II, III, IV, or V, or a pharmaceutical composition thereof, and a diagnostic agent. In other embodiments, the kit comprises a compound of formula I, II, III, IV, or V, or a pharmaceutical composition thereof.

In yet another embodiment, the invention comprises a kit suitable for performing the methods of treatment described herein. In one embodiment, the kit contains a first dosage form comprising one or more compounds of the invention in an amount sufficient to perform the methods of the invention. In another embodiment, a kit comprises one or more compounds of the invention in an amount sufficient to perform the methods of the invention and a container for the dosage.

Preparation of

The compounds of formula I, II, III, IV, or V may be prepared by the general and specific methods described below using common general knowledge of those skilled in the art of synthetic organic chemistry. This common general knowledge can be found in standard reference books, such as, for example, Comprehensive Organic Chemistry, Ed. Barton and Ollis, Elsevier, Comprehensive Organic Transformations, A Guide to Functional Group Preparations, Larock, John Wiley and Sons; and the company of Organic Synthetic Methods, Vol. I-XII (published by Wiley-Interscience). The starting materials used herein are either commercially available or can be prepared by routine methods known in the art.

In the preparation of compounds of formula I, II, III, IV, or V, it is noteworthy that certain preparation methods described herein may require protection of a distal functional group (e.g., primary amine, secondary amine, carboxyl group in the precursor of formula I). The need for such protection will vary depending on the nature of the distal functional group and the conditions of the preparation method. The need for such protection can be 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.

For example, certain compounds contain primary amine or carboxylic acid functional groups, which if unprotected, may interfere with reactions at other sites in the molecule. Thus, such functional groups may be protected by suitable protecting groups, which may be removed in a subsequent step. Suitable protecting groups for amine and carboxylic acid protection include those commonly used in peptide synthesis (e.g., N-tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), and 9-fluorenylmethoxycarbonyl (Fmoc) for amines, and lower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can generally be removed without chemically altering other functional groups in the compounds of formula I.

The schemes described below are intended to provide a general description of the methods employed in the preparation of the compounds of the present invention. Certain compounds of the invention may contain a single or multiple chiral centers, bearing the stereochemical designation (R) or (S). It will be apparent to those skilled in the art that all synthetic transformations can be performed in a similar manner, whether the material is enantiomerically enriched or racemic. Further, resolution into the desired optically active material can be carried out at any desired point within the sequence using well known methods (e.g., as described herein and in the chemical literature). For example, intermediates (e.g., S4, S7, S8, S24, S40, and S41) and final products (e.g., S25 and S42) can be separated using chiral chromatography. Alternatively, chiral salts can be utilized to separate enantiomerically enriched intermediates and final compounds.

In the scheme below, variable X, Y, Z1、Z2、Z3、R、R1、R2、R3、R4M, p, and q are as defined herein for compounds of formula I, II, III, IV, or V, unless otherwise specified. For the sake of simplicity, the variable A is used to denote the ring A and its optional substituents R1. For the scheme provided below, X1、X2、X3And X4Each may independently be a leaving group, for example, any alkyl or aryl sulfonate (e.g., mesylate, tosylate, or triflate), or halogen, or any other group that may be replaced by an amine or used in a metal-mediated coupling reaction. X4And may also be a protected carboxylic acid (i.e., ester). When the protecting group is recognized as Pg1When it is an alkylamineProtecting groups (e.g., benzyl, benzhydryl, allyl, and the like); carbamate protecting groups (e.g., Boc, Cbz, etc.); or an amide protecting group (e.g., trifluoroacetamide). When the protecting group is recognized as Pg2When it is an acid protecting group, for example, methyl, ethyl, benzyl, tert-butyl and the like. When the protecting group is recognized as Pg3When present, it may be an alcohol protecting group (e.g., trimethylsilylethoxyethyl); or an acyl group (e.g., acetyl, benzoyl, etc.); or trialkylsilyl groups (e.g., trimethylsilyl, t-butyldimethylsilyl, triisopropylsilyl, etc.). R 2aIs H or-C1-2Alkyl, wherein alkyl may have 0 to 1 OH. R4aIs C1-2Alkyl radical, C0-2alkylene-C3-6Cycloalkyl radical, C0-2alkylene-R5Or C1-2alkylene-R6Wherein if allowed by valence, said alkyl, alkylene, or cycloalkyl can be independently substituted with 0 to 3F atoms and 0 to 1 independently selected from C0-1alkylene-OROand-N (R)N)2Is substituted with the substituent(s).

Substituted piperidine S8 (wherein R2= H and Y = CH) can be prepared as discussed in scheme 1. The aryl or heteroaryl bromide S1 can be treated with an alkyl lithium (e.g., butyl lithium or tert-butyl lithium) to give an aryl-or heteroaryl-lithium species, which can be reacted with the aldehyde S2 to give the diol S3. Other aryl or heteroaryl organometallic reagents (e.g., without limitation, grignard reagents) may also be used to prepare S3. The reaction is usually carried out at a temperature of about-70 ℃. Then, NaIO can be used4Oxidation of diol S3 to provide the acetal S4 (R)2= H). Compound S4 can then be reacted with a substituted boronic acid or boronic ester (S5) in the presence of a palladium catalyst and a ligand complex in the manner of a suzuki reaction (malueda and navrro, Molecules, 2015, 20, 7528-a 7557) to provide a compound of formula S6. Reduction of the olefin to provide the compound of general structure S7 can be carried out under a hydrogen atmosphere (15-100 psi H) 2) In an alcoholic solvent (e.g. methanol or ethanol) or alternatively in an aprotic organic solvent (e.g. ethyl acetate or THF), in a suitable catalyst (e.g. supported onPalladium on charcoal, Pd (OH) on charcoal2(Pearlman catalyst), PtO2 (Adams catalyst), or tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst)). Using a suitable catalyst, a transfer hydrogenation reagent (e.g., ammonium formate or dihydrobenzene, or the like) may be employed. Alternatively, reduction may be accomplished by alternative methods known to those skilled in the art, using reagents (e.g., triethylsilane or other silanes), under acid or metal catalysis, or using metal reducing agents (e.g., magnesium or the like). Alternatively, the olefin may be functionalized by methods known to those skilled in the art to introduce R3A group. For example, the olefin may be hydroborated to produce an alcohol, which may be alkylated or further converted to a nitrile, F, or alkyl group. Pg1Can be performed using a variety of methods described in the literature to provide the amine S8.

Scheme 1

Scheme 2 provides an alternative preparation of compounds of general structure S4. Reaction of an appropriately substituted diol of general structure S9 with an aldehyde or ketone of general structure S10a in the presence of an acid (e.g., p-toluenesulfonic acid or pyridine p-toluenesulfonic acid) in an aprotic organic solvent (e.g., toluene or benzene) can give a compound of general structure S4. The reaction is heated at reflux, typically using a Dean-Stark trap, to remove water azeotropically. The diol S9 can also be reacted with a cyclic (dotted line present) or acyclic (dotted line absent) acetal or ketal of the general structure S10b under acidic catalysis. The same applies to the use of cyclic or acyclic thioacetals or thioketals of the general structure S10c under the influence of mercury salts, mild oxidizing agents or alkylating agents to give the compound S4. Alternatively, a diol of the general formula S9 can be reacted with an appropriately substituted alkyne S11 in an aprotic solvent (e.g., toluene) in the presence of triruthenium dodecacarbonyl at a temperature of about 100 ℃ to give a compound of the general structure S4 (wherein R is 2=CH2R2a). In which R is2Containing alcoholic functional groups (e.g. CH)2OH), alcohol protecting group (Pg)3) (e.g., acetate) may be incorporated into the compound of general structure S10. The protecting group may then be removed in a subsequent step. Intermediate S4 may then be further modified using the methods described for scheme 1 to provide amines of general structure S8.

Scheme 2

As provided in scheme 3, conversion of S4 to a compound of general structure S7 (where Y = N) can be accomplished by means such as Buchwald-Hartwig C-N coupling between a compound of general structure S4 and an appropriately substituted and protected piperazine S12 in the presence of a palladium or copper catalyst and a ligand complex. These reactions are usually carried out at 0 to 110 ℃ with the addition of a base (e.g., Cs)2CO3LiHMDS or NaOtBu) with an aprotic organic solvent (such as, but not limited to, 1, 4-dioxane and PhCH3) Is carried out in (1). Pg1Can be performed using a variety of methods described in the literature to provide amine S8 (where Y = N).

Scheme 3

Amine compounds S8 prepared via the methods described in schemes 1-3 can be prepared using protected 2-bromoacetate in an appropriate base (e.g., K)2CO3、Et3N, NaH or LiHMDS) in a polar aprotic solvent (such as, but not limited to, DMF, DMAc, DMSO, or NMP) to give a compound of general structure S13 (X = N, L = CH) 2). Standard ester hydrolysis may be performed to provide the acid S14. If Pg2Is t-butyl, standard acidic deprotection methods (e.g., TFA/DCM, HCl/1, 4-dioxane, HCl/EtOAc or other suitable conditions) can be used to afford the acid S14. If Pg2Is methyl or ethyl, standard basic deprotection methods (e.g., aqueous NaOH in methanol or ethanol, or other suitable strips) can be usedPiece) to give the acid S14.

Scheme 4

Compounds of general structure S15 (scheme 5) can be prepared in the presence of a base (e.g., sodium carbonate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, cesium bicarbonate, sodium hydroxide, potassium hydroxide, cesium hydroxide, sodium acetate, potassium acetate, or cesium acetate) or an organic amine base (e.g., Et3N, DIPEA, DBU, etc.), with an amine R in a polar aprotic solvent (such as, but not limited to, THF, DMF, DMAc, DMSO, or NMP) or a protic solvent (such as, water, MeOH, EtOH, or iPrOH), or mixtures thereof4NH2To give a compound of general structure S16. It should be noted that if the examples provide R with a resolved enantiomeric center4The other enantiomer or a racemic mixture thereof may be obtained by selecting the appropriate starting material. Preferred X 3Substituents include F, Cl, and Br, preferably X4The radicals including Cl, Br, and-CO2-Pg2. Reduction of nitro groups can be achieved by using a metal catalyst (e.g., palladium on charcoal or Raney nickel), in a protic solvent (e.g., methanol or ethanol) or an aprotic solvent (e.g., DMF, THF or EtOAC) at 1-6 atm H2By hydrogenation. Alternatively, iron, zinc, SnCl can be used as nitro group2Or other suitable metal in an acidic medium (e.g., 1N HCl, AcOH, or aqueous NH)4Cl (in THF or methanol)) to provide the compound of general structure S17 (scheme 5 a). Compounds such as S18 may be acylated by an acyl halide in standard manner or by a carboxylic acid ester via standard amide coupling schemes to provide compound S19. Reduction to compound S20 can be carried out under standard conditions using a reducing agent (e.g., LAH or BH)3-THF or BH3DMS) (scheme 5 b).

Scheme 5

Diamine compounds S17 and S20, collectively referred to as diamine S21 (scheme 6), prepared via the processes described in schemes 5a and 5b, can be acylated using an acid of general structure S14 under standard amide coupling schemes to afford amide S22, which is present as a mixture of 100% S22a to 100% S22 b. This mixture of amines S22 can be cyclized by various methods to give compounds of general structure S23. The amine S22 can be reacted with a dehydrating agent (e.g., T) under microwave conditions (10-60 minutes at 120 ℃ C. and 180 ℃ C.) 3P®) Or an alkanol (e.g., n-butanol) together to provide compound S23. Alternatively, a mixture of compound S22 can be heated under acidic conditions (e.g., AcOH, 60-100 ℃) or basic conditions (e.g., aqueous NaOH or KOH (in 1, 4-dioxane), 60-100 ℃) to provide S23. Compounds of general structure S23 (X)4= Cl, Br or I) may be converted to an ester of structure S24 by palladium catalysed carbonylation using a suitable alcohol (e.g. MeOH or EtOH or other alkanol) at a temperature of 20 to 100 ℃ under an atmosphere of 15 to 100 psi carbon monoxide. Hydrolysis of the ester S24 can be performed as described in scheme 4 to provide the acid S25. To compound S22 (wherein X4=CO2-Pg2) For the purposes, the conversion to the ester S24 is carried out under similar conditions as described previously except that a basic cyclization process is used in which the compound S25 can be isolated directly from the reaction mixture. To compound S24 (wherein X4Is CO2tBu), deprotection to the acid S25 can be carried out under acidic conditions as described in scheme 4. Alternatively, compound S24 (wherein Pg2Is C1-C8Alkyl groups, e.g., methyl, ethyl, hexyl, or octyl), ester deprotection can be carried out using a variety of enzymes well known to those skilled in the art, including esterases, proteases, peptidases, lipases, and glycosidases. The hydrolysis reaction can also be carried out by using 1,5, 7-triazabicyclo [4.4.0 ]Aqueous solution of dec-5-ene, by treating the ester at room temperature.

Scheme 6

In addition, the diamine S21 can be converted to 2-chloromethylbenzimidazole S26 by several methods (scheme 7). Treatment with 2-chloroacetyl chloride or chloroacetic anhydride in an aprotic solvent (e.g., 1, 4-dioxane) followed by heating at 40-100 deg.C for 2-18 hours affords the desired benzimidazole S26 (where Z is1、Z2And Z3Is CH). In which Z is1、Z2And Z3Are not all CRzIn the case of (a), after treatment with 2-chloroacetyl chloride in an aprotic solvent (e.g., 1, 4-dioxane) for 30 minutes to 4 hours, the solvent is exchanged for an acidic medium (e.g., AcOH or TFA) and then heated at 40-100 ℃ for 2-18 hours to provide the desired compound S26. Diamine S21 can also be treated with chloroacetic anhydride in an aprotic solvent (such as, but not limited to, 1, 4-dioxane, THF, or MeCN) at a temperature of 0 to 80 ℃ followed by heating at 60-100 ℃ for 2 to 18 hours to afford the desired compound S26. In addition, diamine S21 can be treated with 2-chloro-1, 1, 1-trimethoxyethane in an aprotic solvent (such as, but not limited to, 1, 4-dioxane, THF, or MeCN) or a protic solvent (such as MeOH or EtOH) in the presence of an acid catalyst (such as pTSA) at 20-100 ℃. Alternatively, the diamine S21 can be heated with 2-hydroxyacetic acid in an aprotic solvent (such as, but not limited to, mesitylene) at 100 ℃ and 180 ℃ to provide the hydroxymethyl intermediate. Conversion of the hydroxymethyl group to chloromethyl Compound S26 can be carried out by standard methods (including the use of SOCl in an aprotic solvent) 2Process) is completed. Can be in the presence of a base (e.g., sodium carbonate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, or cesium bicarbonate, NaH) or an organic amine base (e.g., Et3N, DIPEA, DBU, etc.) in a polar aprotic solvent (such as, but not limited to, THF, MeCN, DMF, DMAc, DMSO, or NMP), compound S8 is reacted with a compound of general structure S26 to give compound S23 (X)4= Cl, Br, I) or compound S24 (X)4=CO2-Pg2) It was then used to obtain compound S25 via the method described in scheme 6.

Scheme 7

Alternatively (scheme 8), compounds of general structure S26 can be in the presence of a base (e.g., sodium carbonate, potassium carbonate, cesium carbonate, sodium bicarbonate, potassium bicarbonate, or cesium bicarbonate, NaH) or an organic amine base (e.g., Et)3N, DIPEA, DBU, etc.) in a polar aprotic solvent (such as, but not limited to, THF, MeCN, DMF, DMAc, DMSO, or NMP), with an appropriately substituted and protected piperazine S12 to provide compound S27 (scheme 8). Pg1Can be performed using a variety of methods described in the literature to provide the amine S28. General Structure S23 (X)4= Cl, Br or I) or S24 (X)4=CO2-Pg2) The transformation of compounds of (a) can be accomplished by way of a Buchwald-Hartwig C-N coupling between a compound such as that of general structure S4 and that described previously in scheme 3. Compounds of general structure S23 or S24 can then be used to obtain compounds of structure S25 via the methods described in scheme 6.

Scheme 8

Compounds of general structure S25 can also be prepared as discussed in scheme 9. The diol S9 may be protected to give S29. Trimethylsilyl ethoxymethyl group is a preferred protecting group. It is also preferred to protect the diol as the corresponding acetal (e.g., formaldehyde acetal). Compound S29 can then be reacted with a substituted boronic acid or ester (S5) and the olefin then reduced using the method described in scheme 1 to provide a compound of general structure S31 (where Y = CH). Alternatively, compound S29 can be coupled to piperazine of general structure S12 using the method described in scheme 3 to provide S31 (where Y = N). Compounds of general structure S31 can be deprotected using the methods described in scheme 7 and then coupled with S26 to give compounds of general structure 33. Alternatively, compounds of general structure S33 can be prepared from S32 by converting S32 to the corresponding N-acetic acid derivative as described in schemes 4 and 6, and subsequently condensing it with diamine S21. Deprotection of S33 using methods known to those skilled in the art can provide a diol of general structure S34, which can then be reacted with an alkyne of general structure S11 using the methods described in scheme 2 to provide S23 or S24. Alternatively, as discussed in scheme 2, S34 can be converted to S23 or S24 using aldehydes, ketones, or derivatives thereof. Compounds of general structure S23 or S24 can then be used to obtain compounds of structure S25 via the methods described in scheme 6.

Scheme 9

Compounds of general structures S24 and S33 (where Y = N and X-L = cyclopropyl) can be prepared as discussed in scheme 10. Protected piperidone S35 can be homologated to the unsaturated ester S36 using methods well known to those skilled in the art. For example, Horner-Wadsworth-Emmons olefination of S42 with a phosphate ester (e.g., (diethoxyphosphoryl) ethyl acetate) that has been deprotonated using a strong base (e.g., lithium, sodium, or potassium tert-butoxide) can provide S36. The reaction is generally carried out in an aprotic solvent such as THF or DME at a temperature of from about 0 to-50 ℃. The conversion of S36 to the cyclopropane derivative S37 can be accomplished by treatment with a sulfonium ylide (sulfoxonium ylide) derived from trimethyl sulfoxide iodide (trimethylulfoxonium iodide) and a base (e.g., potassium tert-butoxide or sodium hydride). Deprotection of S37 and subsequent resulting carboxylic acids S38 and S21 (wherein X is4=CO2Pg2) Can provide a compound of general structure S39. Deprotection of S39 and coupling with S4 using the method described in scheme 3 gave compounds of general structure S24 (where Y = N and X-L is cyclopropyl). The compound of general structure S24 can then be used to obtain the compound of structure S25 via the methods described in scheme 6. Alternatively, S40 may be reacted with S29 using the method described in scheme 3 to provide S33 (where Y = N and X-L = cyclopropyl). The compounds of general structure S33 can then be used to obtain the compounds of structure S25 via the methods described in schemes 6 and 9.

Scheme 10

Alternatively, compounds of general structure S25 (wherein Y = N and X-L is cyclopropyl) can be prepared as described in scheme 11. Pg removal by S371To provide piperidine derivative S43. Coupling of S43 with S4 in a similar manner as described in scheme 3 provides S13 (where Y = N and X-L is cyclopropyl). Deprotection can then provide a compound of general structure S14, which can then be used to prepare S25 as described in scheme 6.

Scheme 11

[ examples ] A method for producing a compound

The following illustrates the synthesis of non-limiting compounds of the invention. Other compounds within the scope of the invention may be prepared using the methods illustrated in these examples, either alone or in conjunction with techniques generally known in the art.

In general, the experiments are carried out under an inert atmosphere (nitrogen or argon), in particular when reagents or intermediates sensitive to oxygen or moisture are used. Commercial solvents and reagents are typically used without further purification. If desired, anhydrous solvents are used, usually Acroseal from Acros Organics®Product, Aldrich from Sigma-Aldrich® Sure/SealOr Drisolv from EMD Chemicals®And (5) producing the product. In other cases, a commercial solvent was passed through a column packed with 4 a molecular sieve until the QC standard for the following water was reached: a) <100 ppm for methylene chloride, toluene,N,N-dimethylformamide and tetrahydrofuran; b)<180 ppm for methanol, ethanol, 1, 4-dioxane, and diisopropylamine. For very sensitive reactions, the solvent is further treated with sodium metal, calcium hydride, or molecular sieves and distilled just before use. The product is usually dried in vacuum before further reaction or submitted to biological testing. Mass spectrometry data are reported by liquid chromatography-mass spectrometry (LCMS), Atmospheric Pressure Chemical Ionization (APCI), or gas chromatography-mass spectrometry (GCMS) instruments. Symbol ♦ indicates that a chlorine isotope pattern is observed in the mass spectrum.

Chiral separations are used to separate enantiomers or diastereomers of certain intermediates during the preparation of the compounds of the invention. When chiral separation is complete, the separated enantiomers are designated ENT-1 or ENT-2 (or DIAST-1 or DIAST-2) depending on their elution order. In certain embodiments, the enantiomer designated ENT-1 or ENT-2 may be used as a starting material to prepare other enantiomers or diastereomers. In this case, the enantiomers obtained are designated, according to their starting materials, ENT-X1 and ENT-X2, respectively; similarly, the prepared diastereomers are designated DIAST-X1 and DIAST-X2 (or DIAST-), respectively, depending on the starting material. In syntheses using multiple intermediates, the DIAST-Y and DIAST-Z nomenclature is similarly used.

In the case of compounds with two chiral centers, the stereoisomers at each stereocenter are separated at different times. Designation of ENT-1 or ENT-2 (or DIAST-1 or DIAST-2) for intermediates or examples refers to the elution order of the separations performed at the step in question. It will be appreciated that when stereoisomers of chiral centers are separated in a compound with two or more centers, the separated 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. P18 was then used to prepare C70, which had a mixture of one stereoisomer enriched on the cyclopropyl chiral carbon and one stereoisomer on the dioxolane carbon. C70 was then isolated as DIAST-Y1 on the dioxolane carbon to afford intermediate C71, and DIAST-Y2 on the dioxolane carbon to afford intermediate C72, wherein these intermediates were enriched in a single stereoisomer. C71 was then used to prepare example 15, which was named 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ]-6-azaspiro[2.5]Oct-1-yl } -1- (2-methoxyethyl) -1HBenzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate [ starting from P18 via C71]. In these preparations, after the mixture is subjected to a separation procedure, the chiral center is identified as "abs" near the center, with the understanding that the separated enantiomer may not be enantiomerically pure. Typically, the enriched enantiomers at each chiral center are isolated>90 percent. Preferably, the enriched enantiomers at each center are in a mixture>98%。

In certain embodiments, the optical rotation of an enantiomer is measured using a polarimeter. According to their observed rotation data (or their specific rotation data), the clockwise rotated enantiomer is designated as the (+) -enantiomer and the counterclockwise rotated enantiomer is designated as the (-) -enantiomer. Racemic compounds are represented by an undepicted or depicted stereochemistry, or by the appearance of (+/-) next to the structure; in the latter case, the stereochemistry shown represents the relative (rather than absolute) configuration of the substituents of the compound.

Reactions performed by detectable intermediates are typically followed by LCMS and allowed to proceed to full conversion before subsequent reagents are added. The reaction conditions (reaction time and temperature) may be varied for syntheses referring to procedures in other examples or methods. Generally, the reaction is followed by thin layer chromatography or mass spectrometry and, where appropriate, by work-up. Purification may vary from experiment to experiment: in general, the solvent and solvent ratio for the eluent/gradient are selected to provide the appropriate R fs or retention time. All starting materials in these preparations and examples are commercially available or can be prepared by methods known in the art or as described herein.

Preparation P1

4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine-1-carboxylic acid tert-butyl ester (P1)

Step 1: 2-bromo-6- [ (4-chloro-2-fluorophenyl) (hydroxy) methyl]Synthesis of phenol (C1)

This experiment was performed in two batches of the same scale. N-butyllithium (2.5M solution in hexanes; 32.8 mL, 82.0 mmol) was added slowly to a-70 ℃ solution of 1-bromo-4-chloro-2-fluorobenzene (17.2g, 82.1 mmol) in diethyl ether (100 mL) while maintaining the temperature of the reaction mixture below-60 ℃. After stirring the reaction mixture at-70 ℃ for 20 minutes, a solution of 3-bromo-2-hydroxybenzaldehyde (5.5g, 27 mmol) in diethyl ether (100 mL) was added slowly while maintaining the reaction temperature below-60 ℃. After stirring at-70 ℃ for a further 1 h, the reaction was quenched by addition of aqueous ammonium chloride (50 mL) at-70 ℃ 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 sodium chloride (200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (gradient: 0% to 7% ethyl acetate in petroleum ether) afforded C1 as a white solid. And (4) merging yield: 15.7g, 47.4 mmol, 88%. 1H NMR (400 MHz, chloroform-d) δ 7.44 (dd, J = 8.0, 1.5 Hz, 1H), 7.37 (dd, J = 8.1, 8.1 Hz, 1H), 7.15 (br dd, J = 8.5, 2.1 Hz, 1H), 7.12 - 7.05 (m, 2H), 6.80 (dd, J = 7.8, 7.8 Hz, 1H), 6.78 (s, 1H), 6.31 (d, J = 4.8 Hz, 1H), 3.02 (br d, J = 4.9 Hz, 1H)。

Step 2: synthesis of 4-bromo-2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxole (C2)

A solution of sodium periodate (25.4g, 119 mmol) in water (105 mL) was added to a solution of C1(15.7g, 47.4 mmol) in methanol (450 mL) and the reaction mixture was stirred at 30 ℃ for 16 h whereupon it was concentrated in vacuo. After the residue was diluted with dichloromethane (500 mL), 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) afforded C2 as a white solid. Yield: 10.0g, 30.3 mmol, 64%. The following1H NMR data were obtained from experiments performed in the same manner but on a smaller scale.1H NMR (400 MHz, DMSO-d 6) δ 7.67 - 7.61 (m, 2H), 7.50 (s, 1H), 7.43 (br dd, J = 8, 2 Hz, 1H), 7.09 (dd, J = 8.3, 1.1 Hz, 1H), 7.01 (dd, J = 7.9, 1.1 Hz, 1H), 6.86 (dd, J = 8.1, 8.1 Hz, 1H)。

And step 3: 4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]-3, 6-dihydropyridine- 1(2H) Synthesis of tert-butyl formate (C3)

Containing C2(8.00g, 24.3 mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2)H) Tert-butyl formate (9.01g, 29.1 mmol), sodium carbonate (5.15g, 48.6 mmol), and [1, 1' -bis (diphenylphosphino) ferrocene ]Palladium (II) dichloride [ Pd (dppf) Cl2;888 mg,1.21 mmol]The reaction flask, which was a suspension in 1, 4-dioxane (80 mL) and water (32 mL), was evacuated and charged with nitrogen. This cycle of evacuation 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 (200 mL) and water (200 mL). The organic layer was washed with saturated aqueous sodium chloride solution (100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (gradient: 0% to 4.3% ethyl acetate in petroleum ether) afforded the product, which was combined with material from an analogous reaction carried out using C2(2.00g, 6.07 mmol) to give C3 as a pale yellow gum. And (4) merging yield: 10.3g, 23.8 mmol, 78%.1H NMR (400 MHz, chloroform-d) δ 7.53 (dd, J = 8.3, 7.8 Hz, 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, 5 Hz, 2H), 2.59 - 2.50 (br m, 2H), 1.48 (s, 9H)。

And 4, step 4: 4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine-1-carboxylic acid tert-butyl ester Synthesis of butyl ester (P1)

A solution of C3(10.3g, 23.8 mmol) and tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst; 1.54g, 1.66 mmol) in methanol (100 mL) was stirred at 50 ℃ under hydrogen (45 psi) for 18 h. Then, the reaction mixture was filtered through a celite pad, and under reduced pressure, the filtrate was concentrated 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 carried out with C3(1.67g, 3.87 mmol) to give P1 as a colourless gum. And (4) merging yield: 10.3g, 23.7 mmol, 86%. LCMS m/z456.1♦ [M+Na+]。1H NMR (400 MHz, chloroform-d) δ 7.52 (dd, J = 8.5, 7.6 Hz, 1H), 7.23 - 7.17 (m, 2H), 7.16 (s, 1H), 6.83 (dd, J = 7.8, 7.8 Hz, 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 Butyl ester (P2)

Step 1: synthesis of 4-bromo-2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole (C4)

1- (4-chloro-2-fluorophenyl) ethanone (316g, 1.83 mol) and p-toluenesulfonic acid (6.02g, 35.0 mmol) were added to a solution of 3-bromobenzene-1, 2-diol (330g, 1.75 mol) in toluene (1.5L). The reaction apparatus was equipped with a 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); a yellow oil was obtainedC4 in admixture with solids. Yield: 158g, 460 mmol, 26%.1H NMR (400 MHz, chloroform-d): δ 7.54 (dd, J = 8.4, 8.4 Hz, 1H), 7.17 - 7.10 (m, 2H), 6.95 (dd, J= 7.9, 1.4 Hz, 1H), 6.75 (dd, components of the ABX pattern,J= 7.8, 1.4 Hz, 1H), 6.70 (dd, components of the ABX pattern,J = 7.9, 7.9 Hz, 1H), 2.11 (d, J = 1.1 Hz, 3H)。

step 2: 4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]-3, 6-bis Hydropyridine-1 (2)H) Synthesis of tert-butyl formate (C5)

Reacting 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2)H) Tert-butyl formate (62g, 200 mmol) and sodium carbonate (100g, 940 mmol) were added to a solution of C4(58.0g, 169 mmol) in 1, 4-dioxane (600 mL). Adding [1, 1' -bis (diphenylphosphino) ferrocene ]After palladium (II) dichloride (6.0g, 8.2 mmol), the reaction mixture was heated to 90 ℃ and stirred for 16 hours. Then, water (500 mL) was added and the resulting mixture was extracted with ethyl acetate (2 × 500 mL). The combined organic layers were washed with saturated aqueous sodium chloride (2 × 500 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (gradient: 0% to 9% ethyl acetate in petroleum ether) afforded C5 as a yellow oil. Yield: 56.0g, 126 mmol, 75%.1H NMR (400 MHz, chloroform-d) δ 7.50 (dd, J= 8.2, 8.2 Hz, 1H), 7.17-7.09 (m, 2H), 6.83-6.77 (m, 2H), 6.74 (dd, component of ABX pattern,J = 5.4, 3.6 Hz, 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 = 1.1 Hz, 3H), 1.50 (s, 9H)。

and step 3: 4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine- Synthesis of tert-butyl 1-carboxylate (P2)

Tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst; 8.10g, 8.75 mmol) was added to a solution of C5(56.0g, 126 mmol) in methanol (200 mL) and the solution was cooled under hydrogen (45 psi)The reaction mixture was heated to 50 ℃ for 18 hours. 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 give P2 as a yellow solid. Yield: 37.0g, 82.6 mmol, 66%. LCMS m/z392.1 ♦ [ (M-2-methylprop-1-ene) + H]+1H NMR (400 MHz, chloroform-d) δ 7.51 (dd, J= 8.3, 8.0 Hz, 1H), 7.17-7.09 (m, 2H), 6.77 (dd, components of ABC mode,J= 7.8, 7.8 Hz, 1H), 6.70 (dd, components of ABC mode,J= 7.7, 1.3 Hz, 1H), 6.66 (dd, components of ABC mode,J = 7.8, 1.3 Hz, 1H), 4.37 - 4.13 (br m, 2H), 2.92 - 2.73 (m, 3H), 2.05 (d, J = 1.1 Hz, 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 Tosylate salt (P3)

Step 1: 4- [(2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridine-1-carboxylic acid tert-butyl ester (C6) and 4- [ (2)S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole En-4-yl]Isolation of tert-butyl piperidine-1-carboxylate (C7)

P2(75.2g, 168 mmol) was separated into its component enantiomers by SFC (supercritical fluid chromatography) [ column: 5 mu m of Chiral Technologies Chiral pak AD-H; mobile phase: 4: 1 carbon dioxide/(2-propanol containing 0.2% 1-aminopropan-2-ol). The first eluted compound was designated C6, and the second eluted enantiomer was designated C7. The absolute configuration indicated is assigned according to single crystal X-ray structural determination performed on C8 (which is derived from C6, see below).

C6-yield: 38.0g, 84.8 mmol, 50%. Retention time 3.64 min [ column: 5 mu m of Chiral Technologies Chiral pak AD-H, 4.6 x 250 mm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol containing 0.2% 1-aminopropan-2-ol; gradient: 5% B, 1.00 min, then 5% to 60% B over 8.00 min; flow rate: 3.0 mL/min; back pressure: 120 bar ].

C7-yield: 36.8g, 82.2 mmol, 49%. Retention time 4.19 minutes (analytical SFC conditions were the same as those used for C6).

Step 2: 4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Synthesis of pyridine, P-toluenesulfonate (P3)

A solution of C7(1.62g, 3.62 mmol) in ethyl acetate (36 mL) was treated with p-toluenesulfonic acid monohydrate (791 mg, 4.16 mmol) 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, 2 × 15 mL) to give P3 as a white solid. Yield: 1.37g, 2.63 mmol, 73%. LCMSm/z 348.1♦ [M+H]+1H NMR (400 MHz, DMSO-d 6) δ 8.53 (v br s, 1H), 8.29 (v br s, 1H), 7.65 - 7.55 (m, 2H), 7.47 (d, J = 8.1 Hz, 2H), 7.35 (dd, J = 8.4, 2.0 Hz, 1H), 7.11 (d, J = 7.8 Hz, 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- [ (2) for determination of absolute stereochemistryR) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzene And dioxol-4-yl ]Piperidine, mesylate (C8)

P-toluenesulfonic acid (377 mg, 2.19 mmol) was added to a solution of C6(490 mg, 1.09 mmol) in ethyl acetate (5.5 mL) and the reaction mixture was stirred at room temperature overnight. After dilution with additional ethyl acetate, the reaction mixture was washed successively with aqueous sodium bicarbonate, water, and saturated aqueous sodium chloride, dried over sodium sulfate, filtered, and concentrated in vacuo. Yield: 375 mg, 1.08 mmol, 99%.1H NMR (400 MHz, methanol-d 4) δ 7.59 (dd, J = 8.3. 8.3 Hz, 1H), 7.27 (dd, J = 10.9, 2.0 Hz, 1H), 7.20 (br dd, J = 8.4, 2.1 Hz, 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 = 0.7 Hz, 3H), 1.85 - 1.73 (m, 4H)。

Preparation of the free base (4- [ (2)R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine) in 0.1M ethyl acetate and subjected to salt screening. Only the formation of the mesylate salt is described herein. A mixture of methanesulfonic acid (25. mu.L, 39. mu. mol) and substrate solution (0.1M; 0.25 mL, 25. mu. mol) was stirred overnight. Then, sufficient methanol was added to dissolve the solids present, and ethyl acetate (3 mL) was added. The resulting solution was slowly evaporated without stirring to give crystals of C8, one of which was used for single crystal X-ray texture 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 consisted of both omega and phi scans.

By means of internal phasing (inter phasing), using a SHELX software suite, in an orthorhombic space groupP212121The structure is analyzed. The structure is then refined by full matrix least squares. All non-hydrogen atoms were found and refined using anisotropic displacement parameters (anisotropic displacement parameters).

Formation of the mesylate salt was confirmed via N1_ H1X _ O4 proton transfer.

The hydrogen atoms located on nitrogen and oxygen are found from Fourier difference maps (Fourier difference maps) and refined at a limited distance. The remaining hydrogen atoms are placed in the calculated positions and are carried on their carrier atoms. The final refinement includes the isotropic displacement parameters of all hydrogen atoms.

Absolute structural analysis using the likelihood method (Hooft, 2008) was performed using platon (spek). The results indicate that the absolute structure has been correctly assigned; the method calculates the probability of the structure being correct to be 100%. The Hootf parameter is reported as 0.02, esd as 0.0012, and the Parson parameter is reported as 0.07, esd as 0.009. The absolute configuration of C7 is identified as (R)。

The asymmetric unit is composed of one molecule of the C8 protonated free base and one molecule of the deprotonated methanesulfonic acid. The final R index was 4.6%. The final differential fourier shows the electron density without missing or misplacement.

Relevant crystal, data collection, and refinement information are summarized in table a. The atomic coordinates, bond lengths, bond angles, and displacement parameters are listed in tables B-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. Macroe, P.R. Edington, P.McCabe, E.Pidcock, G.P. Shields, R.Taylor, M.Towler and van de street,J. Appl. Cryst. 2006, 39, 453-457.

OLEX2, O.V. Dolomanov, L.J. Bourhis, R.J. Gildea, J.A.K. Howard and Puschmann,J. Appl. Cryst. 2009, 42, 339-341.

R.W.W. Hooft, L.H. Straver and L.Spek,J. Appl. Cryst. 2008, 41, 96-103.

H. D. Flack, Acta Cryst. 1983, A39, 867-881。

table a: crystal data and structure refinement of C8

Table B: atomic coordinate of C8 (x 10)4) And an equivalent isotropic displacement parameter (A)2 x 103). U (eq) is defined as orthogonal UijOne third of the trace amount of tensor.

Table C: the bond length [ A ] and angle [ ] of C8.

For generating a symmetric transformation of equivalent atoms.

Table D: anisotropy displacement parameter (A) of C82 x 103). The anisotropy displacement factor index takes the following form:

-2π2[h2 a*2U11 + ... + 2 h k a* b* U12 ]。

preparation of P3, di-P-toluoyl-L-tartrate salt

4-[(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine, II P-toluoyl-L-tartrate (P3, di-P-toluoyl-L-tartrate)

A solution of C13, free base (519 mg, 1.49 mmol) and di-p-toluoyl-L-tartaric acid (278 mg, 0.719 mmol) in acetonitrile (7.5 mL) 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 afford P3, di-P-toluoyl-L-tartrate (217 mg, 0.296 mmol, 20%, 82% ee) as a white solid.

A solution of P3, di-P-toluoyl-L-tartrate (217 mg, 0.296 mmol, 20%, 82% ee) in acetonitrile (8.0 mL) was cooled to room temperature at 0.2 deg.C/min at 50 deg.C. After 15 h, the solid was collected by filtration and dried in a vacuum oven under nitrogen at 35 ℃ to give P3, di-P-toluoyl-L-tartrate (190 mg, 0.259 mmol, 88%, 88% ee) as a white solid. LCMSm/z 348.1♦ [M+H]+1H NMR (400 MHz, DMSO-d 6) δ 8.9 - 8.5 (br s, 2H), 7.79 (d, J = 8.1 Hz, 4H), 7.64 - 7.54 (m, 2H), 7.34 (dd, J = 8.4, 2.1 Hz, 1H), 7.26 (d, J = 8.0 Hz, 4H), 6.87 - 6.78 (m, 2H), 6.69 (dd, J= 6.7, 2.5 Hz, 1H), 5.58 (s, 2H), 3.37-3.28 (m, 2H, assumed; partially obscured 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) and peak 2 (5.31 min, major) { column: chiralpak IC-U3.0 x 50 mm, 1.6 μm; mobile phase A: carbon dioxide; mobile phase B: 0.1% isopropylamine in methanol; gradient: 10% B, 5.00 min; then 45% B, 0.6 min; 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)

P2(2.00g, 4.46 mmol), zinc cyanide (734 mg, 6.25 mol), zinc (70.1 mg, 1.07 mmol), 1' -bis (diphenylphosphino) ferrocene (dppf; 198 mg, 0.357 mmol) and tris (dibenzylideneacetone) dipalladium (0) (164 mg, 0.179 mmol) were added at 120 ℃ to N,NThe suspension in dimethylacetamide (20 mL) was stirred for 16 hours whereupon it was filtered. The filtrate was mixed with water (50 mL) and extracted with ethyl acetate (3 × 50 mL); the combined organic layers were then washed successively with water (30 mL) and saturated aqueous sodium chloride (20 mL) and concentrated in vacuo. Silica gel chromatography (gradient: 0% to 30% ethyl acetate in petroleum ether) gave a solid, which was treated with acetonitrile (15 mL) and water (15 mL) and subjected to freeze-drying. This gave P4 as a pale yellow solid. Yield: 1.17g, 2.67 mmol, 60%. LCMSm/z 461.3 [M+Na+]。1H NMR (400 MHz, chloroform-d) 7.71 (dd, J = 7.7, 7.6 Hz, 1H), 7.45 (dd, J = 8.0, 1.6 Hz, 1H), 7.42 (dd, J= 10.0, 1.5 Hz, 1H), 6.79 (dd, components of ABC mode,J= 7.7, 7.6 Hz, 1H), 6.72 (dd, components of ABC mode,J= 7.8, 1.3 Hz, 1H), 6.68 (dd, components of ABC mode,J = 7.8, 1.3 Hz, 1H), 4.37 - 4.14 (br m, 2H), 2.91 - 2.73 (m, 3H), 2.07 (d, J = 1.1 Hz, 3H), 1.89 - 1.62 (m, 4H), 1.49 (s, 9H)。

preparation of P5 and P6

4-bromo-2-phenyl-1, 3-benzodioxole, ENT-1 (P5)And4-bromo-2-phenyl-1, 3-benzo-meta-benzene Dioxoles, ENT-2 (P6)

Step 1: 2-bromo-6- [ hydroxy (phenyl) methyl]Synthesis of phenol (C9)

Phenyllithium (1.9M solution in 1-butoxybutane; 78.5 mL, 149 mmol) was slowly added to a-70 ℃ solution of 3-bromo-2-hydroxybenzaldehyde (10.0g, 49.7 mmol) in tetrahydrofuran (70 mL) at a rate that the reaction temperature was maintained below-60 ℃. The resulting suspension was stirred at-70 ℃ for 1 hour and then allowed to warm to room temperature overnight whereupon it was poured into an aqueous ammonium chloride solution (30 mL) at 0 ℃. The mixture was extracted with ethyl acetate (3 × 30mL) and the combined organic layers were washed with saturated aqueous sodium chloride (30 mL), dried over sodium sulfate, filtered and concentrated in vacuo. Silica gel chromatography (gradient: 0% to 5% ethyl acetate in petroleum ether) afforded C9 as a yellow solid. Yield: 6.11g, 21.9 mmol, 44%. 1H NMR (400 MHz, chloroform-d) δ 7.45 - 7.28 (m, 6H), 7.22 - 7.18 (m, 1H), 7.06 (br d, J = 7.7 Hz, 1H), 6.77 (dd, J = 7.9, 7.8 Hz, 1H), 6.06 (br s, 1H), 2.89 (br s, 1H)。

Step 2: synthesis of 4-bromo-2-phenyl-1, 3-benzodioxole (C10)

A solution of sodium periodate (11.7g, 54.7 mmol) in water (175 mL) was added to a solution of C9(6.11g, 21.9 mmol) in methanol (370 mL). The reaction mixture was stirred at 30 ℃ for 40 hours whereupon most of the methanol was removed via vacuum concentration. The resulting mixture was extracted with dichloromethane (5 × 100 mL), and the combined organic layers were washed successively with aqueous sodium sulfite (100 mL) and saturated aqueous sodium chloride (100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (eluent: petroleum ether) afforded C10 as a colorless oil. Yield: 4.50g, 16.2 mmol, 74%. LCMSm/z278.5 (bromine isotope pattern observed) [ M + H [)]+1H NMR (400 MHz, 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, 1.4 Hz, 1H), 6.79 (dd, components of the ABX pattern,J= 7.8, 1.4 Hz, 1H), 6.75 (dd, components of the ABX pattern,J = 7.9, 7.8 Hz, 1H)。

and step 3: 4-bromo-2-phenyl-1, 3-benzodioxole, ENT-1(P5) and 4-bromo-2-phenyl-1, 3- Isolation of benzodioxole, ENT-2(P6)

SFC [ column: chiral Technologies Chiral cel OD, 10 μm; mobile phase: 3: 1 carbon dioxide/(methanol with 0.1% ammonium hydroxide) ], an enantiomer containing C10(5.00g, 18.0 mmol) was isolated. The first eluted enantiomer was designated ENT-1(P5) and the second eluted enantiomer was designated ENT-2 (P6); both were obtained as yellow oil.

Yield of P5: 2.20g, 7.94 mmol, 44%. LCMSm/z277.0 (bromine isotope patterns observed) [ M + H [)]+1H NMR (400 MHz, 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, 1.3 Hz, 1H), 6.80 (dd, components of the ABX pattern,J= 7.8, 1.4 Hz, 1H), 6.75 (dd, components of the ABX pattern,J= 7.9, 7.8 Hz, 1H). Retention time 3.28 min (column: Chiral Technologies Chiral cel OD-H, 4.6 x 150 mm, 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, 7.22 mmol, 40%. LCMSm/z276.9 (bromine isotope pattern observed) [ M + H [)]+1H NMR (400 MHz, 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, 1.4 Hz, 1H), 6.80 (dd, components of the ABX pattern,J= 7.8, 1.4 Hz, 1H), 6.75 (dd, components of the ABX pattern,J= 7.9, 7.9 Hz, 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 Butyl ester (P7)

Step 1: synthesis of 2- (4-bromo-2-methyl-1, 3-benzodioxol-2-yl) -5-chloropyridine (C11) Become into

A mixture of 5-chloro-2-ethynylpyridine (1.80g, 13.1 mmol), 3-bromobenzene-1, 2-diol (2.47g, 13.1 mmol) and triruthenium dodecacarbonyl (167 mg, 0.261 mmol) in toluene (25 mL) was degassed for 1 min and then heated at 100 ℃ for 16 h. The reaction mixture was diluted with ethyl acetate (30 mL) and filtered through a pad of celite; the filtrate was concentrated and purified using silica gel chromatography (gradient: 0% to 1% ethyl acetate in petroleum ether) under vacuum to afford C11 as a yellow oil. Yield: 1.73g, 5.30 mmol, 40%. LCMS m/z325.6 (bromine-chlorine isotope pattern observed) [ M + H]+1H NMR (400 MHz, chloroform-d) δ 8.63 (dd, J= 2.4, 0.7 Hz, 1H), 7.71 (dd, components of the ABX pattern,J= 8.4, 2.4 Hz, 1H), 7.60 (dd, components of the ABX pattern,J = 8.4, 0.7 Hz, 1H), 6.97 (dd, J= 8.0, 1.4 Hz, 1H), 6.76 (dd, components of the ABX pattern,J= 7.8, 1.4 Hz, 1H), 6.72 (dd, components of the ABX pattern,J = 8.0, 7.8 Hz, 1H), 2.10 (s, 3H)。

step 2: 4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]-3, 6-bis Hydropyridine-1 (2)H) Synthesis of tert-butyl formate (C12)

Reacting [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (388 mg, 0.530 mmol) was added to C11(1.73g, 5.30 mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2)H) -tert-butyl formate (1.64g, 5.30 mmol), and cesium carbonate (5.18g, 15.9 mmol) in suspension in 1, 4-dioxane (35 mL) and water (6 mL). The reaction mixture was stirred at 90 ℃ for 4 hoursIt was diluted with ethyl acetate (30 mL) 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 give C12 as a yellow gum. Yield: 1.85g, 4.31 mmol, 81%. LCMS m/z 451.0♦ [M+Na+]。1H NMR (400 MHz, chloroform-d) δ 8.62 (dd, J= 2.5, 0.8 Hz, 1H), 7.69 (dd, components of the ABX pattern,J= 8.4, 2.4 Hz, 1H), 7.57 (dd, components of the ABX pattern,J = 8.4, 0.8 Hz, 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)。

and step 3: 4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine-1- Synthesis of tert-butyl formate (P7)

A solution of C12(2.61g, 6.08 mmol) and tris (triphenylphosphine) rhodium (I) chloride (Wilkinson's catalyst; 563mg, 0.608 mmol) in methanol (100 mL) was degassed under vacuum and then purged with hydrogen; this vacuum-purge cycle was performed a total of three times. The reaction mixture was then stirred under hydrogen (50 psi) at 60 ℃ 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 similarly hydrogenated material from C12(110 mg, 0.256 mmol) to afford P7 as a light yellow gum. And (4) merging yield: 2.05g, 4.76 mmol, 75%. LCMSm/z431.3♦ [M+H]+1H NMR (400 MHz, chloroform-d) δ 8.62 (d, J= 2.3 Hz, 1H), 7.69 (dd, components of the ABX pattern,J= 8.4, 2.4 Hz, 1H), 7.57 (d, half of the AB quartet,J= 8.4 Hz, 1H), 6.79 (dd, components of ABC mode, J= 7.8, 7.7 Hz, 1H), 6.72 (dd, components of ABC mode,J= 7.8, 1.3 Hz, 1H), 6.68 (br d, component of ABC mode,J = 7.9 Hz, 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 of P8 and P9

4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine-1-carboxylic acid tert-butyl ester Butyl esters, ENT-1(P8) and 4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridine-1-carboxylic acid tert-butyl ester, ENT-2(P9)

SFC { column: phenomenex Lux Amylose-1, 5 μm; mobile phase: 9: 1 carbon dioxide/[ 2-propanol containing 0.2% (7M ammonia in methanol) ] }, separation of P7(500 mg, 1.16 mmol) into its component enantiomers was performed. The first eluted enantiomer was designated ENT-1(P8) and the second eluted enantiomer was designated ENT-2 (P9).

Yield of P8: 228 mg, 0.529 mmol, 46%. Retention time 4.00 minutes { column: phenomenex Lux Amylose-1, 4.6 x 250 mm, 5 μm; mobile phase A: carbon dioxide; mobile phase B: [ 2-propanol containing 0.2% (7M ammonia in methanol) ]; gradient: 5% B, 1.00 min, then 5% to 60% B over 8.00 min; flow rate: 3.0 mL/min; back pressure: 120 bar }.

Yield of P9: 229 mg, 0.531 mmol, 46%. Retention time 4.50 minutes (analytical conditions identical to those for P8).

Preparation P10

{4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidin-1-yl } ethano Acid (P10)

Step 1: 4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]A piperidine compound, synthesis of p-toluenesulfonate (C13)Become into

A solution of P2(5.0g, 11 mmol) and P-toluenesulfonic acid (4.81g, 27.9 mmol) in ethyl acetate (100 mL) was stirred at 60 ℃ for 2 hours whereupon it was concentrated in vacuo to give C13 as a yellow gum. This material was used directly in the next step. LCMSm/z 347.9♦ [M+H]+

Step 2: {4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine- Synthesis of ethyl 1-yl } acetate (C14)

Potassium carbonate (7.71g, 55.8 mmol) and ethyl bromoacetate (1.86g, 11.2 mmol) were added to a solution of C13 (from the previous step;. ltoreq.11 mmol) in acetonitrile (150 mL) and the reaction mixture was stirred at 55 ℃ for 16 h. Then, it was filtered, and the filtrate was 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. Warp beam1This material was not completely pure by H NMR analysis. Yield: 3.57g, 8.23 mmol, 75% (over two steps). 1H NMR (400 MHz, chloroform-d) Only C14 peak: δ 7.52 (dd,J= 8.4, 8.0 Hz, 1H), 7.17-7.07 (m, 2H), 6.77 (dd, component of ABC mode,J = 7.8, 7.8 Hz, 1H), 6.72 - 6.67 (m, 2H), 4.21 (q, J = 7.1 Hz, 2H), 3.27 (s, 2H), 3.07 (m, 2H), 2.70 (tt, J = 12.1, 3.8 Hz, 1H), 2.35 (ddd, J = 11.5, 11.5, 2.7 Hz, 2H), 2.04 (d, J = 1.1 Hz, 3H), 2.02 - 1.76 (m, 4H), 1.29 (t, J= 7.1 Hz, 3H)。

and step 3: {4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine- Synthesis of 1-yl } acetic acid (P10)

A solution of C14(3.57g, 8.23 mmol) and aqueous sodium hydroxide (3M; 13.7 mL, 41.1 mmol) in a mixture of methanol (80 mL) and tetrahydrofuran (40 mL) was stirred at 25 ℃ for 16 h. After removal of the solvent in vacuo, the aqueous residue was acidified to pH 7 by addition of 1M hydrochloric acid and then washed with dichloromethane and methanolThe mixture (10: 1, 2X 100 mL) was extracted. The combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure to provide P10 as a yellow solid. Yield: 2.95g, 7.27 mmol, 88%. LCMSm/z 406.2♦ [M+H]+1H NMR (400 MHz, methanol-d 4) δ 7.61 (dd, J = 8.3, 8.3 Hz, 1H), 7.29 (dd, J = 10.9, 2.0 Hz, 1H), 7.22 (ddd, J= 8.4, 2.0, 0.8 Hz, 1H), 6.82 (dd, components of ABC mode,J = 8.3, 7.1 Hz, 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 = 1.1 Hz, 3H)。

preparation P11

2- (chloromethyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P11)

Step 1: 3- [ (2-methoxyethyl) amino group]Synthesis of methyl-4-nitrobenzoate (C15)

Triethylamine (40.7g, 402 mmol, 55.8 mL) was added to a colorless solution of methyl 3-fluoro-4-nitrobenzoate (50g, 250 mmol) in tetrahydrofuran (400 mL), followed by dropwise addition of 2-methoxyethylamine (30.2g, 402 mmol) in tetrahydrofuran (100 mL) 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 (800 mL) and washed with saturated aqueous ammonium chloride (250 mL). The aqueous phase was separated and extracted with ethyl acetate (200 mL). The combined organic layers were washed with saturated aqueous sodium chloride (3 × 250 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give C15(60.2g, 94%) as a yellow solid. 1H NMR (600 MHz, 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-amino-3- [ (2-methoxyethyl) amino group]Synthesis of methyl benzoate (C16)

Pd/C (10g, 94 mmol) was added to a solution of C15(30g, 118 mmol) in methanol (500 mL). The reaction was stirred at room temperature under 15 psi 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 (400 MHz, 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]+

And step 3: 2- (chloromethyl) -1- (2-methoxyethyl) -1HSynthesis of methyl (P11) benzimidazole-6-carboxylate

2-chloro-1, 1, 1-trimethoxyethane (3.31 mL, 24.6 mmol) and p-toluenesulfonic acid monohydrate (84.8 mg, 0.446 mmol) were added sequentially to a solution of C16(5.00g, 22.3 mmol) in tetrahydrofuran (100 mL). The reaction mixture was heated at 45 ℃ for 5 hours whereupon it was concentrated in vacuo; the residual oil was dissolved in ethyl acetate (10 mL) and heated until a solution formed. It was stirred slowly while cooling to room temperature overnight. The precipitate was collected via filtration and washed with heptane to give P11 as a grey solid. Yield: 5.73g, 20.3 mmol, 91%. 1H NMR (600 MHz, chloroform-d) δ 8.12 (br s, 1H), 8.01 (br d, J = 8.6 Hz, 1H), 7.79 (d, J = 8.4 Hz, 1H), 4.96 (s, 2H), 4.52 (t, J = 5.1 Hz, 2H), 3.96 (s, 3H), 3.74 (t, J = 5.1 Hz, 2H), 3.28 (s, 3H)。

And 4, step 4: 2- (chloromethyl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, hydrochloride (P11, HCl salt) synthesis

A solution of C16(5.0g, 24 mmol) in 1, 4-dioxane (100 mL) was heated to 100 ℃ via addition funnel over a 10 hour period, addingA solution of chloroacetic anhydride (4.1g, 24.5 mmol) in 1, 4-dioxane (60 mL) was added and the reaction mixture was stirred at 100 ℃ for another 12 hours. The next day, the reaction was cooled to room temperature and under reduced pressure, the 1, 4-dioxane was removed. 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 solution of 4M hydrogen chloride in 1, 4-dioxane (1.1 eq) was added to the ethyl acetate solution with constant stirring. The hydrochloride salt of P11 precipitated as a pale yellow solid. The suspension was stirred for 1 hour, and then the hydrochloride salt of P11 was collected by filtration to give a yellow solid (6.1g, 86%).1H NMR (600 MHz, CD3OD) δ 8.64 (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/z283.2 [M+H]+

Preparation P12

1- (2-methoxyethyl) -2- (piperazin-1-ylmethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P12)

Step 1: 2- { [4- (tert-Butoxycarbonyl) piperazin-1-yl ]Methyl } -1- (2-methoxyethyl) -1H-benzimidazole Synthesis of oxazole-6-carboxylic acid methyl ester (C17)

Compound P11(1.59g, 5.62 mmol) was added to a 15 ℃ mixture of piperazine-1-carboxylic acid tert-butyl ester (1.00g, 5.37 mmol) and potassium carbonate (2.97g, 21.5 mmol) in acetonitrile (15 mL) and the reaction mixture was stirred at 55 ℃ for 12 hours. Then, it was combined with a similar reaction using P11 and piperazine-1-carboxylic acid tert-butyl ester (200 mg, 1.07 mmol), and the mixture was filtered. After concentration of the filtrate in vacuo, chromatography on silica gel (ladder)Degree: 0% to 60% ethyl acetate in petroleum ether), the residue was purified to provide C17 as a light yellow solid. And (4) merging yield: 2.30g, 5.32 mmol, 83%. LCMSm/z 433.0 [M+H]+1H NMR (400 MHz, chloroform-d) δ8.12 (d, J = 1.5 Hz, 1H), 7.96 (dd, J = 8.4, 1.5 Hz, 1H), 7.73 (d, J = 8.5 Hz, 1H), 4.58 (t, J= 5.4 Hz, 2H), 3.95 (s, 3H), 3.89 (s, 2H), 3.73 (t, J = 5.4 Hz, 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- (2-methoxyethyl) -2- (piperazin-1-ylmethyl) -1H-benzimidazole-6-carboxylic acid methyl ester (P12) Synthesis of (2)

A solution of hydrogen chloride in ethyl acetate (20 mL) was added to a solution of C17(2.30g, 5.32 mmol) in dichloromethane (80 mL). The reaction mixture was stirred at 20 ℃ for 2 hours whereupon it was concentrated in vacuo. The residue was diluted with water (20 mL), the pH adjusted to 9-10 by addition of saturated aqueous sodium bicarbonate, 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, 5.05 mmol, 95%. LCMS m/z 332.8 [M+H]+1H NMR (400 MHz, chloroform-d)δ8.13 (br s, 1H), 7.96 (br d, J = 8.5 Hz, 1H), 7.72 (d, J = 8.5 Hz, 1H), 4.59 (t, J = 5.5 Hz, 2H), 3.95 (s, 3H), 3.86 (s, 2H), 3.75 (t, J = 5.5 Hz, 2H), 3.29 (s, 3H), 2.87 (t, J = 4.8 Hz, 4H), 2.50 (br m, 4H)。

Preparation P13

6-bromo-2- (chloromethyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]Pyridine (P13)

Step 1: 5-bromo-N- (2-methoxyethyl) -2-nitropyridin-3-amine(C18) Synthesis of (2)

A solution of 5-bromo-3-fluoro-2-nitropyridine (400 mg, 1.81 mmol) and 2-methoxyethylamine (408 mg, 5.43 mmol) in tetrahydrofuran (10 mL) was stirred at 25 ℃ for 2 hours whereupon it was diluted with ethyl acetate (100 mL) and washed with water (50 mL). The organic layer was washed with saturated aqueous sodium chloride (50 mL), dried over magnesium sulfate, filtered and concentrated to give C18 as a yellow solid. Yield: 430 mg, 1.56 mmol, 86%.

Step 2: 5-bromo-N 3 Synthesis of- (2-methoxyethyl) pyridine-2, 3-diamine (C19)

A solution of C18(430 mg, 1.56 mmol), ammonium chloride (833 mg, 15.6 mmol), and iron powder (870 mg, 15.6 mmol) in a mixture of methanol (10 mL) and water (2 mL) was stirred at 80 ℃ for 30 minutes. The resulting suspension was poured into water (50 mL) and extracted with ethyl acetate (2 × 50 mL); the combined organic layers were dried over magnesium sulfate, filtered and concentrated to provide C19 as a brown solid. Yield: 350 mg, 1.42 mmol, 91%. 1H NMR (400 MHz, chloroform-d)δ7.63 (d, J = 2.1 Hz, 1H), 6.88 (d, J = 2.0 Hz, 1H), 4.33 - 4.19 (br s, 2H), 3.65 (dd, J = 5.6, 4.6 Hz, 2H), 3.40 (s, 3H), 3.22 (br t, J = 5 Hz, 2H)。

And step 3: 6-bromo-2- (chloromethyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]Synthesis of pyridine (P13) Become into

A solution of C19(400 mg, 1.63 mmol) in 1, 4-dioxane (8 mL) was treated with chloroacetyl chloride (0.284 mL, 3.57 mmol) 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 (8 mL) 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 (50 mL) and neutralized by the addition of saturated aqueous sodium bicarbonate. The aqueous layer was extracted with ethyl acetate (20 mL), and the combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. Silica gel chromatography (gradient: 0% to80% ethyl acetate in heptane)) gave P13 as a solid. Yield: 176 mg, 0.578 mmol, 35%. LCMSm/z306.1 (bromine-chlorine isotope pattern observed) [ M + H]+1H NMR (600 MHz, chloroform-d)δ8.58 (br s, 1H), 7.89 (br s, 1H), 4.92 (s, 2H), 4.44 (t, J = 5.0 Hz, 2H), 3.71 (t, J = 5.0 Hz, 2H), 3.28 (s, 3H)。

Preparation P14

2- { [4- (2, 3-dihydroxyphenyl) piperidin-1-yl]Methyl } -1- (2-methoxyethyl) -1H-benzimidazole-6- Methyl formate (P14)

Step 1: [ (3-bromobenzene-1, 2-diyl) bis (oxymetanediyloxyethane-2, 1-diyl)]Bis (trimethyl methyl) Synthesis of silane) (C20)

The reaction was carried out in two batches on the same scale. Will be provided withN,NDiisopropylethylamine (37.8 mL, 217 mmol) was added dropwise to a solution of 3-bromobenzene-1, 2-diol (10.0g, 52.9 mmol) in tetrahydrofuran (300 mL). After the mixture was stirred at 20 ℃ for 10 minutes, over 5 minutes, [2- (chloromethoxy) ethyl group was added dropwise](trimethyl) silane (19.2 mL, 108 mmol) and stirring was continued at room temperature (18 ℃ C.) for 16 h. Is added againN,N-diisopropylethylamine (27.6 mL, 158 mmol), followed by dropwise addition of [2- (chloromethoxy) ethyl at room temperature (18 deg.C.)](trimethyl) silane (14.0 mL, 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. Warp beam1H NMR analysis of this productThe quality is not completely pure. And (4) merging yield: 22.9g, 50.9 mmol, 48%. 1H NMR (400 MHz, chloroform-d) Only C20 peak: delta 7.19 (dd,J = 8.1, 1.5 Hz, 1H), 7.12 (dd, J = 8.3, 1.4 Hz, 1H), 6.90 (dd, J = 8.2. 8.2 Hz, 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- (2, 3-bis { [2- (trimethylsilyl) ethoxy group]Methoxy } phenyl) -3, 6-dihydropyridine-1 (2H) Synthesis of tert-butyl formate (C21)

Containing C20(6.11 g, 13.6 mmol), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2)H) Tert-butyl formate (5.04g, 16.3 mmol), aqueous sodium carbonate solution (1M; 40.8 mL, 40.8 mmol), and [1, 1' -bis (diphenylphosphino) ferrocene]The reaction vessel for the suspension of palladium (II) dichloride (497 mg, 0.679 mmol) in 1, 4-dioxane (100 mL) 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 (40 mL) and extracted with ethyl acetate (3 × 150 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. Purification was performed via silica gel chromatography (gradient: 0% to 8% methanol in dichloromethane)) to afford C21 as a yellow oil. Yield: 5.47g, 9.91 mmol, 73%.1H NMR (600 MHz, chloroform-d)δ7.10 (br d, J = 8.2 Hz, 1H), 6.98 (dd, J = 7.9, 7.9 Hz, 1H), 6.81 (br d, J = 7.7 Hz, 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), 0.01 (s, 9H)。

And step 3: 4- (2, 3-bis { [2- (trimethylsilyl) ethoxy group]Methoxy } phenyl) piperidine-1-carboxylic acid tert-butyl ester Synthesis of butyl ester (C22)

A solution of C21(12.5g, 22.6 mmol) in methanol (300 mL) was treated with 10% palladium on charcoal (2.94g, 2.76 mmol) and at 4%Hydrogenation at 0psi and 25 ℃ for 16 hours. LCMS analysis at this point indicated conversion to product: LCMSm/z 576.0 [M+Na+]. After filtering the reaction mixture and washing the filter cake with methanol (2 × 100 mL), the combined filtrates were concentrated in vacuo to give C22 as a colorless oil. Yield: 11.2g, 20.1 mmol, 89%.1H NMR (400 MHz, chloroform-d)δ7.05 - 6.97 (m, 2H), 6.83 (dd, J = 6.9, 2.5 Hz, 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, 3.5 Hz, 1H), 2.79 (br dd, J = 12.8, 12.8 Hz, 2H), 1.78 (br d, J = 13 Hz, 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)。

And 4, step 4: 4- (2, 3-bis { [2- (trimethylsilyl) ethoxy group]Synthesis of methoxy } phenyl) piperidine (C23) Become into

2, 6-lutidine (2.39g, 22.3 mmol) was added to a room temperature (15 deg.C) solution of C22(7.23g, 13.0 mmol) in dichloromethane (90 mL), followed by dropwise addition of trimethylsilyl trifluoromethanesulfonate (3.80g, 17.1 mmol). The reaction mixture was stirred for 16 h at 15 ℃ whereupon additional 2, 6-lutidine (909 mg, 8.48 mmol) and trimethylsilyl trifluoromethanesulfonate (1.45g, 6.52 mmol) 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: LCMSm/z 454.1 [M+H]+. The reaction mixture was concentrated in vacuo, and the residue was washed successively with aqueous ammonium chloride (3 × 100 mL) and saturated aqueous sodium chloride (100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give C23(6.6 g) as a brown oil. This material was used directly in the next step.

And 5: 2- { [4- (2, 3-bis { [2- (trimethylsilyl) ethoxy group]Methoxy } phenyl) piperidin-1-yl] Methyl } -1- (2-methoxyethyl) -1HSynthesis of methyl (C24) benzimidazole-6-carboxylate

P11(3.08g, 10.9 mmol) was added to C23 (from previous step; 6.6g,. ltoreq.13)mmol) in acetonitrile (150 mL), then potassium carbonate (10.1g, 73.1 mmol) is added and the reaction mixture is stirred at room temperature (15 deg.C) for 16 hours. LCMS analysis at this point indicated the presence of product: LCMSm/z 700.2 [M+H]+. The reaction mixture was filtered and the filtrate was concentrated in vacuo; purification was performed via silica gel chromatography (gradient: 34% to 56% ethyl acetate in petroleum ether) to give C24 as a yellow oil. Yield: 5.4g, 7.7 mmol, 59% (via two steps).1H NMR (400 MHz, chloroform-d)δ8.16 - 8.12 (m, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.73 (d, J = 8.5 Hz, 1H), 7.04 - 6.96 (m, 2H), 6.86 (dd, J = 6.7, 2.6 Hz, 1H), 5.21 (s, 2H), 5.12 (s, 2H), 4.63 (t, J = 5.5 Hz, 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= 11.2 Hz, 2H), 2.35-2.22 (m, 2H), 1.81 (br d, half of AB quartet,J = 12.6 Hz, 2H), 1.75 - 1.61 (m, 2H), 1.04 - 0.91 (m, 4H), 0.05 (s, 9H), -0.01 (s, 9H)。

step 6: 2- { [4- (2, 3-dihydroxyphenyl) piperidin-1-yl]Methyl } -1- (2-methoxyethyl) -1H-benzo (b) Synthesis of imidazole-6-carboxylic acid methyl ester (P14)

A solution of hydrogen chloride in 1, 4-dioxane (4M; 96 mL, 384 mmol) was added to a room temperature (18 ℃) solution of C24(6.40g, 9.14 mmol) 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, 1.43 mmol) and concentrated in vacuo. The residue was treated with a mixture of dichloromethane and methanol (20: 1, 150 mL) and stirred at room temperature (18 ℃) for 1 hour, whereupon a solid (4.85 g) was collected via filtration. This material was treated with water (100 mL) and the mixture was adjusted to pH 7-8 by the addition of aqueous sodium bicarbonate, stirred at room temperature (18 ℃) for 30 minutes, and filtered. The filter cake was washed with water (2 × 20 mL), then mixed with methanol (100 mL) and concentrated in vacuo. The resulting material was treated with petroleum ether (100 mL) and stirred at room temperature (18 ℃ C.) for 30 minutes. After filtration, the filter cake is taken up in toluene (30) mL) were mixed and concentrated in vacuo to afford P14 as a gray solid. And (4) merging yield: 2.92g, 6.64 mmol, 63%. LCMSm/z 440.1 [M+H]+1H NMR (400 MHz, DMSO-d 6)δ8.21 (d, J = 1.6 Hz, 1H), 7.81 (dd, J = 8.5, 1.6 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 6.64 - 6.51 (m, 3H), 4.63 (t, J = 5.3 Hz, 2H), 3.88 (s, 3H), 3.84 (s, 2H), 3.75 (t, J = 5.3 Hz, 2H), 3.22 (s, 3H), 2.97 - 2.78 (m, 3H), 2.18 (br dd, J = 11, 11 Hz, 2H), 1.75 - 1.64 (m, 2H), 1.64 - 1.49 (m, 2H)。

Preparation P15

2- (chloromethyl) -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid methyl ester (P15)

This entire sequence is carried out on a large scale. Generally, the reactor is evacuated to-0.08 to-0.05 MPa before the reaction and after the addition of the reagents, and then filled to normal pressure with nitrogen. This process is typically repeated three times and then the oxygen content is evaluated to ensure that it is ≦ 1.0%. For the course of extraction and washing of the organic layer, the mixture is typically stirred for 15 to 60 minutes and then allowed to stand for 15 to 60 minutes before the layers separate.

Step 1: (2S) -2- [ (benzyloxy) methyl group]Synthesis of oxetane (C25)

The reaction was carried out in three batches on approximately the same scale. 2-methylpropan-2-ol (774.7kg) was charged to a 2000L glass lined reactor. Potassium tert-butoxide (157.3 kg, 1402 mol) was added via the solid addition funnel and the mixture was stirred for 30 minutes. Then, trimethyl sulphoxide iodide (308.2 kg, 1400 mol) was added in the same manner and the reaction mixture was heated at 55 ℃ to 65 ℃ for 2 to 3 hours whereupon (2) was added at a rate of 5 to 20 kg/hour S) -2- [ (benzyloxy) methyl group]Oxirane (92.1 kg, 561 mol). In the reaction mixtureAfter holding 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 rinsed with tert-butyl methyl ether (3 x 340 kg) and the combined filtrates were transferred to a 5000L reactor, treated with pure water (921 kg), and stirred at 15 ℃ to 30 ℃ for 15 to 30 minutes. The organic layer was then washed twice with a solution of sodium chloride (230.4 kg) in pure water (920.5 kg) and concentrated under reduced pressure (. ltoreq. -0.08 MPa) at ≦ 45 ℃. N-heptane (187 kg) was added and the resulting mixture was concentrated under reduced pressure (. ltoreq. -0.08 MPa.) at ≦ 45 ℃; the organic phase was purified using silica gel chromatography (280 kg) with sodium chloride (18.5 kg) at the top of the column. The crude was packed into a column using n-heptane (513 kg), and then eluted with a mixture of n-heptane (688.7kg) and ethyl acetate (64.4 kg). The three batches were combined to provide C25 as 85% pure pale yellow oil (189.7 kg, 906 mmol, 54%).1H NMR (400 MHz, 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, 3.7 Hz, 1H), 4.72-4.55 (m, 4H), 3.67 (dd, component of ABX pattern, J= 11.0, 4.9 Hz, 1H), 3.62 (dd, components of the ABX pattern,J = 11.0, 3.7 Hz, 1H), 2.72 - 2.53 (m, 2H)。

step 2: (2S) Synthesis of (2-oxetaneme) -methanols (C26)

10% palladium on charcoal (30.7 kg) was added via an addition funnel to a 10 ℃ -30 ℃ solution of 85% pure C25 (185.3 kg, 884.8 mol from previous step) in tetrahydrofuran (1270 kg) in a 3000L stainless steel autoclave reactor. The addition funnel was rinsed with pure water and tetrahydrofuran (143 kg), and the rinse was added to the reaction mixture. After the reactor contents were purged with nitrogen, they were similarly purged with hydrogen, the pressure was increased to 0.3 to 0.5MPa, and then vented to 0.05 MPa. The hydrogen purging was repeated five times, whereupon the hydrogen pressure was increased to 0.3 to 0.4 MPa. The reaction mixture was then heated to 35-45 ℃. After 13 hours, the mixture was vented to 0.05MPa while maintaining the hydrogen pressure at 0.3 to 0.5MPa, and purged five times with nitrogen via increasing the pressure to 0.15-0.2 MPa and then venting 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). The filter cake was soaked twice with this flush and then filtered; concentration under reduced pressure (. ltoreq. -0.06 MPa) at ≦ 40 deg.C to give C26(62.2 kg, 706 mol, 80%) in tetrahydrofuran (251 kg).

And step 3: 4-Methylbenzenesulfonic acid (2)S) Synthesis of (E) -Oxetadin-2-ylmethyl ester (C27)

4- (dimethylamino) pyridine (17.5kg, 143 mol) was added to a 10 ℃ to 25 ℃ solution of C26 (from previous step; 62.2kg, 706 mol) in tetrahydrofuran (251 kg) and triethylamine (92.7kg, 916 mol in 1240 kg dichloromethane). After 30 minutes, p-toluenesulfonyl chloride (174.8kg, 916.9 mol) was added portionwise at 20 to 40 minute intervals, 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 53.8 kg of sodium bicarbonate and 622 kg of pure water), and then washed with an aqueous ammonium chloride solution (prepared using 230 kg of ammonium chloride and 624 kg of pure water). After the final washing with pure water (311kg), the organic layer was filtered through a stainless steel Nutsche filter which had been previously loaded with silica gel (60.2 kg). The filter cake was soaked with dichloromethane (311kg) for 20 minutes and then filtered; the combined filtrates are concentrated under reduced pressure (. ltoreq. -0.05 MPa) and. ltoreq.40 ℃ until 330 to 400L remain. Tetrahydrofuran (311kg) was then added at 15-30 ℃ 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, again to a volume of 330 to 400L, to give a pale yellow solution of C27 (167.6 kg, 692 mmol, 98%) in tetrahydrofuran (251.8 kg). 1H NMR (400 MHz, chloroform-d) Only C27 peak: delta 7.81 (d,J = 8.4 Hz, 2H), 7.34 (d, J = 8.1 Hz, 2H), 4.91 (ddt, J = 8.0, 6.7, 3.9 Hz, 1H), 4.62 - 4.55 (m, 1H), 4.53 - 4.45 (m, 1H), 4.14 (d, J = 3.9 Hz, 2H), 2.75 - 2.63 (m, 1H), 2.60 - 2.49 (m, 1H), 2.44 (s, 3H)。

and 4, step 4: (2S) Synthesis of (E) -2- (azidomethyl) oxetane (C28)

At 10 to 25 ℃ ofN,NDimethylformamide (473 kg), sodium azide (34.7kg, 534 mol), and potassium iodide (5.2kg, 31 mol) were combined in a 3000L glass-lined reactor. After addition of C27(83.5kg, 344.6 mol in 125.4 kg of tetrahydrofuran), the reaction mixture was heated to 55 ℃ to 65 ℃ for 17 hours 40 minutes whereupon it was cooled to 25-35 ℃ and nitrogen was bubbled through the bottom valve for 15 minutes. Then, t-butyl methyl ether (623 kg) and pure water (840 kg) were added, and the resulting aqueous layer was extracted twice with t-butyl methyl ether (312 kg and 294 kg). The combined organic layers were washed with pure water (2 × 419 kg) while maintaining the temperature at 10 ℃ to 25 ℃ to obtain C28(31.2 kg, 276 mol, 80%) in the aforementioned organic layer solution (1236.8 kg).

And 5: 1- [(2S) -oxetan-2-yl]Synthesis of methylamine (C29)

10% Palladium on charcoal (3.7 kg) was added via addition funnel to C28[ from previous step, 1264kg (31.1 kg of C28, 275 mol.) in a 3000L stainless steel autoclave reactor]Solution in tetrahydrofuran (328 kg) at 10 ℃ to 30 ℃. The addition funnel was rinsed with tetrahydrofuran (32 kg) and the rinse was added to the reaction mixture. After the reactor contents were purged with nitrogen, they were similarly purged with hydrogen, the pressure was increased to 0.05 to 0.15MPa, and then vented to 0.03-0.04 MPa. This hydrogen purging was repeated five times, whereupon the hydrogen pressure was increased to 0.05 to 0.07 MPa. The reaction temperature was increased to 25-33 ℃ and the hydrogen pressure was maintained at 0.05-0.15MPa for 22 hours while the hydrogen was changed every 3-5 hours. The mixture was then purged five times with nitrogen by increasing the pressure to 0.15-0.2 MPa and then venting to 0.05 MPa. After filtration, tetrahydrofuran (92kg and 93 kg) was used to wash the reactor, and then the filter cake was soaked. The combined filtrates were concentrated under reduced pressure (. ltoreq. -0.07MPa) and. ltoreq.45 ℃ to give C29(18.0 kg, 207 mol, 75%) in tetrahydrofuran (57.8 kg). 1H NMR (400 MHz, DMSO-d 6) Only C29 peak: delta 4.62 (ddt,J= 7.6, 6.6, 5.1 Hz, 1H), 4.49 (ddd, J = 8.6, 7.3, 5.6 Hz, 1H), 4.37 (dt, J = 9.1, 5.9 Hz, 1H), 2.69 (d, J = 5.1 Hz, 2H), 2.55 - 2.49 (m, 1H), 2.39 (m, 1H)。

step 6: 4-Nitro-3- { [ (2)S) -Oxetazedin-2-ylmethyl]Synthesis of methyl amino } benzoate (C30)

Potassium carbonate (58.1 kg, 420 mol) was added to a solution of methyl 3-fluoro-4-nitrobenzoate (54.8 kg, 275 mol) in tetrahydrofuran (148 kg) in a 100L glass-lined reactor and the mixture was stirred for 10 minutes. A solution of C29(29.3 kg, 336 mol) in tetrahydrofuran (212.9 kg) was added and the reaction mixture was stirred at 20 to 30 ℃ for 12 hours whereupon ethyl acetate (151 kg) was added and the mixture was filtered through silica gel (29 kg). The filter cake was washed with ethyl acetate (150 kg and 151 kg) and the combined filtrates were concentrated to a volume of 222-281L under reduced pressure (. ltoreq. -0.08MPa) and. ltoreq.45 ℃. After the mixture was cooled to 10 deg.C-30 deg.C, n-heptane (189 kg) was added, stirring was conducted for 20 minutes, and the mixture was concentrated to a volume of 222L under reduced pressure (. ltoreq. -0.08MPa) and. ltoreq.45 deg.C. N-heptane (181 kg) was again 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 40-45 ℃ and stirred for 1 hour, whereupon it is cooled to 15-25 ℃ at a rate of 5-10 ℃ per hour, and then stirred for 1 hour at 15-25 ℃. Filtration using a stainless steel centrifuge provided a filter cake, which was rinsed with a mixture of ethyl acetate (5.0 kg) and n-heptane (34 kg), and then stirred with tetrahydrofuran (724kg) at 10 ℃ -30 ℃ for 15 minutes; filtration afforded a yellow solid (57.3 kg, 210 mol, 76%) consisting mostly of C30. 1H NMR (400 MHz, DMSO-d 6) 8.34 (t, J = 5.8 Hz, 1H), 8.14 (d, J = 8.9 Hz, 1H), 7.63 (d, J = 1.7 Hz, 1H), 7.13 (dd, J = 8.9, 1.8 Hz, 1H), 4.99 (dddd, J = 7.7, 6.7, 5.3, 4.1 Hz, 1H), 4.55 (ddd, J = 8.6, 7.3, 5.8 Hz, 1H), 4.43 (dt, J = 9.1, 6.0 Hz, 1H), 3.87 (s, 3H), 3.67 - 3.61 (m, 2H), 2.67 (dddd, J = 11.1, 8.6, 7.7, 6.2 Hz, 1H), 2.57 - 2.47 (m, 1H)。

And 7: 2- (chloromethyl) -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid methyl ester Synthesis of (P15)

A solution of C30 (51.8 kg, 190 mol from the previous step) in tetrahydrofuran (678 kg) in a 3000L autoclave reactor was treated with 10% palladium on charcoal (5.2 kg) 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, the pressure was increased to 0.1 to 0.2MPa, and then vented to 0.02-0.05 MPa. This hydrogen purging was repeated five times, whereupon the hydrogen pressure was increased to 0.1 to 0.25 MPa. The reaction mixture was stirred at 20-30 ℃ and every 2-3 hours, the mixture was purged three times with nitrogen and then five times with hydrogen; after each final hydrogen change, the hydrogen pressure was increased to 0.1 to 0.25 MPa. After 11.25 hours total reaction time, the reaction mixture was vented to normal pressure and purged five times with nitrogen by increasing the pressure to 0.15-0.2 MPa and then venting to 0.05 MPa. Then, it was filtered and the filter cake was rinsed twice with tetrahydrofuran (64 kg and 63 kg); the combined washings and filtrates were concentrated to a volume of 128-160L at reduced pressure (. ltoreq. -0.08 MPa) and. ltoreq.40 ℃. Tetrahydrofuran (169kg) was added and the mixture was again concentrated to a volume of 128-160L; this procedure was repeated a total of four times to give intermediate 4-amino-3- { [ (2) S) -Oxetazedin-2-ylmethyl]Amino } benzoic acid methyl ester.

Tetrahydrofuran (150 kg) was added to this solution followed by 2-chloro-1, 1, 1-trimethoxyethane (35.1kg, 227 mol) 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 (142 kg) was added and the mixture was again concentrated to a volume of 135-181L whereupon 2-propanol (36.5 kg) and pure water (90 kg) were added and stirring was continued until a solution was obtained. Using an in-line liquid filterIt was filtered and then treated with pure water (447 kg) at 20 to 40 ℃ at a reference rate of 150 to 400 kg/hour. After the mixture was cooled to 20-30 ℃, it was stirred for 2 hours and the solid was collected via filtration using a centrifuge. The filter cake was rinsed with a solution of 2-propanol (20.5 kg) and pure water (154 kg); after drying P15 was obtained as a white solid (32.1 kg, 109 mol, 57%).1H NMR (400 MHz, chloroform-d)δ8.14 - 8.11 (m, 1H), 8.01 (dd, J = 8.5, 1.1 Hz, 1H), 7.79 (br d, J= 8.6 Hz, 1H), 5.26-5.18 (m, 1H), 5.04 (s, 2H), 4.66-4.58 (m, 2H), 4.53 (dd, component of ABX pattern, J = 15.7, 2.7 Hz, 1H), 4.34 (dt, J = 9.1, 6.0 Hz, 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 (206 mg, 1.14 mmol) was dissolved in 1, 4-dioxane (11.5 mL) and treated with chloroacetyl chloride (109 μ L, 1.37 mmol). The mixture was stirred at 100 ℃ for 3 hours and cooled to room temperature. Triethylamine (0.8 mL, 7 mmol) and heptane (10 mL) were added and filtered. The filtrate was concentrated under reduced pressure and the crude was purified by silica gel chromatography (eluent: 40% ethyl acetate in heptane) to give 120 mg of P16 (44%).1H NMR (400 MHz, chloroform-d)δ8.14 (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 of 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 Ester, ENT-2 (P18)

Step 1: synthesis of tert-butyl 4- (2-ethoxy-2-oxoethylene) piperidine-1-carboxylate (C31)

A solution of potassium tert-butoxide (65.9g, 587 mmol) in tetrahydrofuran (500 mL) was added to a 0 ℃ solution of ethyl (diethoxyphosphoryl) acetate (132g, 589 mmol) in tetrahydrofuran (500 mL) 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, 452 mmol) in tetrahydrofuran (1.5L) is added dropwise at-50 ℃ and the reaction mixture is then slowly warmed to 20 ℃ and then stirred at 20 ℃ for 16 hours. After addition of water (1L), the mixture was concentrated in vacuo to remove tetrahydrofuran. The aqueous residue was extracted with ethyl acetate (2 × 800 mL) and the combined organic layers were washed with saturated aqueous sodium chloride solution (500 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting material was washed several times with petroleum ether (200 mL) to afford C31 as a white solid. Yield: 95.0g, 353 mmol, 78%. 1H NMR (400 MHz, chloroform-d)δ5.71 (s, 1H), 4.16 (q, J = 7.2 Hz, 2H), 3.55 - 3.43 (m, 4H), 2.94 (br t, J = 5.5 Hz, 2H), 2.28 (br t, J = 5.5 Hz, 2H), 1.47 (s, 9H), 1.28 (t, J = 7.0 Hz, 3H)。

Step 2: 6-azaspiro [2.5 ]]Octane-1, 6-dicarboxylic acid 6-Tert-butyl esterSynthesis of 1-Ethyl ester (C32)

Potassium tert-butoxide (71.2g, 634 mmol) was added in one portion to a solution of trimethyl sulphoxide iodide (140g, 636 mol) in dimethyl sulphoxide (800 mL) at 20 ℃. After stirring the reaction mixture at 20 ℃ for 1.5 hours, C31(95.0g, 353 mmol) was added dropwise to dimethyl sulfoxide(800 mL) and stirring was continued at 20 ℃ for 16 h. Then, a saturated aqueous sodium chloride solution (2.0L) was added; the resulting mixture was neutralized by the addition of ammonium chloride and extracted with ethyl acetate (3.0L). The combined organic layers were washed successively with water (2 × 1.0L) and saturated aqueous sodium chloride (2.0L), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification was performed by silica gel chromatography (eluent: 10: 1 petroleum ether/ethyl acetate) to give C32 as a yellow oil.1H NMR analysis indicated the presence of additional aliphatic species. Yield: 80g, 280 mmol, 79%.1H NMR (400 MHz, chloroform-d) Only C32 peak: δ 4.19-4.09 (m, 2H), 3.55-3.39 (m, 3H), 3.27 (ddd,J = 13.0, 7.0, 4.5 Hz, 1H), 1.76 - 1.64 (m, 2H), 1.56 (dd, J= 8.0, 5.5 Hz, 1H, assumed; partially obscured by water peaks), 1.47 (s, 9H), 1.47-1.37 (m, 2H), 1.27 (t, J = 7.0 Hz, 3H), 1.17 (dd, J = 5.0, 5.0 Hz, 1H), 0.93 (dd, J = 8.0, 4.5 Hz, 1H)。

And step 3: 6- (Tert-butoxy radicalCarbonyl) -6-azaspiro [2.5]Synthesis of octane-1-carboxylic acid (C33)

Lithium hydroxide monohydrate (37.4g, 891 mmol) was added in one portion to a mixture of C32(80g, 280 mmol) in tetrahydrofuran (500 mL) and water (500 mL). The reaction mixture was stirred at 25 ℃ for 16 h, whereupon it was diluted with water (600 mL) and washed with ethyl acetate (3 × 300 mL). The organic layer was discarded and the aqueous layer was acidified to pH 3-4 by addition of 6M hydrochloric acid. The resulting mixture was extracted with ethyl acetate (3 × 600 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was triturated with petroleum ether (300 mL) to provide C33 as a white solid. Yield: 42.0g, 164 mmol, 59%. LCMSm/z 278.2 [M+Na+]。1H NMR (400 MHz, DMSO-d 6) δ 12.15-12.03 (br s, 1H), 3.43-3.25 (m, 3H, hypothetical; partially obscured by a water peak), 3.23-3.12 (m, 1H), 1.64-1.50 (m, 2H), 1.52 (dd,J = 7.5, 5.5 Hz, 1H), 1.39 (s, 9H), 1.39 - 1.28 (m, 2H), 0.96 - 0.88 (m, 2H)。

and 4, step 4: 1- ({4- (methoxycarbonyl) -2- [ (2-methoxyethyl) amino]Phenyl } carbamoyl) -6-nitrogen Hetero spiro [2.5 ]]Synthesis of tert-butyl octane-6-carboxylate (C34)

C33(570 mg, 2.23 mmol), C16(500 mg, 2.23 mmol) andO- (7-azabenzotriazol-1-yl) - N,N,N’,N’Tetramethyluronium hexafluorophosphate (HATU; 1.27g, 3.34 mmol) inN,NThe solution in dimethylformamide (10 mL) was stirred for 30 min whereupon triethylamine (902 mg, 8.91 mmol) was added and stirring continued at 30 ℃ for 16 h. The reaction mixture was then poured into water (60 mL) and extracted with ethyl acetate (3 × 50 mL). The combined organic layers were washed with saturated aqueous sodium chloride (3 × 50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (eluent: 1: 1 petroleum ether/ethyl acetate) gave C34 as a brown oil, which was used directly in the following step.

And 5: 2- [6- (tert-Butoxycarbonyl) -6-azaspiro [2.5 ]]Oct-1-yl]-1- (2-methoxyethyl) -1H- Synthesis of methyl benzimidazole-6-carboxylate (C35)

A solution of C34 (from the previous step,. ltoreq.2.23 mmol) in acetic acid (15 mL) was stirred at 50 ℃ for 16 h whereupon it was concentrated in vacuo to afford C35 as a brown oil. This material was used directly in the next step. LCMSm/z 444.1 [M+H]+

Step 6: 2- (6-azaspiro [2.5 ]]Oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester Synthesis of ester (C36)

Trifluoroacetic acid (5 mL) was added to a solution of C35 (from the previous step;. ltoreq.2.23 mmol) in dichloromethane (10 mL) and the reaction mixture was stirred for 2 hours at 25 ℃. After removal of the solvent in vacuo, the residue was basified via addition of saturated aqueous potassium carbonate (40 mL) and extracted with a mixture of dichloromethane and methanol (10: 1, 3 × 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 give C36 as a yellow solid. Yield: 640 mg, 1.86 mmol, 83% (via three steps). LCMSm/z 344.1 [M+H]+

And 7: 2- (6-azaspiro [2.5 ]]Oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester Esters, ENT-1(P17) and 2- (6-azaspiro [2.5 ]]Oct-1-yl) -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid Isolation of methyl ester, ENT-2(P18)

SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 55: 45 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ], was performed to separate C36(630 mg, 1.83 mmol) into its component enantiomers. The first eluted peak was designated ENT-1(P17) and the second eluted enantiomer was designated ENT-2 (P18); both were isolated as a pale yellow solid.

Yield of P17: 300 mg, 0.874 mmol, 48%. LCMSm/z 344.1 [M+H]+. Retention time: 5.10 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 150 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol with 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then held at 40% B for 3.0 minutes; flow rate: 2.5 mL/min).

Yield of P18: 240 mg, 0.699 mmol, 38%. LCMSm/z 344.1 [M+H]+. Retention time: 7.35 minutes (analytical conditions identical to those used for P17).

Preparation P19

4-amino-3- { [ (1-ethyl-1)H-imidazol-5-yl) methyl]Amino } benzoic acid methyl ester (P19)

Step 1: 3- { [ (1-Ethyl-1)H-imidazol-5-yl) methyl]Synthesis of methyl amino } -4-nitrobenzoate (C37)

Triethylamine (3.65 mL, 26.2 mmol) was added to 3-fluoro-4-nitrobenzylMethyl ester acid (1.00g, 5.02 mmol) and 1- (1-ethyl-1)H-imidazol-5-yl) methylamine, dihydrochloride salt (1.00g, 5.05 mmol) in a mixture of tetrahydrofuran (12 mL) and methanol (8 mL). 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, 4.17 mmol, 83%.1H NMR (400 MHz, chloroform-d)δ8.24 (d, J = 8.8 Hz, 1H), 7.98 - 7.91 (m, 1H), 7.68 (d, J = 1.7 Hz, 1H), 7.57 (br s, 1H), 7.33 (dd, J = 8.8, 1.7 Hz, 1H), 7.11 (br s, 1H), 4.53 (d, J = 4.9 Hz, 2H), 3.99 (q, J = 7.3 Hz, 2H), 3.95 (s, 3H), 1.47 (t, J = 7.3 Hz, 3H)。

Step 2: 4-amino-3- { [ (1-ethyl-1)H-imidazol-5-yl) methyl]Synthesis of methyl amino } benzoate (P19) Become into

A wet mixture of palladium on charcoal (144 mg) and C37(412 mg, 1.35 mmol) in methanol (13 mL) 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: 340 mg, 1.24 mmol, 92%. 1H NMR (400 MHz, methanol-d 4)δ7.66 (br s, 1H), 7.38 - 7.29 (m, 2H), 6.97 (br s, 1H), 6.67 (d, J = 7.9 Hz, 1H), 4.35 (s, 2H), 4.11 (q, J = 7.3 Hz, 2H), 3.81 (s, 3H), 1.44 (t, J= 7.3 Hz, 3H)。

Preparation P20

4-amino-3- (methylamino) benzoic acid methyl ester (P20)

Step 1: synthesis of methyl 3- (methylamino) -4-nitrobenzoate (D1)

Methylamine (38.4 mL, 76.8 mmol, 2M in tetrahydrofuran) was added dropwise over 10 minutesTo a solution of methyl 3-fluoro-4-nitrobenzoate (5.10g, 25.6 mmol) in tetrahydrofuran (60 mL). Upon addition, the pale yellow solution turned dark orange immediately and was stirred at room temperature for 2 hours. Then, the reaction mixture was diluted with ether (100 mL) and the organic layer was washed successively with water (50 mL) and saturated aqueous sodium chloride solution (50 mL). 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. LCMSm/z 211.1 [M+H]+1H NMR (400 MHz, chloroform-d)δ8.22 (d, J = 8.9 Hz, 1H), 8.00 (br s, 1H), 7.56 (d, J = 1.7 Hz, 1H), 7.25 (dd, J= 8.9, 1.7 Hz, 1H, assumed; partially obscured by solvent peaks), 3.95 (s, 3H), 3.09 (d,J = 5.1 Hz, 3H)。

step 2: synthesis of methyl 4-amino-3- (methylamino) benzoate (P20)

A solution of D1(5.26g, 25.0 mmol) in ethanol (150 mL) was added to 500 mL Parr prefilled with 10% palladium on charcoal (50% water; 1 g)®In a bottle. The mixture was shaken for 1 hour at room temperature under a 50 psi hydrogen atmosphere 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 (400 MHz, chloroform-d)δ7.46 (dd, J= 8.0, 1.9 Hz, 1H), 7.34 (d, J = 1.8 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 3.87 (s, 3H), 3.72 (br s, 2H), 3.21 (br s, 1H), 2.91 (s, 3H)。

Preparation of P21 and P22

5-bromo-N 3 -methylpyridine-2, 3-diamine (P21)And5-bromo-N 3 6-dimethylpyridine-2, 3-diamine (P22)

Intermediate P21 was according to the literature procedure (Choi, J.Y, et al,J. Med. Chem. 2012, 55852-870). Intermediate P22 was synthesized using the same method.

Preparation P23

2- (chloromethyl) -1- [ (1-methyl-1)H-imidazol-5-yl) methyl]-1H-benzimidazole-6-carboxylic acid methyl ester (P23)

Step 1: 3- { [ (1-methyl-1)H-imidazol-5-yl) methyl]Synthesis of methyl amino } -4-nitrobenzoate (D2)

1- (1-methyl-1)H-imidazol-5-yl) methylamine (670 mg, 6.0 mmol) and triethylamine (762 mg, 7.53 mmol) were slowly added to methyl 3-fluoro-4-nitrobenzoate (1.0g, 5.0 mmol)N,N-in a colorless solution in dimethylformamide (10 mL). The reaction mixture was stirred at 60 ℃ for 16 h whereupon it was poured into water (30 mL) and extracted with dichloromethane (3 × 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). Using a mixed solution of 30: the resulting yellow solid was triturated with 1 petroleum ether/ethyl acetate to give D2(1.2g, 82%) as a yellow solid. LCMS m/z 290.9 [M+H]+1H NMR (400 MHz, chloroform-d)δ8.25 (d, J = 8.9 Hz, 1H), 7.98 - 7.92 (m, 1H), 7.70 (d, J = 1.7 Hz, 1H), 7.49 (s, 1H), 7.34 (dd, J = 8.9, 1.7 Hz, 1H), 7.12 (s, 1H), 4.54 (d, J = 5.0 Hz, 2H), 3.96 (s, 3H), 3.67 (s, 3H)。

Step 2: 4-amino-3- { [ (1-methyl-1)H-imidazol-5-yl) methyl]Synthesis of methyl amino } benzoate (D3)

Wet 10% palladium on charcoal (1g) was added to a suspension of D2 (5.46g, 18.8 mmol) in methanol (160 mL). The mixture was stirred at 20 ℃ under 1 atm of hydrogen for 36 hours. The reaction mixture was filtered and the filter cake was rinsed with methanol (200 mL). Under reduced pressure, mixingThe filtrate was concentrated to give D3(4.8g, 98%) as a brown solid. LCMSm/z260.9 [M+H]+1H NMR (400 MHz, DMSO-d 6)δ7.56 (s, 1H), 7.18 (br d, J = 8.1 Hz, 1H), 7.12 (br s, 1H), 6.87 (s, 1H), 6.55 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.84 (t, J = 5.2 Hz, 1H), 4.23 (d, J = 5.0 Hz, 2H), 3.73 (s, 3H), 3.63 (s, 3H)。

And step 3: 2- (hydroxymethyl) -1- [ (1-methyl-1)H-imidazol-5-yl) methyl]-1H-benzimidazole-6-carboxylic acid methyl ester Synthesis of ester (D4)

A mixture of D3(780 mg, 3.00 mmol) and 2-hydroxyacetic acid (342 mg, 4.49 mmol) in 1,3, 5-trimethylbenzene (8 mL) 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 (50 mL) and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (eluent: 20% methanol in dichloromethane) to provide D4(318 mg, 35%) as a yellow foam. LCMSm/z 300.9 [M+H]+1H NMR (400 MHz, DMSO-d 6)δ8.13 - 8.11 (m, 1H), 7.83 (dd, J = 8.4, 1.6 Hz, 1H), 7.71 (d, J = 8.5 Hz, 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)。

And 4, step 4: 2- (chloromethyl) -1- [ (1-methyl-1)H-imidazol-5-yl) methyl ]-1H-benzimidazole-6-carboxylic acid methyl ester Synthesis of (P23)

Thionyl chloride (990 mg, 0.60 mL, 8.32 mmol) was added dropwise to D4(500 mg, 1.66 mmol) in dichloromethane (10 mL) and at room temperatureN,N-suspension in dimethylformamide (3 mL). The reaction mixture was stirred at room temperature for 1 hour, then concentrated under reduced pressure. The resulting brown residue was triturated with dichloromethane (10 mL). The solid was collected by filtration and washed with dichloromethane (5 mL) to provide P23(431 mg, 73%) as an off-white solid. LCMSm/z318.9♦ [M+H]+1H NMR (400 MHz, DMSO-d 6)δ9.17 (s, 1H), 8.31 (s, 1H), 7.93 (br d, J = 8.5 Hz, 1H), 7.82 (d, J = 8.5 Hz, 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-3HImidazo [4,5-b]Pyridine (P24)

Step 1: 6-chloro-NSynthesis of (D5) methyl-3-nitropyridin-2-amine

A solution of methylamine in tetrahydrofuran (2.0M; 622 mL, 1.24 mol) was added dropwise to 2, 6-dichloro-3-nitropyridine (200g, 1.04 mol) and Na via syringe at 0 deg.C2CO3(132g, 1.24 mol) in a suspension in ethanol (1L). 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 give a yellow solid. The crude was purified by silica gel chromatography (gradient: 0% to 5% ethyl acetate in petroleum ether) to give D5(158g, 81% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d 6)δ8.72 (br s, 1H), 8.41 (d, J = 8.6 Hz, 1H), 6.76 (d, J = 8.6 Hz, 1H), 3.00 (d, J = 4.8 Hz, 3H)。

Step 2: 6-chloro-N 2 Synthesis of (D6) methylpyridine-2, 3-diamine

Iron powder (15.4g, 276 mmol) was added to a mixture of D5(15.8g, 84.2 mmol) in acetic acid (100 mL). The reaction mixture was stirred at 80 ℃ for 3 hours whereupon it was allowed to cool 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 was purified by silica gel chromatography (eluent: 1: 1 ethyl acetate/petroleum ether) to give D6 as a brown solid (8.40g, 63% yield).1H NMR (400 MHz, chloroform-d)δ6.79 (d, J = 7.7 Hz, 1H), 6.49 (d, J = 7.7 Hz, 1H), 3.00 (s, 3H)。

And step 3: 5-chloro-2- (chloromethyl) -3-methyl-3HImidazo [4,5-b]Synthesis of pyridine (P24)

Chloroacetyl chloride (55.5 mL, 698 mmol) was added to a solution of D6(50.0g, 317 mmol) in 1, 4-dioxane (1.2L) and the reaction mixture was stirred at 15 ℃ for 50 minutes. Then, it was concentrated under reduced pressure to give a brown solid, which was taken up 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 by addition of saturated aqueous sodium bicarbonate. After carbon dioxide evolution ceased, 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 was purified by silica gel chromatography (gradient: 10% to 25% ethyl acetate in petroleum ether) to give P24 as a yellow solid (61.0g, 79% yield). LCMS m/ z215.7 (dichloro isotope pattern observed) [ M + H]+1H NMR (400 MHz, DMSO-d 6)δ8.13 (d, J = 8.3 Hz, 1H), 7.37 (d, J = 8.4 Hz, 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 ]](ii) a And 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl)]Piperazin-1-yl } methyl) -1- (2-methoxy Ethyl) -1H-benzimidazole-6-carboxylic acid, ENT-X2, trifluoroacetate (2) [ from C40 ]]

Step 1: 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperazin-1-yl } Methyl) -1- (2-methoxyethyl) -1HSynthesis of methyl (C38) benzimidazole-6-carboxylate

This experiment was performed in two batches of the same scale. Comprises reacting a mixture of C2(500 mg, 1.52 mmol), P12(530 mg, 1.59 mmol) and [2 ', 6' -bis (prop-2-yloxy) biphenyl-2-yl]A reaction vessel for a mixture of (dicyclohexyl) phosphane (Ruphos; 142 mg, 0.304 mmol), tris (dibenzylideneacetone) dipalladium (0) (139 mg, 0.152 mmol), and cesium carbonate (1.48g, 4.54 mmol) in toluene (15 mL) was evacuated and charged with nitrogen. This cycle of evacuation was repeated twice, whereupon the reaction mixture was stirred at 100 ℃ for 16 hours, combined with a 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. And (4) merging yield: 600 mg, 1.03 mmol, 34%. LCMS m/z 581.0♦ [M+H]+

Step 2: 2- ({4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperazin-1-yl } Methyl) -1- (2-methoxyethyl) -1HMethyl benzimidazole-6-carboxylate, ENT-1(C39) and 2- ({4- [2- (4-chloro-2-) Fluorophenyl) -1, 3-benzodioxol-4-yl]Piperazin-1-yl } methyl) -1- (2-methoxyethyl) -1H-benzo (b) Isolation of imidazole-6-carboxylic acid methyl ester, ENT-2(C40)

SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 3: 2 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]Separation of C38(780 mg, 1.34 mmol) into its component enantiomers was performed. The first eluted enantiomer was designated ENT-1(C39) and was obtained as a white solid. Yield: 282 mg, 0.485 mmol, 36%. LCMSm/z 581.0♦ [M+H]+. Retention time 1.90 min (column: Chiral Technologies Chiralpak AD-3, 4.6 × 50 mm, 3 μm; flow)Moving phase A: carbon dioxide; mobile phase B: ethanol containing 0.05% diethylamine; gradient: 5% B, 0.20 min, then 5% to 40% B, over 1.4 min, then held at 40% B for 1.05 min; flow rate: 4.0 mL/min).

The second eluted enantiomer was designated ENT-2(C40) and 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 afforded C40 as a light brown solid. Yield: 280 mg, 0.482 mmol, 36%. LCMSm/z581.0♦ [M+H]+. Retention time 2.18 minutes (analytical conditions identical to those for C39).

And step 3: 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 ]]Is/are as follows Synthesis of

Aqueous lithium hydroxide (2M; 0.30 mL, 0.60 mmol) was added to a solution of C39(70 mg, 0.12 mmol) in a mixture of methanol (3 mL) and tetrahydrofuran (3 mL). After the reaction mixture was stirred at 25 ℃ for 16 h, an aqueous solution of lithium hydroxide (2M; 0.30 mL, 0.60 mmol) was again added and stirring continued for another 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 5-6 by 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: 40.5 mg, 59.5. mu. mol, 50%. LCMS m/z 567.0♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.37 (br s, 1H), 8.07 (dd, J = 8.5, 1.5 Hz, 1H), 7.79 (d, J = 8.6 Hz, 1H), 7.59 (dd, J = 8.0, 8.0 Hz, 1H), 7.34 (dd, J = 10.2, 2.0 Hz, 1H), 7.30 (br dd, J = 8.3, 2.0 Hz, 1H), 7.22 (s, 1H), 6.87 (dd, J = 8.1, 8.1 Hz, 1H), 6.63 (br d, J = 8 Hz, 1H), 6.60 (br d, J = 8 Hz, 1H), 4.70 (s, 2H), 4.65 (t, J = 4.8 Hz, 2H), 3.75 (t, J = 4.8 Hz, 2H), 3.59 - 3.42 (m, 8H), 3.29 (s, 3H)。

And 4, step 4: 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 ]]Is/are as follows Synthesis of

Aqueous lithium hydroxide (2M; 0.30 mL, 0.60 mmol) was added to a solution of C40(69 mg, 0.12 mmol) in a mixture of methanol (3 mL) and tetrahydrofuran (3 mL). After the reaction mixture was stirred at 25 ℃ for 16 h, an aqueous solution of lithium hydroxide (2M; 0.30 mL, 0.60 mmol) was again added and stirring continued for another 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 5-6 by 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: 22.9 mg, 33.6 μmol, 28%. LCMSm/z 567.0♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.40 - 8.35 (m, 1H), 8.07 (dd, J = 8.6, 1.5 Hz, 1H), 7.79 (d, J = 8.6 Hz, 1H), 7.59 (dd, J = 8.0, 8.0 Hz, 1H), 7.35 (dd, J = 10.2, 2.0 Hz, 1H), 7.31 (br dd, J = 8, 2 Hz, 1H), 7.22 (s, 1H), 6.87 (dd, J = 8.3, 8.0 Hz, 1H), 6.63 (br d, J = 8 Hz, 1H), 6.60 (br d, J = 8 Hz, 1H), 4.68 (s, 2H), 4.65 (t, J = 4.9 Hz, 2H), 3.76 (t, J = 4.8 Hz, 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 ]Piperidine-1- Methyl } methyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]6-pyridinesAcid, trifluoroacetate salt (3)

Step 1: 4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine derivatives Synthesis of (C13, free base)

P-toluenesulfonic acid monohydrate (318 mg, 1.67 mmol) was added to a solution of P2(300 mg, 0.670 mmol) in ethyl acetate (3.5 mL). The reaction mixture was stirred at 60 ℃ for 1 hour whereupon it was basified by addition of saturated aqueous potassium carbonate (20 mL) and extracted with a mixture of dichloromethane and methanol (10: 1,3 × 50 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo to afford C13 as a brown solid, the free base. Yield: 230 mg, 0.661 mmol, 99%.

Step 2: 6-bromo-2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4-) Base of]Piperidin-1-yl } methyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]Synthesis of pyridine (C41)

A suspension of C13, free base (130 mg, 0.374 mmol), P13(130 mg, 0.427 mmol), and potassium carbonate (172 mg, 1.24 mmol) in acetonitrile (2 mL) was stirred at 50 ℃ for 16 h. Then, the reaction mixture was purified using preparative thin layer chromatography (eluent: ethyl acetate) to give C41 as a brown oil. Yield: 114 mg, 0.185 mmol, 49%. LCMS m/z617.1 (bromine-chlorine isotope pattern observed) [ M + H]+

And step 3: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]Process for preparing methyl pyridine-6-carboxylate (C42)Synthesis of

C41(114 mg, 0.185 mmol), 1, 3-bis (diphenylphosphino) propane (15.3 mg, 37.1. mu. mol), palladium (II) acetate (8.3 mg, 37. mu. mol), and triethylamine (187 mg, 1.85 mmol) in methanol (5 mL) and carbon monoxide (50 psi) at 80 deg.CN,NThe solution in the mixture of-dimethylformamide (1 mL) was stirred for 16 hours. After the reaction mixture was diluted with ethyl acetate (50 mL), it was washed with saturated aqueous sodium chloride solution (2 × 50 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure. Purification was performed using preparative thin layer chromatography (eluent: ethyl acetate) to afford C42 as a colorless oil. Yield: 60.0 mg, 0.101 mmol, 55%. LCMSm/z617.2 (chlorine isotope pattern observed) [ M + Na+]。

And 4, step 4: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- (2-methoxyethyl) -1HImidazo [4,5-b]Process for preparing pyridine-6-carboxylic acid, trifluoroacetate salt (3) Synthesis of

Aqueous sodium hydroxide (3M; 1.0 mL, 3.0 mmol) was added to a solution of C42(60.0 mg, 0.101 mmol) in methanol (2.0 mL) and the reaction mixture was stirred at 20 ℃ for 2 h. Then, it was adjusted to pH 7 by addition of 1M hydrochloric acid and extracted with a mixture of dichloromethane and methanol (10: 1,3 × 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: 29.6 mg, 42.6. mu. mol, 42%. LCMSm/z 581.0♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ9.13 (d, J = 1.9 Hz, 1H), 8.74 (d, J = 1.9 Hz, 1H), 7.63 (dd, J = 8.3, 8.3 Hz, 1H), 7.30 (dd, J = 10.9, 2.0 Hz, 1H), 7.24 (ddd, J= 8.4, 2.0, 0.7 Hz, 1H), 6.89-6.84 (m, 1H), 6.82-6.77 (m, 2H), 4.98-4.89 (m, 2H, assumed; mostly obscured by water peaks), 4.64 (t,J = 4.8 Hz, 2H), 4.04 - 3.92 (br m, 2H), 3.75 (dd, J = 5.4, 4.2 Hz, 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 = 1.0 Hz, 3H)。

examples 4 and 5

2-({4-[(2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidin-1-yl } Methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-ammonium formate (4) and 2- ({4- [ (2)S)-2-(4- Chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetanyl-2- Radical methyl]-1H-benzimidazole-6-ammonium formate (5)

Step 1: 4- [2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine, trifluoroacetic acid Synthesis of salt (C43)

Trifluoroacetic acid (1.3 mL) was added to a solution of P1(300 mg, 0.691 mmol) in dichloromethane (5 mL). The reaction mixture was stirred at 29 ℃ for 2 hours whereupon it was concentrated in vacuo to give 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]Piperidine- 1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HMethyl (C44) benzimidazole-6-carboxylate and 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidin-1-yl } methyl) -1- [ (2S)- Oxetazedin-2-ylmethyl]-1HSynthesis of methyl (C45) benzimidazole-6-carboxylate

P15(204 mg, 0.692 mmol) was added to a solution of C43 (from the previous step,. ltoreq.0.691 mmol) in acetonitrile (10 mL), followed by potassium carbonate (956 mg, 6.92 mmol). The reaction mixture was stirred at 29 deg.CFor 16 hours, whereupon it is 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 provide a mixture of diastereomeric products as yellow gums (178 mg). Via SFC [ column: 5 μm Chiral Technologies ChiralCel OD; mobile phase: 55:45 carbon dioxide/(methanol with 0.1% ammonium hydroxide) ]Two products were separated. The first eluted diastereomer obtained as a yellow oil was designated C44. Yield: 44.3 mg, 74.8. mu. mol, 11% (in two steps). LCMSm/z592.1♦ [M+H]+. Retention time 4.26 min (column: Chiral Technologies ChiralCel OD-3, 4.6 x 100 mm, 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 maintained at 40% B for 2.5 min; flow rate: 2.8 mL/min).

Via SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 3: 2 carbon dioxide/(methanol containing 0.1% ammonium hydroxide)]The second eluted diastereomer was subjected to a second purification to provide a second eluted diastereomer as a colorless oil, designated C45. Yield: 38 mg, 64. mu. mol, 9% (in two steps). LCMSm/z 592.1♦ [M+H]+. Retention time 4.41 minutes (analytical conditions identical to those for C44).

Absolute stereochemistry represented at dioxolane was assigned via 5 and 5, potency correlation of free acid samples (synthesized from intermediate C48); the absolute stereochemistry of the intermediates was determined via single crystal X-ray structural determination of C49 (hemisulfate salt of C48) (see below).

And step 3: 2- ({4- [(2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine- 1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of (4) -benzimidazole-6-ammonium formate

Aqueous lithium hydroxide (2M; 0.80 mL, 1.6 mmol) was added to a mixture of C44(44.3 mg, 74.8. mu. mol) in methanol (1 mL) and tetrahydrofuran (1 mL)And the reaction mixture was stirred at 26 ℃ for 3 hours. Then, it was adjusted to pH 7 by addition of trifluoroacetic acid and 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: 26.6 mg, 44.7 μmol, 60%. LCMSm/z578.0♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.31 (d, J = 1.4 Hz, 1H), 7.96 (dd, J= 8.5, 1.6 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 7.57 (dd, J = 8.0, 8.0 Hz, 1H), 7.34 (dd, J = 10.1, 2.0 Hz, 1H), 7.29 (br dd, J= 8.3, 2.0 Hz, 1H), 7.20 (s, 1H), 6.86-6.79 (m, 1H), 6.77 (br dd, component of ABC mode,J= 7.9, 1.3 Hz, 1H), 6.73 (dd, components of ABC mode, J= 7.5, 1.4 Hz, 1H), 5.29-5.18 (m, 1H), 4.9-4.78 (m, 1H, assumed; partially obscured by a water peak), 4.68 (dd,J = 15.3, 2.7 Hz, 1H), 4.54 (td, J = 8.0, 5.9 Hz, 1H), 4.44 (dt, J= 9.2, 5.9 Hz, 1H), 4.02 (AB quartet,J AB = 13.9 Hz, ΔνAB = 49.0 Hz, 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)。

and 4, step 4: 2- ({4- [(2 S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine- 1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of (5) -benzimidazole-6-ammonium formate

Aqueous lithium hydroxide (2M; 0.80 mL, 1.6 mmol) was added to a solution of C45(38 mg, 64. mu. mol) in a mixture of methanol (1 mL) and tetrahydrofuran (1 mL) and the reaction mixture was stirred at 24 ℃ for 2.5 hours. Then, it is adjusted to pH 7 by addition of 1M hydrochloric acid, and the resulting mixture is 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) to5 was provided as a white solid. Yield: 27.9 mg, 46.9. mu. mol, 73%. LCMSm/z577.9♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.32 (d, J = 1.4 Hz, 1H), 7.96 (dd, J= 8.5, 1.5 Hz, 1H), 7.66 (d, J = 8.5 Hz, 1H), 7.56 (dd, J = 8.0, 8.0 Hz, 1H), 7.34 (dd, J = 10.2, 2.0 Hz, 1H), 7.29 (br dd, J= 8.3, 2.0 Hz, 1H), 7.20 (s, 1H), 6.85-6.80 (m, 1H), 6.77 (dd, component of ABC mode,J= 8.0, 1.3 Hz, 1H), 6.73 (dd, components of ABC mode,J= 7.5, 1.4 Hz, 1H), 5.30-5.20 (m, 1H), 4.9-4.79 (m, 1H, assumed; partially obscured by a water peak), 4.68 (dd,J = 15.4, 2.7 Hz, 1H), 4.62 - 4.54 (m, 1H), 4.44 (dt, J= 9.2, 5.9 Hz, 1H), 4.02 (AB quartet,J AB = 13.9 Hz, ΔνAB = 44.6 Hz, 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) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid (5, free acid)

Step 1: 4- [(2R) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine-1-carboxylic acid methyl ester Tert-butyl ester (C46) and 4- [ (2)S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine-1- Isolation of tert-butyl formate (C47)

Reverse phase HPLC was used [ column: phenomenex Lux Amylose-1, 5 μm; mobile phase: 9: 1 carbon dioxide/(2-propanol containing 0.2% 1-aminopropan-2-ol) ], separation of P1(10g, 23 mmol) into its constituent enantiomers was carried out. The first eluted enantiomer was designated C46 and the second eluted enantiomer was designated C47, both obtained as colorless oil. The absolute stereochemistry denoted by C46 and C47 was assigned based on single crystal X-ray structural determinations performed on C49 (synthesized from C47) (see below).

Yield of C46: 4.47g, 10.3 mmol, 45%. Retention time: 3.98 min [ column: phenomenex Lux Amylose-1, 4.6 x 250 mm, 5 μm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol containing 0.2% 1-aminopropan-2-ol; gradient: 5% B, 1.00 min, then 5% to 60% B over 8.00 min; flow rate: 3.0 mL/min; back pressure: 120 bar ].

Yield of C47: 4.49g, 10.3 mmol, 45%. Retention time: 4.32 minutes (analytical SFC conditions identical to those used for C46).

Step 2: 4- [(2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine derivatives (C48) Synthesis of (2)

P-toluenesulfonic acid monohydrate (566 mg, 2.98 mmol) was added to a solution of C47(1.12g, 2.58 mmol) in ethyl acetate (26 mL). After heating the reaction mixture at 45 ℃ for 16 h, it was concentrated in vacuo, dissolved in ethyl acetate and washed with saturated aqueous 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(947 mg) as a foamy white solid, LCMSm/z 334.0♦ [M+H]+. A portion of this material, which still contained some p-toluenesulfonic acid, was used in the synthesis of C50 below.

A second foamy white solid (440 mg) was dissolved in ethyl acetate (25 mL) and washed with saturated aqueous sodium bicarbonate (2 × 15 mL); the organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo to afford C48 as a colorless oil (350 mg) containing no more p-toluenesulfonic acid. And (3) regulating the yield: 350 mg, 1.05 mmol, 88%. 1H NMR (400 MHz, chlorine)Imitation-d)δ7.53 (dd, J = 8.4, 7.8 Hz, 1H), 7.22 - 7.13 (m, 3H), 6.87 - 6.80 (m, 1H), 6.79 - 6.71 (m, 2H), 3.23 - 3.14 (m, 2H), 2.86 - 2.69 (m, 3H), 1.90 - 1.68 (m, 4H)。

And step 3: 4- [(2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine, hemithio Synthesis of acid salt (C49)

A 0.1M solution of C48 (from the previous colorless oil) in ethyl acetate was prepared and subjected to salt screening. Only sulfate formation is described herein. The mixture of sulfuric acid (25 μmol) and substrate solution (0.1M, 250 μ L, 25 μmol) 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 (approximately 150 μ L) until a solution was formed; it was allowed to slowly evaporate overnight until approximately 50 μ Ι _ of solvent remained. One of the resulting crystals was analyzed by single crystal X-ray texture analysis, and absolute stereochemistry was established as the absolute stereochemistry 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 consisted of both omega and phi scans.

By using the SHELX software suite, the internal phase is determined by the internal phaseP 1The structure is analyzed. The structure is then refined by full matrix least squares. All non-hydrogen atoms were found and refined using anisotropic shift parameters.

The hydrogen atoms located on the nitrogen are found from the fourier difference plot and refined at a limited distance. The remaining hydrogen atoms are placed in the calculated positions and are carried on their carrier atoms. The final refinement includes the isotropic displacement parameters of all hydrogen atoms.

The asymmetric unit is composed of two molecules of protonated C48, one molecule of doubly deprotonated sulfuric acid, and one molecule of water in its complete position. Thus, the structures are hemisulfate and hemihydrate. The chlorofluorophenyl ring is disordered and modeled with an occupancy of 60/40, with the ring flipped over in two positions.

Absolute structural analysis using the likelihood method (Hooft, 2008) was performed using platon (spek). The results indicate that the absolute structure has been correctly assigned; the method calculates the probability of the structure being correct to be 100.0. The Hooft parameter is reported as 0.061, esd as 0.004, and the Parson parameter is reported as 0.063, esd as 0.005.

The final R index was 3.1%. The final differential fourier shows the electron density without missing or misplacement.

Relevant crystal, data collection, and refinement information are summarized in table E. The atomic coordinates, bond lengths, bond angles, and displacement parameters are listed in tables F-H.

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. 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, 453-457.

OLEX2, O.V. Dolomanov, L.J. Bourhis, R.J. Gildea, J.A.K. Howard and H.Puschmann,J. Appl. Cryst. 2009, 42, 339-341.

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。

table E: crystal data and structure refinement of C49.

Table F: atomic coordinate of C49 (x 10)4) And an equivalent isotropic displacement parameter (A)2 x 103). U (eq) is defined as orthogonal UijOne third of the trace amount of tensor.

Table G: the bond length [ A ] and angle [ ] of C49.

For generating a symmetric transformation of equivalent atoms.

Table H: anisotropy displacement parameter (A) of C492 x 103). The anisotropy displacement factor index takes the following form:

-2π2[h2 a*2U11 + ... + 2 h k a* b* U12 ]。

and 4, step 4: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine- 1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of methyl (C50) benzimidazole-6-carboxylate

By usingN,NDiisopropylethylamine (0.68 mL, 3.9 mmol) A solution of C48(500 mg, from the previous foamy white solid, corrected for p-toluenesulfonic acid: 1.25 mmol) in acetonitrile (6 mL) was treated and allowed to stir at 45 ℃ for 5 min. After addition of P15(319 mg, 1.08 mmol), stirring was continued at 45 ℃ for 7.25 h whereupon the reaction mixture was diluted with water (6 mL) and acetonitrile (2 mL) at 45 ℃. The resulting heterogeneous mixture was allowed to cool to room temperature and stirred for 72 hours. More water (5 mL) was added and after a further 30 minutes of stirring, via The solid was collected by filtration and washed with a mixture of acetonitrile and water (15:85, 3 × 5 mL) to give C50 as a white solid with a slightly pink character. Yield: 605 mg, 1.02 mmol, 82%. LCMSm/z 592.0♦ [M+H]+1H NMR (400 MHz, chloroform-d)δ8.17 (d, J = 1.6 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 7.51 (dd, J= 8.0, 8.0 Hz, 1H), 7.19 (br s, 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, components of ABX pattern,J= 15.3, 5.9 Hz, 1H), 4.67 (dd, components of the ABX pattern,J = 15.3, 3.5 Hz, 1H), 4.63 - 4.55 (m, 1H), 4.38 (ddd, J = 9.1, 6.0, 5.9 Hz, 1H), 3.94 (s, 5H), 3.03 - 2.89 (m, 2H), 2.77 - 2.65 (m, 2H), 2.51 - 2.39 (m, 1H), 2.34 - 2.20 (m, 2H), 1.91 - 1.76 (m, 4H)。

and 5: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzodioxol-4-yl]Piperidine- 1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of (5, free acid) benzimidazole-6-carboxylic acid

A suspension of C50(595 mg, 1.00 mmol) in methanol (10 mL) was heated to 45 ℃ and treated with aqueous sodium hydroxide (1M; 2.01 mL, 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, 1 mL) to bring the pH to 5 to 6. Water (10 mL) 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, 3 × 5 mL) to give a solid (433 mg). A portion of this material (300 mg) was stirred with a mixture of heptane and ethyl acetate (1: 3, 5 mL) at 40 ℃ for 1 hour; after cooling to room temperature with continued stirring, the solid was collected via filtration and washed with a mixture of heptane and ethyl acetate (3: 1,3 × 3 mL) to give 5, the free acid as a white solid. Yield: 260 mg, 0.450 mmol, corresponding to 65% for the entire reaction. LCMS m/z 578.0♦ [M+H]+1H NMR (400 MHz, DMSO-d 6) δ12.75 (v br s, 1H), 8.26 (br s, 1H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.66 - 7.56 (m, 3H), 7.40 (dd, J= 8.3, 2.0 Hz, 1H), 7.35 (s, 1H), 6.87-6.75 (m, 3H), 5.13-5.03 (m, 1H), 4.76 (dd, component of ABX pattern,J= 15.3, 7.2 Hz, 1H), 4.62 (dd, components of the ABX pattern,J = 15.2, 2.8 Hz, 1H), 4.46 - 4.38 (m, 1H), 4.34 (ddd, J= 9.0, 5.9, 5.8 Hz, 1H), 3.84 (AB quartet,J AB = 13.5 Hz, ΔνAB = 67.7 Hz, 2H), 3.00 (br d, J = 11.2 Hz, 1H), 2.84 br (d, J = 11.3 Hz, 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 with that of 4 and 5, the 5, free acid sample exhibiting an EC of 25 nM in assay 250(geometric mean of 3 replicates). The activities of the ammonium salts of example 4 and example 5 in assay 2 were respectively>20000 nM (2 replicates) and 20 nM (geometric mean of 3 replicates).

Example 5 Synthesis of a 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt

1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 2- ({4- [ (2)S) -2- (4-chloro-2-fluorophenyl) -1, 3-benzene And dioxol-4-yl]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole- 6-formate (5, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt)

A mixture of 5, the free acid (0.50g, 0.86 mmol) in tetrahydrofuran (4 mL) was treated with an aqueous solution of 2-amino-2- (hydroxymethyl) propane-1, 3-diol (Tris, 1.0M; 0.5 mL, 1.0 mmol). After 20 hours, the mixture was concentrated with ethanol (2 × 6 mL) 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 × 10 mL) and dried in vacuo to The 5, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt was obtained as a white solid. Yield: 410 mg, 0.586 mmol, 68%.1H NMR (600 MHz, DMSO-d 6) And characteristic peak: δ 8.19 (s, 1H), 7.78 (br d,J = 8.4 Hz, 1H), 7.62 - 7.58 (m, 2H), 7.55 (d, J = 8.3 Hz, 1H), 7.40 (dd, J = 8.4, 2.0 Hz, 1H), 7.35 (s, 1H), 6.85 - 6.80 (m, 2H), 6.79 (dd, J = 6.9, 2.4 Hz, 1H), 5.11 - 5.05 (m, 1H), 4.73 (dd, J = 15.2, 7.2 Hz, 1H), 4.60 (dd, J = 15.3, 2.9 Hz, 1H), 4.45 - 4.39 (m, 1H), 4.34 (ddd, J = 9.0, 6.0, 5.8 Hz, 1H), 3.91 (d, J = 13.5 Hz, 1H), 3.74 (d, J = 13.5 Hz, 1H), 2.99 (br d, J = 11.1 Hz, 1H), 2.85 (br d, J= 11.3 Hz, 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]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-ammonium formate (6) and 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidin-1-yl } methyl) -1- [(2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-ammonium formate (7)

Step 1: 4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]A piperidine compound, synthesis of p-toluenesulfonate (C13)

A solution of P2(150 mg, 0.335 mmol) and P-toluenesulfonic acid monohydrate (159 mg, 0.836 mmol) in ethyl acetate (2.0 mL) was stirred at 60 ℃ for 3.5 h. The reaction mixture was concentrated in vacuo to give C13 as a brown oil, which was used directly in the next step. LCMSm/z 348.1♦ [M+H]+

Step 2: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of methyl (C51) benzimidazole-6-carboxylate

P15(99.1 mg, 0.336 mmol) was added to a suspension of C13 (from the previous step;. ltoreq.0.335 mmol) and potassium carbonate (232 mg, 1.68 mmol) 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 (390 mg) was combined with material from a similar reaction using C13(≦ 0.11 mmol), it was diluted with water (20 mL) 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 dichloromethane/methanol) to provide C51, a mixture of diastereomers as a colorless oil. And (4) merging yield: 80.6 mg, 0.133 mmol, 30% (in two steps). LCMSm/z 606.2♦ [M+H]+

And step 3: 2- ({4- [(2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-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- [ (2) yl ] -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1HIsolation of methyl (C53) benzimidazole-6-carboxylate

Via repeated SFC [ column: chip Technologies chipak AD, 10 μm; mobile phase: 65: 35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]Separation of C51(180 mg, 0.297 mmol) into its constituent diastereomers was carried out. The first diastereomer eluted was designated C52. Yield: 61.2 mg, 0.101 mmol, 34%. LCMSm/z 627.9♦ [M+Na+]. Retention time: 5.03 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 150 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol with 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then maintained at 40% B for 3.0 minutes; flow rate: 2.5 mL/min).

The second eluted diastereomer is designated C53. Upon analysis, this material proved to be contaminated with the corresponding ethyl ester; it is brought to the hydrolysis step (to give 7) in the form of this mixture. Yield: 40.0 mg, 66.0. mu. mol, 22%. LCMSm/z 606.0♦ [M+H]+. Retention time: 5.19 minutes (analytical conditions identical to those for C52).

Absolute stereochemistry indicated at dioxolane via 7 and 7, assignment of potency correlation of free acid samples (synthesized from intermediate P3, see example 7 below, alternative synthesis of free acid); the absolute stereochemistry of P3 was determined via single crystal X-ray structural determination of C8 (see below).

And 4, step 4: 2- ({4- [(2R) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of (6) -ammonium benzimidazole-6-formate

Aqueous lithium hydroxide (2M; 0.990 mL, 1.98 mmol) was added to a solution of C52(60 mg, 99. mu. mol) in a mixture of methanol (1.0 mL) and tetrahydrofuran (1.0 mL) and the reaction mixture was stirred at 20 ℃ for 16 h. Trifluoroacetic acid is added until the pH of the reaction mixture reaches 7 whereupon it is concentrated in vacuo and the residue is 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♦ [M+H]+1H NMR (400 MHz, methanol-d 4) And characteristic peak: delta 8.35 (d,J = 1.3 Hz, 1H), 7.97 (dd, J = 8.5, 1.5 Hz, 1H), 7.67 (d, J = 8.5 Hz, 1H), 7.58 (dd, J = 8.3, 8.3 Hz, 1H), 7.28 (dd, J = 10.9, 2.0 Hz, 1H), 7.21 (br dd, J = 8.4, 1.9 Hz, 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, 2.7 Hz, 1H), 4.49 (dt, J= 9.1, 6.0 Hz, 1H), 4.03 (AB quartet,J AB = 13.9 Hz, ΔνAB = 47.8 Hz, 2H), 3.14 (br d, J = 11 Hz, 1H), 3.02 (br d, J = 11.5 Hz, 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 = 1.1 Hz, 3H), 2.01 - 1.87 (m, 2H), 1.87 - 1.78 (br m, 2H)。

and 5: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of (7) -benzimidazole-6-ammonium formate

Aqueous lithium hydroxide (2M; 0.642 mL, 1.28 mmol) was added to a solution of C53(38.9 mg, 64.2. mu. mol) in a mixture of methanol (1.0 mL) and tetrahydrofuran (1.0 mL). After stirring the reaction mixture at 20 ℃ for 16 h, it was adjusted to pH 7 by 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: 25.1 mg, 41.2 μmol, 64%. LCMS m/z 591.9♦ [M+H]+1H NMR (400 MHz, methanol-d 4) And characteristic peak: delta 8.34 (d,J = 1.5 Hz, 1H), 7.98 (dd, J = 8.5, 1.6 Hz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.58 (dd, J = 8.3, 8.3 Hz, 1H), 7.28 (dd, J = 10.9, 2.0 Hz, 1H), 7.20 (br dd, J = 8.4, 1.9 Hz, 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, 2.7 Hz, 1H), 4.68 - 4.61 (m, 1H), 4.48 (dt, J= 9.1, 5.9 Hz, 1H), 4.05 (AB quartet,J AB = 13.9 Hz, ΔνAB = 44.1 Hz, 2H), 3.15 (br d, J = 11.7 Hz, 1H), 3.03 (br d, J = 11.6 Hz, 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]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid (7, free acid)

Step 1: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HOf methyl benzimidazole-6-carboxylate (C53) Synthesis of

Will be provided withN,N-diisopropylethylamine (15.1 mL, 86.9 mmol) was added to a mixture of P3(8.22g, 15.8 mmol) in acetonitrile (185 mL); after stirring for 5 min, P15(4.57g, 15.5 mmol) was added and the reaction mixture was heated at 45 ℃. After 4 hours, the reaction mixture was concentrated to half its original volume in vacuo, and the resulting mixture was diluted with water (100 mL) and extracted with ethyl acetate (2 × 100 mL). The combined organic layers were washed with water (50 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (gradient: 50% to 100% ethyl acetate in heptane) afforded C53 as a white solid. Yield: 8.4g, 13.9 mmol, 88%. LCMS m/z 606.1♦ [M+H]+1H NMR (600 MHz, DMSO-d 6) δ8.30 (s, 1H), 7.82 (br d, J = 8.4 Hz, 1H), 7.67 (d, J = 8.4 Hz, 1H), 7.58 - 7.53 (m, 2H), 7.33 (dd, J = 8.4, 2.1 Hz, 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, 7.2 Hz, 1H), 4.67 (dd, J = 15.3, 2.8 Hz, 1H), 4.51 - 4.44 (m, 1H), 4.37 (ddd, J = 8.9, 5.9, 5.9 Hz, 1H), 3.97 (d, J = 13.6 Hz, 1H), 3.87 (s, 3H), 3.78 (d, J = 13.5 Hz, 1H), 3.02 (br d, J = 11.1 Hz, 1H), 2.86 (br d, J = 11.3 Hz, 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- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HOf (7, free acid) benzimidazole-6-carboxylic acid Synthesis of

A mixture of C53(8.40g, 14.0 mmol) in methanol (135 mL) was heated to 45 ℃ and treated with aqueous sodium hydroxide (1M; 27.7 mL, 27.7 mmol). After 20 hours, the reaction mixture was concentrated to half its original volume in vacuo. The resulting mixture was diluted with water (100 mL) and the pH adjusted to 5-6 using aqueous citric acid (1M, 15 mL). The resulting solid was filtered, washed with water (2 × 15 mL), and transferred to a separatory funnel as a solution in ethyl acetate (50 mL); in this way, residual water is removed. The organic layer was dried over magnesium sulfate, filtered, and combined with four batches prepared in advance by a similar procedure (amount of C53 used for these reactions was 987 mg, 1.63 mmol; 1.15g, 1.90 mmol; 8.57g, 14.1 mmol; and 12.6g, 20.8 mmol) 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 × 25 mL) to give 7 as a white solid, the free acid. Yield: 29.4g, 0.527 mmol, 74% (for the combined reactions). LCMS 592.2 ♦ [ M + H ] ]+1H NMR (600 MHz, DMSO-d 6 ):δ12.74 (br s, 1H), 8.28 (s, 1H), 7.80 (br d, J = 8.4 Hz, 1H), 7.64 (d, J = 8.4 Hz, 1H), 7.59 - 7.52 (m, 2H), 7.33 (dd, J = 8.4, 2.1 Hz, 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, 7.3 Hz, 1H), 4.65 (dd, J = 15.2, 2.8 Hz, 1H), 4.51 - 4.45 (m, 1H), 4.38 (ddd, J = 9.0, 5.9, 5.9 Hz, 1H), 3.96 (br d, J = 13.6 Hz, 1H), 3.78 (br d, J = 13.5 Hz, 1H), 3.02 (br d, J = 11.1 Hz, 1H), 2.86 (br d, J= 11.1 Hz, 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 with that of 6 and 7, and in assay 2, this 7, free acid sample exhibited an EC of 4.3 nM50(geometric mean of 3 replicates). The activity of the ammonium salts of example 6 and example 7 in assay 2 was 2400 nM (geometric mean of 5 replicates) and 2.9 nM (geometric mean of 8 replicates), respectively.

Synthesis of 7S-1. example 7, synthesis of 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt

1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 2- ({4- [ (2)S) -2- (4-chloro-2-fluorophenyl) -2-methyl- 1, 3-benzodioxol-4-yl]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzene Benzimidazole-6-formate (7, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt)

A mixture of 7, the free acid (2.00g, 3.38 mmol) in tetrahydrofuran (16 mL) was treated with an aqueous solution of 2-amino-2- (hydroxymethyl) propane-1, 3-diol (Tris, 1.0M; 3.55 mL, 3.55 mmol). After 18 hours, the reaction mixture was concentrated and treated with ethanol (30 mL) in vacuo. After stirring the mixture for 23 hours, the solid was collected via filtration and washed with ethyl acetate (2 × 10 mL) to give 7, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt as a white solid. Yield: 1.41g, 1.98 mmol, 59%. LCMS m/z592.3♦ [M+H]+1H NMR (600 MHz, DMSO-d 6) And characteristic peak: δ 8.20 (s, 1H), 7.79 (d,J = 8.4 Hz, 1H), 7.59 - 7.52 (m, 3H), 7.33 (br d, J = 8.5 Hz, 1H), 6.81 - 6.72 (m, 3H), 5.14 - 5.07 (m, 1H), 4.76 (dd, J = 15.2, 7.2 Hz, 1H), 4.63 (br d, J = 15.4 Hz, 1H), 4.50 - 4.44 (m, 1H), 4.37 (ddd, J = 8.9, 5.9, 5.9 Hz, 1H), 3.94 (d, J = 13.4 Hz, 1H), 3.76 (d, J = 13.4 Hz, 1H), 3.01 (br d, J = 11.1 Hz, 1H), 2.86 (br d, J= 11.2 Hz, 1H), 2.73-2.60 (m, 2H), 2.5-2.41 (m, 1H), 2.27-2.20 (m, 1H), 2.20-2.13 (m, 1H), 2.02 (s, 3H), 1.83-1.64 (m, 4H). mp = 175 ℃ to 180 ℃.

Synthesis of 7S-2: example 7 alternative Synthesis of a 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt

A3.3M solution of 2-amino-2- (hydroxymethyl) -1, 3-propanediol (1.0 eq., 1.93L) in water was added to a solution of 7, the free acid (3.74 kg) in isopropanol (20L) at 65 ℃. Additional isopropanol (19L) was added followed by methanol (19L) while maintaining the temperature at 65 ℃. Over a period of 2 hours, the mixture was slowly cooled to 45 ℃ and then maintained at 45 ℃ for at least 12 hours. Then, over a period of 3 hours, the mixture was cooled to 5 ℃ and then maintained at 5 ℃ for at least 3 hours. Then, the mixture was filtered and the solid was collected and washed with ethyl acetate (2 × 10 mL) to give 7, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt as a white solid (yield: 3.64 kg, 80.9%). To obtain LCMS and1h NMR data substantially identical to those in the synthesis of 7S-1 shown above.

In the case of the example 7, the following examples are given,1, 3-dihydroxy Powder X-ray diffraction of form I of the (E) -2- (hydroxymethyl) propan-2-aminium salt (PXRD) data acquisition

PXRD analysis was performed on the white solid of the tris salt of example 7 (from Synthesis 7S-1 and Synthesis 7S-2) and it was found to be a crystalline material (designated form I for this anhydrous crystalline form). Powder X-ray diffraction analysis was performed using a Bruker AXS D8 Endevidor diffractometer equipped with a Cu radiation sourceIn (1). The divergence slit was set at 15 mm continuous illumination. The diffracted radiation was detected by a PSD-Lynx Eye detector with the detector PSD on set at 2.99 degrees. The X-ray tube voltage and current intensity were set at 40 kV and 40 mA, respectively. At Cu wavelength (CuK)= 1.5418 λ), data was collected from 3.0 to 40.0 degrees 2 θ in a θ - θ goniometer using a 0.01 degree step and a 1.0 second step time. The anti-scatter screen is set to a fixed distance of 1.5 mm. During data collection, the sample was rotated. Samples were prepared by placing them in a silicon low background sample holder and rotating them during collection. Data were collected and analyzed by EVA DIFFRAC Plus software using Bruker DIFFRAC Plus software. Prior to peak searching, the PXRD data file is unprocessed. The peak selected by threshold 1 is used for preliminary peak assignment using a peak search algorithm in the EVA software. To ensure effectiveness, the adjustment is made manually; the automatically assigned output is visually inspected and the peak position is adjusted to the peak maximum. The relative intensities are generally selected >Peak 3%. In general, unresolved peaks or peaks that coincide with noise are not selected. Typical errors associated with peak position over PXRD specified by USP are up to +/-0.2-theta (USP-941). One diffractogram was always observed and is provided in figure 1. Diffraction peaks expressed in degrees 2 theta and relative intensities (relative intensities) of PXRD of the sample obtained by the above synthesis of 7S-2>3%) is provided in table X1.

TABLE X1

Corner (2 theta) Relative Strength (%)
3.7 74.3
7.3 83.3
8.1 12.5
8.5 6.5
10.1 6.6
13.6 3.5
14.7 49.8
15.2 7.9
15.5 28.7
15.9 18.3
16.9 60.8
17.4 26.3
17.7 11.4
17.9 13.5
18.9 75.4
19.7 18.7
20.2 100.0
20.9 24.8
21.5 14.8
22.2 31.7
22.9 10.1
23.5 34.6
23.7 8.2
24.4 6.5
24.9 8.7
25.2 6.4
25.9 14.7
26.4 48.6
26.7 12.5
27.5 15.8
27.9 6.1
28.3 10.5
29.5 15.5
29.8 12.6
30.2 12.1
30.9 3.4
31.7 16.4
33.3 17.2
34.0 14.9
35.8 4.8
37.5 3.2
38.6 5.3

Examples 8 and 9

2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine-1- Methyl group) -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1HBenzimidazole-6-carboxylic acid, DIAST-X1 (8) [ from C56](ii) a And 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperidine-1- Methyl group) -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1HBenzimidazole-6-carboxylic acid, DIAST-X2 (9) [ from C57]

Step 1: 3-fluoro-4- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl]A process for the preparation of a benzonitrile, synthesis of p-toluenesulfonate (C54)

P-toluenesulfonic acid (158 mg, 0.919 mmol) was added to a solution of P4(161 mg, 0.367 mmol) in ethyl acetate (8 mL) and the reaction mixture was stirred at 65 ℃ for 16 h. The solvent was removed in vacuo to afford C54 as a dark yellow gum; this material was used directly in the next step.

Step 2: 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl] Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HSynthesis of methyl (C55) benzimidazole-6-carboxylate

Potassium carbonate (219 mg, 1.58 mmol) was added to C54 (from the previous step;. ltoreq.0.367 mmol) inTo a solution in acetonitrile (3.7 mL) was then added P15(115 mg, 0.390 mmol). The reaction mixture was stirred at 50 ℃ for 20 hours, whereupon it was diluted with ethyl acetate (10 mL) and filtered. The filter cake was washed with ethyl acetate (3 × 10 mL) and the combined filtrates were concentrated in vacuo. Silica gel chromatography (gradient: 0% to 100% ethyl acetate in petroleum ether) afforded C55 as a dark yellow oil. Yield: 191.0 mg, 0.320 mmol, 87% (in two steps). LCMSm/z 619.1 [M+Na+]。

And step 3: 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl] Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-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]Piperidine-1- Methyl group) -1- [ (2)S) -Oxetazedin-2-ylmethyl]-1HIsolation of-benzimidazole-6-carboxylic acid methyl ester, ENT-2 (C57)

Via SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 3: 2 carbon dioxide/2-propanol]Separation of C55(191 mg, 0.320 mmol) into its constituent stereoisomers in dioxolane was carried out. The first eluting isomer was obtained as a white gum and was designated ENT-1 (C56). Yield: 114 mg; this material contained the remaining ethanol. LCMSm/z 597.1 [M+H]+. Retention time 4.40 min (column: Chiral Technologies Chiral cel OD-3, 4.6 x 100 mm, 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 for 2.5 min; flow rate: 2.8 mL/min).

SFC [ column: chiral Technologies Chiral cel OD, 5 μm; mobile phase: 55: 45 carbon dioxide/(2-propanol containing 0.1% ammonium hydroxide)]The second eluted isomer was repurified to give a colorless gum designated ENT-2 (C57). Yield: 50 mg, 83.8. mu. mol, 26%. LCMSm/z 597.1 [M+H]+. Retention time 4.74 minutes (analytical conditions identical to those for C56).

And 4, step 4: 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl] Piperidin-1-yl } methyl) -1- [ (2 S) -Oxetazedin-2-ylmethyl]-1HBenzimidazole-6-carboxylic acid, DIAST-X1 (8) [ from C56]Synthesis of (2)

With 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a]An aqueous solution of pyrimidine (0.97M, 394 μ L, 0.382 mmol) was treated with a solution of C56(114 mg, 0.191 mmol) in acetonitrile (10 mL) and the reaction mixture was stirred at room temperature for 23 h. Adding more 1,3,4,6,7, 8-hexahydro-2H-pyrimido [1,2-a]An aqueous solution of pyrimidine (0.97M, 394 μ L, 0.382 mmol) and stirring was continued for 6 hours whereupon the pH was carefully adjusted to 7 to 8 by 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 afford 8 as a white solid. Yield: 22.2 mg, 38.1. mu. mol, 20%. LCMSm/z 583.3 [M+H]+1H NMR (400 MHz, methanol-d 4):δ8.19 (d, J = 1.4 Hz, 1H), 7.94 (dd, J = 8.4, 1.5 Hz, 1H), 7.77 (dd, J = 7.7, 7.7 Hz, 1H), 7.64 (dd, J = 10.6, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.57 (dd, J = 8.0, 1.5 Hz, 1H), 6.81 - 6.75 (m, 1H), 6.75 - 6.68 (m, 2H), 5.34 - 5.25 (m, 1H), 4.73 (dd, J = 15.3, 3.0 Hz, 1H), 4.67 - 4.59 (m, 1H), 4.49 (dt, J= 9.2, 6.0 Hz, 1H), 3.96 (AB quartet,J AB = 13.7 Hz, ΔνAB = 41.2 Hz, 2H), 3.06 (br d, J = 11 Hz, 1H), 2.95 (br d, J = 11 Hz, 1H), 2.87 - 2.76 (m, 1H), 2.71 (tt, J = 12.0, 3.9 Hz, 1H), 2.61 - 2.50 (m, 1H), 2.36 - 2.21 (m, 2H), 2.06 (s, 3H), 1.95 - 1.72 (m, 4H)。

and 5: 2- ({4- [2- (4-cyano-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl] Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HBenzimidazole-6-carboxylic acid, DIAST-X2 (9) [ from C57]Synthesis of (2)

With 1,3,4,6,7, 8-hexahydro-2 H-pyrimido [1,2-a]An aqueous solution of pyrimidine (0.97M, 173. mu.L, 0.168 mmol) was treated with a solution of C57(50 mg, 84. mu. mol) in acetonitrile (10 mL). The reaction was stirred at room temperature (about 25 deg.C) for 16 hours whereupon an additional amount of 1,3,4,6,7, 8-hexahydro-2 was addedH-pyrimido [1,2-a]An aqueous solution of pyrimidine (0.97M, 173. mu.L, 0.168 mmol) and stirring was continued at 25 ℃ for 29 h. The reaction mixture was then carefully adjusted to 7 to 8 by addition of 1M hydrochloric acid; the resulting mixture was concentrated and subjected to reverse phase HPLC (column: Xtimate. C18, 5 μm; mobile phase A: 0.05% ammonium hydroxide in water; mobile phase B: acetonitrile; gradient: 27% to 67% B) in vacuo to give 9 as a white solid. Yield: 18.0 mg, 30.9. mu. mol, 37%. LCMSm/z 583.3 [M+H]+1H NMR (400 MHz, methanol-d 4) δ 8.36 - 8.33 (m, 1H), 7.97 (dd, J = 8.5, 1.5 Hz, 1H), 7.78 (dd, J = 7.7, 7.7 Hz, 1H), 7.70 - 7.63 (m, 2H), 7.57 (dd, J= 8.0, 1.5 Hz, 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, hypothetical; partially obscured by a water peak), 4.73 (dd, components of the ABX pattern,J = 15.3, 2.7 Hz, 1H), 4.68 - 4.60 (m, 1H), 4.50 (dt, J= 9.2, 6.0 Hz, 1H), 4.02 (AB quartet,J AB = 13.8 Hz, ΔνAB = 48.2 Hz, 2H), 3.13 (br d, J = 11 Hz, 1H), 3.01 (br d, J = 11.5 Hz, 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]Piperidine-1- Methyl group) -1- [ (2)S) -Oxetazedin-2-ylmethyl ]-1HBenzimidazole-6-carboxylic acid, DIAST-X2 (10) [ from P9]

Step 1: 5-chloro-2- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl]The amount of pyridine, ENT-X2, P-toluenesulfonate (C58) [ from P9]Synthesis of (2)

A solution of P9(228 mg, 0.529 mmol) in ethyl acetate (2.7 mL) was treated with P-toluenesulfonic acid monohydrate (116 mg, 0.610 mmol) and the reaction mixture was heated at 50 ℃ for 16 h. Then, it was allowed to stir at room temperature overnight whereupon the precipitate was collected via filtration and washed with a mixture of ethyl acetate and heptane (1: 1, 2 × 20 mL) to provide C58 as a white solid. Yield: 227 mg, 0.451 mmol, 85%. LCMSm/z331.0♦ [M+H]+1H NMR (400 MHz, DMSO-d 6): δ8.73 (d, J = 2.4 Hz, 1H), 8.61 - 8.46 (br m, 1H), 8.35 - 8.18 (br m, 1H), 8.02 (dd, J = 8.5, 2.5 Hz, 1H), 7.64 (d, J = 8.5 Hz, 1H), 7.47 (d, J = 7.8, 2H), 7.11 (d, J= 7.8 Hz, 2H), 6.89-6.81 (m, 2H), 6.72 (quintuple,J= 4.0 Hz, 1H), 3.45-3.27 (m, 2H, assumed; partially obscured 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: 2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid methyl ester, DIAST-Y2 (C59) [ from P9]Synthesis of (2)

Will be provided withN,NDiisopropylethylamine (0.234 mL, 1.34 mmol) was added to a solution of C58(225 mg, 0.447 mmol) in acetonitrile (2.2 mL). After stirring the mixture at 45 ℃ for 5 min, P15(120 mg, 0.407 mmol) was added and stirring was continued at 45 ℃ for 16 h whereupon P15(11 mg, 37. mu. mol) was added again. After stirring for another 3 hours, the reaction mixture was treated with water (2.5 mL) and allowed to cool to room temperature. More water (5 mL) 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 × 5 mL) to give C59(252 mg) as an off-white solid.1H NMR analysis showed that this material contained someN,NDiisopropylethylamine and was used directly in the following step. LCMSm/z589.1♦ [M+H]+1H NMR (400 MHz, chloroform-d) 8.61 (d, J = 2.3 Hz, 1H), 8.18 (d, J = 1.5 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.74 (d, J= 8.5 Hz, 1H), 7.67 (dd, components of the ABX pattern,J= 8.4, 2.4 Hz, 1H), 7.59-7.51 (m, 1H), 6.82-6.75 (m, 1H), 6.74-6.66 (m, 2H), 5.28-5.19 (m, 1H), 4.75 (dd, component of ABX pattern,J= 15.3, 6.0 Hz, 1H), 4.68 (dd, components of the ABX pattern,J = 15.3, 3.4 Hz, 1H), 4.67 - 4.58 (m, 1H), 4.41 (ddd, J = 9.1, 5.9, 5.9 Hz, 1H), 3.95 (s, 2H), 3.95 (s, 3H), 3.07 - 2.89 (m, 2H), 2.81 - 2.69 (m, 2H), 2.53 - 2.41 (m, 1H), 2.37 - 2.22 (m, 2H), 2.05 (s, 3H), 1.93 - 1.74 (m, 4H)。

and step 3: 2- ({4- [2- (5-Chloropyridin-2-yl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1HBenzimidazole-6-carboxylic acid, DIAST-X2 (10) [ from P9]Synthesis of (2)

A suspension of C59 (from the previous step; 250 mg,. ltoreq.0.407 mmol) in methanol (2 mL) was heated to 40 ℃ whereupon aqueous sodium hydroxide solution (1M; 0.81 mL, 0.81 mmol) was added. After 17 hours, the reaction mixture was cooled to room temperature and the pH was adjusted to 5 to 6 with 1M aqueous citric acid. The resulting mixture was diluted with water (2 mL), stirred for 2 hours, and extracted with ethyl acetate (3 × 5 mL); the combined organic layers were washed with saturated aqueous sodium chloride (5 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford a foamy solid. This material was taken up in a mixture of ethyl acetate and heptane (1: 1, 4 mL), heated to 50 ℃, and then allowed to cool and stir overnight. Filtration afforded 10 as a white solid. Yield: 179 mg, 0.311 mmol, 76% (in two steps). LCMS m/z575.1♦ [M+H]+1H NMR (400 MHz, DMSO-d 6) δ12.73 (br s, 1H), 8.71 (d, J = 2.5 Hz, 1H), 8.27 (d, J = 1.5 Hz, 1H), 8.00 (dd, J = 8.5, 2.5 Hz, 1H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.64 (d, J = 8.4 Hz, 1H), 7.60 (d, J= 8.5 Hz, 1H), 6.83-6.72 (m, 3H), 5.14-5.06 (m, 1H), 4.77 (dd, component of ABX pattern,J= 15.2, 7.2 Hz, 1H), 4.63 (dd, components of the ABX pattern,J = 15.2, 2.8 Hz, 1H), 4.50 - 4.42 (m, 1H), 4.37 (ddd, J= 9.0, 5.9, 5.9 Hz, 1H), 3.85 (AB quartet,J AB = 13.6 Hz, ΔνAB = 71.5 Hz, 2H), 3.01 (br d, J = 11.2 Hz, 1H), 2.85 (br d, J = 11.2 Hz, 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)。

synthesis of 10S-1. example Synthesis of 10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt

1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium 2- ({4- [2- (5-chloropyridin-2-yl) -2-methyl-1, 3- Benzodioxol-4-yl]Piperidin-1-yl } methyl) -1- [ (2S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole Azole-6-formate, DIAST-X2 (10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt) [ from P9]Synthesis of (2)

A mixture of 10(1.54g, 2.68 mmol) in tetrahydrofuran (10 mL) was treated with an aqueous solution of 2-amino-2- (hydroxymethyl) propane-1, 3-diol (Tris, 1.0M; 2.81 mL, 2.81 mmol). After 24 hours, the reaction mixture was concentrated with ethanol (2 × 50 mL) in vacuo. The residue was treated with ethanol (15 mL). After stirring for 20 hours, the solid was collected via filtration and washed with cold ethanol (5 mL) to give 10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt as a white solid. Yield: 1.41g, 2.03 mmol, 76%. LCMSm/z575.3♦ [M+H]+1H NMR (600 MHz, DMSO-d 6)δ8.71 (d, J = 2.5 Hz, 1H), 8.21 (br s, 1H), 8.00 (dd, J = 8.5, 2.5 Hz, 1H), 7.79 (br d, J = 8.4 Hz, 1H), 7.60 (d, J = 8.5 Hz, 1H), 7.57 (d, J = 8.4 Hz, 1H), 6.82 - 6.73 (m, 3H), 5.13 - 5.07 (m, 1H), 4.74 (dd, J = 15.3, 7.2 Hz, 1H), 4.61 (dd, J = 15.3, 2.9 Hz, 1H), 4.49 - 4.43 (m, 1H), 4.37 (ddd, J = 9.0, 5.9, 5.9 Hz, 1H), 3.93 (d, J = 13.6 Hz, 1H), 3.75 (d, J = 13.5 Hz, 1H), 3.01 (br d, J = 11.3 Hz, 1H), 2.86 (br d, J= 11.4 Hz, 1H), 2.73-2.59 (m, 2H), 2.48-2.37 (m, 1H), 2.27-2.20 (m, 1H), 2.19-2.12 (m, 1H), 2.01 (s, 3H), 1.82-1.66 (m, 4H). mp = 184 to 190 ℃.

Synthesis of 10S-2. example 10 alternative Synthesis of 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt

A mixture of 10(8.80 gm, 15.3 mmol) in 2-methyltetrahydrofuran (90 ml) was concentrated in vacuo on a rotary evaporator in a 37 deg.C water bath to reduce the total volume to-54 ml. Isopropanol (90 ml) was added to the mixture and then the resulting mixture was again concentrated to a volume of-54 ml. Isopropanol (135 ml) was added to the mixture followed by aqueous tris amine (3M; 5.0ml, 0.98 equiv). Stirring the resulting mixture/solution at ambient temperature; and within 15 minutes a solid precipitate began to form. The mixture was then stirred at ambient temperature for an additional 5 hours. The resulting mixture/slurry was cooled to 0 ℃ and the cooled slurry was stirred for an additional about 2 hours. The slurry was filtered and washed with cold isopropanol (3 × 15 ml). The collected solid was allowed to air dry on the collection funnel for about 90 minutes and then transferred to a vacuum oven to dry overnight. After 50 ℃/23inHg vacuum (with a small nitrogen bleed) for 16 hours, 8.66 gm of 10, 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium salt was obtained as a white solid; 99.8 area% (by UPLC) (yield: 12.5 mmol, 81%). To obtain LCMS and 1H NMR data substantially identical to those in the synthesis of 10S-1 shown previously.

In the light of the above example 10,1, 3-dihydroxyForms of (E) -2- (hydroxymethyl) propan-2-aminium saltsA (also referred to as Compound example) 10 form a) of the anhydrous tris salt powder X-ray diffraction (PXRD) data acquisition

PXRD analysis was performed on the white solid of the tris salt of example 10 (from Synthesis 10S-1 and Synthesis 10S-2) and was found to be a crystalline material (designated form A). 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 15 mm continuous illumination. The diffracted radiation was detected by a PSD-Lynx Eye detector with the detector PSD on set at 2.99 degrees. The X-ray tube voltage and current intensity were set at 40 kV and 40 mA, respectively. At Cu wavelength (CuK)= 1.5418 λ), data was collected from 3.0 to 40.0 degrees 2 θ in a θ - θ goniometer using a 0.01 degree step and a 1.0 second step time. The anti-scatter screen is set to a fixed distance of 1.5 mm. During data collection, the sample was rotated. Samples were prepared by placing them in a silicon low background sample holder and rotating them during collection. Data were collected and analyzed by EVA DIFFRAC Plus software using Bruker DIFFRAC Plus software. Prior to peak searching, the PXRD data file is unprocessed. The peak selected by threshold 1 is used for preliminary peak assignment using a peak search algorithm in the EVA software. To ensure effectiveness, the adjustment is made manually; the automatically assigned output is visually inspected and the peak position is adjusted to the peak maximum. The relative intensities are generally selected >Peak 3%. In general, unresolved peaks or peaks that coincide with noise are not selected. Typical errors associated with peak position over PXRD specified by USP are up to +/-0.2-theta (USP-941). Diffraction peaks expressed in degrees 2 theta and relative intensities (relative intensities) of PXRD of samples obtained from the previous synthesis of 10S-2>3%) is provided in table X2.

TABLE X2

Corner (2 theta) Relative Strength (%)
3.9 18.4
7.7 36.3
8.1 10.4
8.7 3.4
10.2 4.1
14.6 5.8
15.2 30.1
15.7 45.5
16.0 31.3
16.8 8.7
17.6 86.0
19.2 46.6
19.5 25.4
19.8 31.4
20.2 25.0
21.1 100.0
21.4 40.2
22.2 37.0
23.0 19.8
24.3 43.0
25.0 9.9
26.0 15.8
27.3 35.3
28.2 14.1
29.3 19.7
29.8 11.7
31.6 9.3
32.8 6.0
34.0 14.4
34.5 12.1
35.4 3.0
36.5 4.1

Example 11

1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxole- 4-radical]Piperidin-1-yl } methyl) -1H-benzimidazole-6-carboxylic acid, formate (11)

This entire synthesis sequence is performed in library form.

Step 1: 1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxole Penten-4-yl radical]Piperidin-1-yl } methyl) -1HSynthesis of methyl (C60) benzimidazole-6-carboxylate

A mixture of P14(44 mg, 100. mu. mol) and 3-ethynylpyridine (21 mg, 200. mu. mol) in toluene (800. mu.L) was treated with sodium bicarbonate (100. mu. mol) followed by triruthenium dodecacarbonyl (6 mg, 9. mu. mol). Then, the reaction flask is capped andshaking was carried out at 120 ℃ for 16 hours. Using Speedvac®The concentrator removed the solvent to afford C60, which was used directly in the next step.

Step 2: 1- (2-methoxyethyl) -2- ({4- [ 2-methyl-2- (pyridin-3-yl) -1, 3-benzodioxole Penten-4-yl radical]Piperidin-1-yl } methyl) -1HSynthesis of-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 flask was capped and shaken at 80 ℃ for 16 hours whereupon a Speedvac was used®The reaction mixture 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: 2.2 mg, 4.2. mu. mol, 4% (in two steps). LCMSm/z 529 [M+H]+. Retention time: 2.47 minutes (column: Waters Xbridge C18, 2.1X 50 mm, 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/minute).

Example 12

2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ]Piperidine-1- Methyl) -1- [2- (dimethylamino) ethyl]-1H-benzimidazole-6-carboxylic acid (12)

This entire synthesis sequence is performed in library form.

Step 1: 3- { [2- (dimethylamino) amino) Ethyl radical]Synthesis of methyl amino } -4-nitrobenzoate (C61)

By usingN,NDimethylethane-1, 2-diamine (18 mg, 200. mu. mol) andN,Ntreatment of methyl 3-fluoro-4-nitrobenzoate (78 mg, 600. mu. mol)N,N-a 0.2M solution in dimethylformamide; 1 mL, 200. mu. mol). The reaction flask was capped and shaken at 50 ℃ for 16 hours whereupon a Speedvac was used®The reaction mixture was evaporated to give C61. This material was used directly in the next step.

Step 2: 4-amino-3- { [2- (dimethylamino) ethyl]Synthesis of methyl amino } benzoate (C62)

The zinc powder was activated using dilute hydrochloric acid. Methanol (2 mL) was added to C61 (from previous step,. ltoreq.200. mu. mol), followed by an aqueous solution of calcium chloride (1.0M; 200. mu.L, 200. mu. mol) and activated zinc powder (130 mg, 2.0 mmol). The reaction flask was capped and shaken at 70 ℃ for 16 hours whereupon the reaction mixture was filtered. Using Speedvac®The filtrate was concentrated, and the residue was taken up in water (2 mL) and then extracted with ethyl acetate (2 × 3 mL). Using Speedvac ®The combined organic layers were evaporated to give C62 (estimated 150 μmol) which was used directly in the next step.

And step 3: 4- [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl)]Piperazine derivatives Pyridin-1-yl } acetyl) amino]-3- { [2- (dimethylamino) ethyl]Synthesis of methyl amino } benzoate (C63)

Compound P10(41 mg, 100. mu. mol) was added to C62 (from the previous step, approximately 150. mu. mol) and reacted with 2-hydroxypyridine 1-oxide and 1- [3- (dimethylamino) propyl]Method for producing (E) -3-ethylcarbodiimide hydrochlorideN,NDimethylacetamide solutions (0.1M each; 1 mL, 100. mu. mol each) to the mixture. Then, addN,NDiisopropylethylamine (39 mg, 300. mu. mol), and the reaction flask was capped and shaken at 50 ℃ for 16 hours. Then, Speedvac was used®A concentrator, concentrating the reaction mixture, and allowingPurification by preparative thin layer chromatography provided C63, which proceeded directly to the next step.

And 4, step 4: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [2- (dimethylamino) ethyl]-1HSynthesis of methyl (C64) benzimidazole-6-carboxylate

In a capped vial, a mixture of acetic acid (500. mu.L) and C63 (from the previous step,. ltoreq.100. mu. mol) was shaken for 2 hours at 150 ℃ whereupon Speedvac was used®The reaction mixture was evaporated. The resulting C64 proceeded directly to the following step.

And 5: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [2- (dimethylamino) ethyl]-1HSynthesis of (12) -benzimidazole-6-carboxylic acid

A solution of C64 (from the previous step,. ltoreq.100. mu. mol) in ethanol (500. mu.L) was treated with aqueous lithium hydroxide (2.0M; 500. mu.L, 1 mmol) 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, Speedvac was used®The resulting mixture was concentrated by a concentrator, 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% B]Purification to give 12. Yield: 7.0 mg, 12. mu. mol, 12% (in three steps). LCMSm/z 593 [M+H]+. Retention time: 2.45 minutes (column: Waters Xbridge C18, 2.1X 50 mm, 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]Piperidine-1- Methyl group) -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 group]Synthesis of methyl (C65) -5-nitropyridine-2-carboxylate

Triethylamine (532 mg, 5.26 mmol) was added to a suspension of 1- (1, 3-oxazol-2-yl) methylamine, hydrochloride salt (236 mg, 1.75 mmol) and methyl 6-chloro-5-nitropyridine-2-carboxylate (386 mg, 1.78 mmol) in tetrahydrofuran (5 mL). After the reaction mixture was stirred for 14 hours at 25 ℃, it was poured into water (30 mL) and extracted with dichloromethane (2 × 50 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo; silica gel chromatography (gradient: 0% to 5% methanol in dichloromethane) afforded C65 as a yellow solid. Yield: 310 mg, 1.11 mmol, 63%. LCMSm/z 278.7 [M+H]+1H NMR (400 MHz, chloroform-d) δ8.69 - 8.61 (br m, 1H), 8.58 (d, J = 8.4 Hz, 1H), 7.65 (d, J = 0.8 Hz, 1H), 7.46 (d, J = 8.4 Hz, 1H), 7.11 (d, J = 1.0 Hz, 1H), 5.07 (d, J = 5.3 Hz, 2H), 3.97 (s, 3H)。

The remainder of this synthesis sequence proceeds in library form.

Step 2: 5-amino-6- [ (1, 3-oxazol-2-ylmethyl) amino]Synthesis of pyridine-2-carboxylic acid methyl ester (C66)

Aqueous ammonium chloride (5.0M; 400. mu.L, 2.0 mmol) and activated zinc (131 mg, 2.0 mmol) were added sequentially to a solution of C65(56 mg, 200. mu. mol) in methanol (2.0 mL). Then, the reaction flask was capped and shaken at 30 ℃ for 16 hours, whereupon the reaction mixture was filtered. Using Speedvac ®Concentrator, concentrate filtrate, then mix it with water (1.0 mL) and extract with dichloromethane (3 × 1.0 mL); use ofSpeedvac®The combined organic layers were evaporated to afford C66, which was used directly in the next step.

And step 3: 5- [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl)]Piperazine derivatives Pyridin-1-yl } acetyl) amino]-6- [ (1, 3-oxazol-2-ylmethyl) amino]Synthesis of pyridine-2-carboxylic acid methyl ester (C67)

A mixture of P10(81 mg, 200. mu. mol) and C66 (from the previous step,. ltoreq.200. mu. mol) was reacted withN,N-dimethylacetamide and then withN,NTreatment with diisopropylethylamine (100. mu.L, 600. mu. mol). Adding 1- [3- (dimethylamino) propyl group]Of 3-ethylcarbodiimide hydrochloride (0.24M) and 2-hydroxypyridine 1-oxide (0.1M)N,N-solution in dimethylacetamide (1.0 mL, containing 240. mu. mol of 1- [3- (dimethylamino) propyl group]3-ethylcarbodiimide hydrochloride and 100. mu. mol of 2-hydroxypyridine 1-oxide), and the reaction flask was capped and shaken at 50 ℃ for 16 hours. Then, Speedvac was used®The concentrator, volatiles removed, and the residue was subjected to preparative thin layer chromatography to give C67, which was directly advanced to the next step.

And 4, step 4: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3HImidazo [4,5-b]Synthesis of pyridine-5-carboxylic acid methyl ester (C68) Become into

A mixture of acetic acid (1.0 mL) and C67 (from the previous step,. ltoreq.200. mu. mol) was shaken for 2 hours at 150 ℃ whereupon a Speedvac was used®The concentrator evaporates the reaction mixture. The resulting C68 was used directly in the following step.

And 5: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -3- (1, 3-oxazol-2-ylmethyl) -3HImidazo [4,5-b]Synthesis of pyridine-5-carboxylic acid (13)

Aqueous lithium hydroxide (2.0M; 1.0 mL, 2.0 mmol) was added to C68 (fromPrevious step,. ltoreq.200. mu. mol) in tetrahydrofuran (1.0 mL). After addition of methanol (500. mu.L), the reaction flask was capped and shaken at 50 ℃ for 16 hours. In the use of Speedvac®After the concentrator removed volatiles, dimethyl sulfoxide (1.0 mL) was added and the pH was adjusted to 7-8 using concentrated hydrochloric acid. Reverse phase HPLC was used [ column: agela Durashell C18, 5 μm; mobile phase A: ammonium hydroxide (in water) (pH 10); mobile phase B: acetonitrile; gradient: 24% to 64% B ]The resulting mixture was purified to give 13. Yield: 3.9 mg, 6.5. mu. mol, 3% (through four steps). LCMSm/z 604 [M+H]+. Retention time: 3.14 minutes (column: Waters Xbridge C18, 2.1X 50 mm, 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/minute).

Example 14

2-({4-[(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1-methyl-1H-benzimidazole-6-carboxylic acid (14)

Step 1: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1-methyl-1HSynthesis of methyl (C69) benzimidazole-6-carboxylate

Will be provided withN,N-diisopropylethylamine (683 μ L, 3.92 mmol) was added to a mixture of P3(680 mg, 1.31 mmol) in acetonitrile (5.2 mL); this was stirred at 45 ℃ for 5 minutes whereupon P16(319 mg, 1.34 mmol) was added. Stirring was continued for 2.75 hours at 45 ℃ and then water (6 mL) was added and stirred for 30 minutes before the reaction mixture was allowed to cool to room temperature. The solid was collected via filtration and washed with a mixture of acetonitrile and water (1: 4, 3 × 5 mL) to give C69 as a white solid. Yield: 635 mg, 1.15 mmol, 88%. LCMS m/z 550.1♦ [M+H]+1H NMR (400 MHz, chloroform-d) δ 8.15 - 8.12 (m, 1H), 7.97 (dd, J = 8.5, 1.6 Hz, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.50 (dd, J = 8.2, 8.2 Hz, 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 s, 3H), 1.89 - 1.78 (m, 4H)。

Step 2: 2- ({4- [(2S) -2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]Piperidin-1-yl } methyl) -1-methyl-1HSynthesis of (14) -benzimidazole-6-carboxylic acid

A mixture of C69(600 mg, 1.09 mmol) in methanol (11 mL) was heated to 45 ℃ and then treated with aqueous sodium hydroxide (1M; 2.2 mL, 2.2 mmol). After 24 hours, the reaction mixture was adjusted to pH 5-6 via addition of aqueous citric acid (1M; 1.1 mL) 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 via filtration and washed with a mixture of methanol and water (1: 4; 3 × 5 mL). This provided 14 as a white solid. Yield: 535 mg, 0.998 mmol, 92%. LCMSm/z536.1♦ [M+H]+1H NMR (400 MHz, DMSO-d 6)δ8.16 (d, J = 1.5 Hz, 1H), 7.81 (dd, J = 8.4, 1.6 Hz, 1H), 7.64 (d, J = 8.4 Hz, 1H), 7.59 - 7.52 (m, 2H), 7.33 (dd, J = 8.3, 2.1 Hz, 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) -1HBenzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate (15) [ from P18, via C71](ii) a And 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxole-4- Base of]-6-azaspiro [2.5]Oct-1-yl } -1- (2-methoxyethyl) -1 H-benzimidazole-6-carboxylic acid, DIAST-X2, trifluro Acetate (16) [ from P18, Via C72]

Step 1: 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 (C70) [ from P18]Of (2) Become into

A mixture of P18(240 mg, 0.699 mmol), C4 (275 mg, 0.800 mmol), cesium carbonate (455 mg, 1.40 mmol), tris (dibenzylideneacetone) dipalladium (0) (40.0 mg, 43.7. mu. mol), and 1,1 '-biphenyl-2, 2' -diylbis (diphenylphospholane) (BINAP; 52.2 mg, 83.8. mu. mol) in toluene (5 mL) 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 as a mixture of diastereomers as a yellow oil. Yield: 165 mg, 0.272 mmol, 39%. LCMSm/z 628.1♦ [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](ii) a And 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl ]6-nitrogen Hetero spiro [2.5 ]]Oct-1-yl } -1- (2-methoxyethyl) -1H-benzimidazole-6-carboxylic acid methyl ester, DIAST-Y2(C72) [ to From P18]Separation of

SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65: 35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ], C70(165 mg, 0.272 mmol) was used for the separation of stereoisomers on dioxolane. The first eluted isomer was designated DIAST-Y1(C71) and the second eluted isomer was designated DIAST-Y2 (C72); both were isolated as white solids.

Yield of C71: 55.0 mg, 90.7. mu. mol, 33%. LCMSm/z 605.9♦ [M+H]+. Retention time 4.47 min (column: Chiral Technologies Chiralpak AD-3, 4.6 x 100 mm, 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 maintained at 40% B for 2.5 min; flow rate: 2.8 mL/min).

Yield of C72: 58.0 mg, 95.7. mu. mol, 35%. LCMSm/z 628.0♦ [M+Na+]. Retention time 4.88 minutes (analytical conditions identical to those for C71).

And step 3: 2- {6- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]-6- Azaspiro [2.5 ]]Oct-1-yl } -1- (2-methoxyethyl) -1 H-benzimidazole-6-carboxylic acid, DIAST-X1, trifluoroacetate salt (15) [ from P18, via C71]Synthesis of (2)

Aqueous sodium hydroxide (3M; 1.0 mL, 3.0 mmol) was added to a solution of C71(55.0 mg, 90.7. mu. mol) in a mixture of methanol (2.0 mL) 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) afforded 15 as a white solid. Yield: 35.8 mg, 50.7 μmol, 56%. LCMSm/z 592.3♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.46 (s, 1H), 8.21 (d, J = 8.6 Hz, 1H), 7.78 (d, J = 8.6 Hz, 1H), 7.54 (dd, J = 8.3, 8.3 Hz, 1H), 7.16 - 7.08 (m, 2H), 6.76 (dd, J= 8.2, 8.1 Hz, 1H), 6.55-6.47 (m, 2H), 4.9-4.70 (m, 2H, assumed; partially obscured by a water peak), 3.82 (t,J = 4.9 Hz, 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)。

and 4, step 4: 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 salt (16) [ from P18, via C72 ]Synthesis of (2)

Aqueous sodium hydroxide (3M; 1.0 mL, 3.0 mmol) was added to a solution of C72(58.0 mg, 95.7. mu. mol) in a mixture of methanol (2.0 mL) 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) afforded 16 as a white solid. Yield: 33.4 mg, 47.3. mu. mol, 49%. LCMSm/z 592.2♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ8.53 - 8.50 (m, 1H), 8.25 (dd, J = 8.6, 1.4 Hz, 1H), 7.80 (br d, J = 8.6 Hz, 1H), 7.57 (dd, J = 8.4, 8.2 Hz, 1H), 7.25 (dd, J = 10.8, 2.0 Hz, 1H), 7.19 (br dd, J = 8.4, 2.1 Hz, 1H), 6.77 (dd, J= 8.2, 8.1 Hz, 1H), 6.55-6.50 (m, 2H), 4.9-4.72 (m, 2H, assumed; partially obscured by water peaks), 3.93-3.80 (m, 2H), 3.68-3.58 (m, 1H), 3.41-3.3 (m, 1H, hypothetical; 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, 5.8 Hz, 1H), 2.28 - 2.17 (m, 1H), 2.01 (d, J = 1.0 Hz, 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]Piperidine-1- Methyl) -1- [ (1-ethyl-1) H-imidazol-5-yl) methyl]-1HBenzimidazole-6-ammonium formate, 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-1)H-imidazol-5-yl) methyl]-1H-benzimidazole-6-ammonium formate, ENT-2 (18)

Step 1: 4- [ ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl)]Piperazine derivatives Pyridin-1-yl } acetyl) amino]-3- { [ (1-ethyl-1)H-imidazol-5-yl) methyl]Synthesis of methyl amino } benzoate (C73) Become into

Will be provided withO- (7-azabenzotriazol-1-yl) -N,N,N’,N’Tetramethyluronium hexafluorophosphate (566 mg, 1.49 mmol) was added to P19(340 mg, 1.24 mmol) inN,N-dimethylformamide (10 mL) and the mixture is stirred for 10 minutes at 25 ℃. Then, P10(503 mg, 1.24 mmol) andN,Ndiisopropylethylamine (615. mu.L, 3.53 mmol) inN,N-solution in dimethylformamide (7.7 mL) and the reaction mixture was stirred at 25 ℃ for 16 hours whereupon it was poured into water (10 mL) and extracted with ethyl acetate (3 × 50 mL). The combined organic layers were washed successively with aqueous ammonium chloride (3 × 20 mL) and saturated aqueous sodium chloride (2 × 20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. After purification using silica gel chromatography (gradient: 0% to 5% methanol in ethyl acetate), C73 was obtained as a light brown gum. Yield: 316 mg, 0.477 mmol, 38%. LCMS m/z 662.2♦ [M+H]+

Step 2: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl]-1H-benzimidazole-6-carboxylic acid methyl ester (C74) complex Become into

A solution of C73(316 mg, 0.477 mmol) in acetic acid (14 mL) was stirred at 55 ℃ for 16 h. Under high vacuum, the solvent was removed and the residue was purified using preparative thin layer chromatography (eluent: 10: 1 dichloromethane/methanol) to give C74 as a colorless oil. Yield: 200 mg, 0.310 mmol, 65%. LCMSm/z 644.3♦ [M+H]+

And step 3: 2- ({4- [2- (4-chloro-2-fluorophenyl) -2-methyl-1, 3-benzodioxol-4-yl]Piperazine derivatives Pyridin-1-yl } methyl) -1- [ (1-ethyl-1H-imidazol-5-yl) methyl]-1H-benzimidazole-6-ammonium formate, 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-1) yl ] -1- [ (1-ethyl-1)H-imidazol-5-yl) methyl]-1HSynthesis of (E) -benzimidazole-6-ammonium formate, ENT-2(18)

A mixture of C74(150 mg, 0.233 mmol) and aqueous sodium hydroxide (2M; 233. mu.L, 0.466 mmol) in a mixture of methanol (3 mL) and tetrahydrofuran (3 mL) was stirred at 45 ℃ for 16 h. After the reaction mixture was adjusted to pH 7 by addition of 1M hydrochloric acid, it was concentrated in vacuo to give a mixture of 17 and 18. Via SFC [ column: chiral Technologies Chiral cel OD, 10 μm; mobile phase: 1: 1 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide) ], separating these enantiomers. The first eluted enantiomer was designated ENT-1(17) and the second eluted enantiomer was designated ENT-2(18), both isolated as white solids.

17 yield: 45.0 mg, 69.5. mu. mol, 30%. LCMSm/z 630.3♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ 8.15 (br s, 1H), 8.00 (br d, J = 8.4 Hz, 1H), 7.81 (s, 1H), 7.72 (d, J = 8.2 Hz, 1H), 7.56 (dd, J = 8.3, 8.3 Hz, 1H), 7.28 (dd, J = 10.9, 2.0 Hz, 1H), 7.21 (dd, J= 8.3, 2.1 Hz, 1H), 6.77 (dd, components of ABC mode,J= 8.0, 7.7 Hz, 1H), 6.69 (dd, components of ABC mode,J = 7.8, 1.2 Hz, 1H), 6.67 - 6.60 (m, 2H), 5.82 (s, 2H), 4.12 (q, J= 7.2 Hz, 2H), 3.89 (AB quartet,J AB = 14.3 Hz, ΔνAB = 6.9 Hz, 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= 7.3 Hz, 3H). Retention time 5.66 min (column: Chiral Technologies Chiralpak AD-3, 4.6 x 150 mm, 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 maintained at 40% B for 3.0 min; flow rate: 2.5 mL/min).

18 yield: 32.8 mg, 50.7. mu. mol, 22%. LCMSm/z 630.3♦ [M+H]+1H NMR (400 MHz, methanol-d 4)δ 8.15 (s, 1H), 8.00 (d, J = 8.5 Hz, 1H), 7.81 (s, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.56 (dd, J = 8.3, 8.3 Hz, 1H), 7.28 (dd, J = 10.9, 2.0 Hz, 1H), 7.21 (dd, J= 8.3, 2.0 Hz, 1H), 6.77 (dd, components of ABC mode,J= 7.8, 7.8 Hz, 1H), 6.69 (dd, components of ABC mode,J = 7.9, 1.2 Hz, 1H), 6.67 - 6.60 (m, 2H), 5.82 (s, 2H), 4.12 (q, J= 7.3 Hz, 2H), 3.89 (AB quartet,J AB = 14.1 Hz, ΔνAB = 7.4 Hz, 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= 7.3 Hz, 3H). Retention time 5.34 minutes (analytical SFC conditions identical to those used for 17).

The compounds listed in table 1 were prepared using procedures analogous to the examples identified in table 2, using the appropriate intermediates identified in table 2. Using the methods discussed herein, the compound is purified. The final compound can be isolated as a neutral or acid or base salt.

Table 1: structures and IUPAC names of examples 19-102

Table 2: preparation and materialization data for examples 19-102

Examples Numbering Method 1H NMR (400 MHz, methanol-d 4) Delta, Mass Spectrometry, ions observed m/z [M+H]+Or HPLC retention time; mass spectrometrym/z [M+H]+(unless otherwise indicated Point out)
19 Examples 15 and 16; C4, P12 8.39 (br s, 1H), 8.08 (d, J = 8.5 Hz, 1H), 7.81 (d, J = 8.6 Hz, 1H), 7.60 (dd, J = 8.3, 8.3 Hz, 1H), 7.28 (dd, J = 10.9, 2.1 Hz, 1H), 7.22 (dd, J = 8.4, 2.0 Hz, 1H), 6.83 (dd, J = 8.1, 8.1 Hz, 1H), 6.60 (d, J = 7.8 Hz, 1H), 6.55 (d, J = 8.4 Hz, 1H), 4.73 (s, 2H), 4.66 (t, J = 4.9 Hz, 2H), 3.77 (t, J = 4.8 Hz, 2H), 3.59 – 3.43 (m, 8H), 3.30 (s, 3H^^), 2.05 (s, 3H); 581.0
20 example 4 and 51; C43, P11 8.34 – 8.31 (m, 1H), 8.03 (dd, J = 8.5, 1.5 Hz, 1H), 7.78 (d, J = 8.6 Hz, 1H), 7.60 (dd, J = 8.0, 8.0 Hz, 1H), 7.36 (dd, J = 10.2, 1.9 Hz, 1H), 7.31 (dd, J = 8.4, 1.8 Hz, 1H), 7.25 (s, 1H) 6.90 (dd, the components of ABC mode,J = 8.9, 6.6 Hz, 1H), 6.86 – 6.80 (m, 2H), 4.79 (s, 2H), 4.60 (br t, J = 4.8 Hz, 2H), 3.95 – 3.85 (m, 2H), 3.74 (dd, J = 5.3, 4.2 Hz, 2H), 3.44 – 3.33 (m, 2H), 3.28 (s, 3H), 3.15 – 3.05 (m, 1H), 2.37 – 2.12 (m, 4H); 566.0♦
21 example 4 and 51; C43, P11 8.32 (dd, J = 1.6, 0.7 Hz, 1H), 8.03 (dd, J = 8.5, 1.5 Hz, 1H), 7.78 (dd, J = 8.5, 0.7 Hz, 1H), 7.60 (dd, J = 8.1, 7.9 Hz, 1H), 7.36 (dd, J = 10.2, 2.0 Hz, 1H), 7.31 (br dd, J = 8.3, 1.8 hz, 1H), 7.25 (s, 1H), 6.90 (dd, the components of ABC mode,J = 8.8, 6.7 Hz, 1H), 6.87 – 6.80 (m, 2H), 4.79 (s, 2H), 4.60 (t, J = 4.8 Hz, 2H), 3.90 (br d, J = 12.3 Hz, 2H), 3.74 (dd, J = 5.3, 4.2 Hz, 2H), 3.38 (br dd, J = 12.6, 12.5 Hz, 2H), 3.28 (s, 3H), 3.10 (tt, J = 11.9, 4.0 Hz, 1H), 2.37 – 2.11 (m, 4H); 566.0♦
22 examples 1 and 2; P12, P5 8.37 (d, J = 1.5 Hz, 1H), 8.07 (dd, J = 8.5, 1.5 Hz, 1H), 7.79 (d, J = 8.6 Hz, 1H), 7.60 – 7.54 (m, 2H), 7.50 – 7.42 (m, 3H), 6.98 (s, 1H), 6.86 (dd, J = 8.1, 8.1 Hz, 1H), 6.61 (dd, J = 7.9, 0.9 Hz, 1H), 6.59 (dd, J = 8.4, 0.9 Hz, 1H), 4.73 (s, 2H), 4.64 (t, J = 4.8 Hz, 2H), 3.75 (dd, J = 5.4, 4.3 Hz, 2H), 3.61 – 3.44 (m, 8H), 3.28 (s, 3H); 515.1
23 examples 1 and 2; P12, P6 8.37 (br s, 1H), 8.07 (dd, J = 8.6, 1.5 Hz, 1H), 7.79 (d, J = 8.5 Hz, 1H), 7.61 – 7.54 (m, 2H), 7.51 – 7.42 (m, 3H), 6.98 (s, 1H), 6.86 (dd, J = 8.2, 8.1 Hz, 1H), 6.61 (br d, J = 8 Hz, 1H), 6.59 (br d, J = 8.5 Hz, 1H), 4.69 (s, 2H), 4.64 (t, J = 4.9 Hz, 2H), 3.75 (t, J = 4.9 Hz, 2H), 3.59 – 3.43 (m, 8H), 3.29 (s, 3H); 515.1
24 examples 4 and 5; C13, P11 8.33 (dd, J = 1.5, 0.6 Hz, 1H), 8.03 (dd, J = 8.5, 1.5 Hz, 1H), 7.79 (dd, J = 8.5, 0.5 Hz, 1H), 7.62 (dd, J = 8.4, 8.3 Hz, 1H), 7.29 (dd, J = 10.9, 2.0 Hz, 1H), 7.22 (ddd, J = 8.4, 2.0, 0.7 Hz, 1H), 6.88 – 6.82 (m, 1H), 6.82 – 6.76 (m, 2H), 4.83 (s, 2H), 4.63 (t, J = 4.8 Hz, 2H), 3.98 – 3.88 (m, 2H), 3.75 (dd, J = 5.3, 4.2 Hz, 2H), 3.47 – 3.36 (m, 2H), 3.31 (s, 3H^^), 3.10 (tt, J = 12.0, 4.1 Hz, 1H), 2.36 – 2.10 (m, 4H), 2.05 (d, J = 1.0 Hz, 3H); 580.1♦
25 examples 15 and 162; C4, P17 8.51 (dd, J = 1.5, 0.7 Hz, 1H), 8.25 (dd, J = 8.6, 1.4 Hz, 1H), 7.79 (dd, J = 8.6, 0.7 Hz, 1H), 7.57 (dd, J = 8.3, 8.3 Hz, 1H), 7.25 (dd, J = 10.8, 2.0 Hz, 1H), 7.19 (ddd, J = 8.4, 2.0, 0.7 Hz, 1H), 6.80 – 6.73 (m, 1H), 6.55 – 6.50 (m, 2H), 4.9 – 4.73 (m, 2H^), 3.92 – 3.81 (m, 2H), 3.66 – 3.58 (m, 1H), 3.41 – 3.3 (m, 1H^^), 3.25 (s, 3H), 3.20 – 3.12 (m, 1H), 3.05 – 2.97 (m, 1H), 2.70 – 2.63 (m, 1H), 2.27 – 2.17 (m, 1H), 2.01 (d, J = 1.0 Hz, 3H), 1.84 – 1.71 (m, 2H), 1.67 – 1.58 (m, 2H), 1.31 (br d, J = 13 Hz, 1H); 592.3♦
26 examples 15 and 162; C4, P17 8.53 – 8.50 (m, 1H), 8.26 (dd, J = 8.6, 1.4 Hz, 1H), 7.81 (d, J = 8.6 Hz, 1H), 7.55 (dd, J = 8.3, 8.2 Hz, 1H), 7.16 – 7.08 (m, 2H), 6.77 (dd, J = 8.3, 7.9 Hz, 1H), 6.52 (br d, J = 8.3 Hz, 1H), 6.51 (br d, J = 7.7 Hz, 1H), 4.9 – 4.74 (m, 2H^), 3.83 (t, J = 4.8 Hz, 2H), 3.68 – 3.60 (m, 1H), 3.54 – 3.46 (m, 1H), 3.18 – 3.09 (m, 1H), 3.14 (s, 3H), 3.09 – 3.01 (m, 1H), 2.69 – 2.62 (m, 1H), 2.31 – 2.21 (m, 1H), 2.01 (br s, 3H), 1.78 – 1.69 (m, 2H), 1.63 – 1.52 (m, 2H), 1.33 – 1.25 (m, 1H); 592.3♦
27 example 4 and 53; P11 8.32 (br s, 1H), 8.02 (dd, J = 8.5, 1.5 Hz, 1H), 7.82 – 7.76 (m, 2H), 7.73 (br d, J = 10.0 Hz, 1H), 7.67 (br d, J = 8.0 Hz, 1H), 7.35 (s, 1H), 6.96 – 6.89 (m, 1H), 6.88 – 6.83 (m, 2H), 4.76 (s, 2H), 4.61 (t, J = 4.8 Hz, 2H), 3.87 (br d, J = 12.3 Hz, 2H), 3.74 (t, J = 4.8 Hz, 2H), 3.39 – 3.3 (m, 2H^^), 3.29 (s, 3H), 3.15 – 3.05 (m, 1H), 2.35 – 2.10 (m, 4H); 557.1
28 example 11; P14 3.08 minutes4; 596
29 Example 11; P14 3.12 minutes4; 556
30 Example 11; P14 2.90 minutes4; 576
31 Example 11; P14 2.92 minutes4; 546
32 Example 11; P14 2.88 minutes4; 558
33 Example 11; P14 3.04 minutes4; 562
34 Example 11; P14 2.99 minutes5; 553
35 Example 11; P14 2.92 minutes4; 576
36 Example 11; P14 2.81 minutes5; 543
37 Example 11; P14 2.90 minutes4; 558
38 Example 11; P14 2.91 minutes4; 546
39 Example 11; P14 2.89 minutes4; 558
40 Example 11; P14 3.11 minutes4; 596
41 Example 11; P14 2.97 minutes4; 564
42 Example 11; P14 2.40 minutes5; 543
43 Example 12; P10 2.70 minutes4; 621
44 Example 12; P10 2.49 minutes4; 635
45 Example 12; P10 2.79 minutes4; 613
46 Example 12; P10 2.71 minutes4; 635
47 Example 12; P10 2.85 minutes4; 657
48 Example 12; P10 2.71 minutes4; 633
49 Example 12; P10 2.66 minutes4; 607
50 Example 12; P10 2.43 minutes4; 616
51 Example 12; P10 2.74 minutes4; 630
52 Example 12; P10 2.73 minutes4; 593
53 Example 12; P10 2.79 minutes 4; 616
54 Example 12; P10 2.67 minutes4; 631
55 Example 12; P10 2.44 minutes4; 630
56 Example 12; P10 2.77 minutes4; 606
57 Example 12; P10 2.72 minutes4; 617
58 Example 12; P10 2.78 minutes4; 603
59 Example 12; P10 2.82 minutes4; 621
60 Example 12; P10 2.74 minutes4; 631
61 Example 12; P10 2.76 minutes4; 592
62 Example 12; P10 2.45 minutes4; 630
63 Example 12; P10 2.78 minutes4; 617
64 Example 12; P10 2.84 minutes4; 606
65 Example 12; P10 2.56 minutes4; 613
66 Example 12; P10 2.75 minutes4; 607
67 Example 12; P10 2.48 minutes4; 619
68 Example 12; P10 2.75 minutes4; 646
69 Example 12; P10 2.73 minutes4; 603
70 Example 12; P10 2.86 minutes5; 661
71 Example 12; P10 2.77 minutes4; 657
72 Example 12; P10 2.79 minutes4; 606
73 Example 12; P10 2.70 minutes4; 602
74 Example 12; P10 2.45 minutes4; 616
75 Example 13; P10 2.92 minutes4; 566
76 Example 13; P10 2.99 minutes4; 631
77 Example 13; P10 2.94 minutes4; 616
78 Example 13; P10 3.08 minutes5; 617
79 Example 13; P10 3.09 minutes4; 606
80 Example 13; P10 3.02 minutes4; 603
81 Example 13; P10 3.10 minutes5; 604
82 Example 11; P14 2.87 minutes4; 528
83 Example 11; P14 3.00 minutes4; 592
84 Example 11; P14 2.99 minutes4; 542
85 Example 11; P14 2.98 minutes4; 542
86 Example 11; P14 2.97 minutes4; 562
87 Example 11; P14 2.97 minutes5; 553
88 Example 11; P14 2.90 minutes4; 542
89 Example 4 and 56,7; P11 8.63 (ddd, J = 4.9, 1.8, 0.9 Hz, 1H), 8.34 (dd, J = 1.6, 0.7 Hz, 1H), 8.03 (dd, J = 8.5, 1.5 Hz, 1H), 7.90 (ddd, J = 7.8, 7.8, 1.7 Hz, 1H), 7.80 (dd, J = 8.5, 0.7 Hz, 1H), 7.74 (ddd, J = 7.9, 1.1, 1.0 Hz, 1H), 7.45 (ddd, J = 7.6, 4.9, 1.2 Hz, 1H), 6.88 – 6.83 (m, 1H), 6.83 – 6.76 (m, 2H), 4.83 (s, 2H), 4.63 (t, J = 4.8 Hz, 2H), 3.99 – 3.88 (m, 2H), 3.75 (dd, J = 5.3, 4.2 Hz, 2H), 3.45 – 3.34 (m, 2H), 3.31 (s, 3H), 3.15 – 3.03 (m, 1H), 2.41 – 2.20 (m, 2H), 2.19 – 2.08 (m, 2H), 2.05 (s, 3H); 529.3
90 example 4 and 56,7; P11 8.63 (ddd, J = 4.9, 1.8, 0.9 Hz, 1H), 8.34 (dd, J = 1.6, 0.7 Hz, 1H), 8.04 (dd, J = 8.5, 1.5 Hz, 1H), 7.90 (ddd, J = 7.8, 7.8, 1.7 Hz, 1H), 7.80 (dd, J = 8.5, 0.7 Hz, 1H), 7.73 (ddd, J = 8.0, 1.1, 1.0 Hz, 1H), 7.45 (ddd, J = 7.6, 4.9, 1.2 Hz, 1H), 6.88 – 6.83 (m, 1H), 6.83 – 6.75 (m, 2H), 4.83 (s, 2H), 4.63 (t, J = 4.9 Hz, 2H), 3.98 – 3.88 (m, 2H), 3.75 (t, J = 4.8 Hz, 2H), 3.44 – 3.34 (m, 2H), 3.32 (s, 3H^^), 3.15 – 3.03 (m, 1H), 2.40 – 2.19 (m, 2H), 2.18 – 2.08 (m, 2H), 2.05 (s, 3H); 529.3
91 examples 6 and 7; P8, P11 8.59 (d, J = 2.4 Hz, 1H), 8.26 (d, J = 1.4 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.87 (dd, J = 8.5, 2.5 Hz, 1H), 7.68 – 7.61 (m, 2H), 6.83 – 6.75 (m, 1H), 6.75 – 6.67 (m, 2H), 4.67 (t, J = 5.2 Hz, 2H), 4.00 (s, 2H), 3.82 (t, J = 5.1 Hz, 2H), 3.29 (s, 3H), 3.13 – 3.05 (m, 2H), 2.81 – 2.70 (m, 1H), 2.45 – 2.34 (m, 2H), 2.01 (s, 3H), 1.98 – 1.77 (m, 4H); 563.3♦
92 examples 6 and 7; P9, P11 8.59 (d, J = 2.3 Hz, 1H), 8.26 (d, J = 1.4 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.87 (dd, J = 8.5, 2.5 Hz, 1H), 7.68 – 7.62 (m, 2H), 6.82 – 6.76 (m, 1H), 6.74 – 6.68 (m, 2H), 4.67 (t, J = 5.2 Hz, 2H), 4.00 (s, 2H), 3.82 (t, J = 5.1 Hz, 2H), 3.29 (s, 3H), 3.13 – 3.04 (m, 2H), 2.76 (tt, J = 11.8, 4 Hz, 1H), 2.45 – 2.34 (m, 2H), 2.01 (s, 3H), 1.97 – 1.78 (m, 4H); 563.3♦
93 example 8 and 98; P8, P11 8.97 (dd, J = 2.1, 0.9 Hz, 1H), 8.27 – 8.25 (m, 1H), 8.21 (dd, J = 8.2, 2.1 Hz, 1H), 7.96 (dd, J = 8.5, 1.5 Hz, 1H), 7.81 (dd, J = 8.3, 0.9 Hz, 1H), 7.64 (d, J = 8.6 Hz, 1H), 6.83 – 6.77 (m, 1H), 6.76 – 6.68 (m, 2H), 4.68 (t, J = 5.2 Hz, 2H), 3.95 (s, 2H), 3.83 (t, J = 5.2 Hz, 2H), 3.30 (s, 3H), 3.08 – 2.99 (m, 2H), 2.79 – 2.69 (m, 1H), 2.39 – 2.28 (m, 2H), 2.04 (s, 3H), 1.96 – 1.76 (m, 4H); 554.4
94 example 8 and 98; P9, P11 8.97 (dd, J = 2.2, 0.9 Hz, 1H), 8.26 (br s, 1H), 8.21 (dd, J = 8.2, 2.1 Hz, 1H), 7.96 (dd, J = 8.4, 1.4 Hz, 1H), 7.81 (dd, J = 8.2, 0.9 Hz, 1H), 7.64 (br d, J = 8.5 Hz, 1H), 6.83 – 6.77 (m, 1H), 6.76 – 6.69 (m, 2H), 4.68 (t, J = 5.3 Hz, 2H), 3.95 (s, 2H), 3.83 (t, J = 5.2 Hz, 2H), 3.30 (s, 3H), 3.08 – 2.99 (m, 2H), 2.79 – 2.69 (m, 1H), 2.39 – 2.28 (m, 2H), 2.04 (s, 3H), 1.96 – 1.76 (m, 4H); 554.4
95 example 10; P8, P15 8.61 (dd, J = 2.5, 0.7 Hz, 1H), 8.41 (s, 1H), 8.33 (dd, J = 1.6, 0.7 Hz, 1H), 7.97 (dd, J = 8.5, 1.5 Hz, 1H), 7.88 (dd, J = 8.5, 2.5 Hz, 1H), 7.66 (dd, J = 8.5, 0.6 Hz, 1H), 7.65 (dd, J = 8.5, 0.7 Hz, 1H), 6.82 – 6.77 (m, 1H), 6.76 – 6.69 (m, 2H), 5.32 – 5.24 (m, 1H), 4.9 – 4.83 (m, 1H^), 4.71 (dd, J = 15.4, 2.6 Hz, 1H), 4.65 – 4.58 (m, 1H), 4.48 (ddd, J = 9.2, 6.0, 5.9 Hz, 1H) 4.03 (AB quartet,J AB = 13.9 Hz, ΔνAB = 49.7 Hz, 2H), 3.18 – 3.11 (m, 1H), 3.06 – 2.98 (m, 1H), 2.87 – 2.69 (m, 2H), 2.60 – 2.49 (m, 1H), 2.46 – 2.31 (m, 2H), 2.02 (s, 3H), 1.98 – 1.79 (m, 4H); 574.9♦
96 example 6 and 79,10; P15 7.01 minutes11; 610.5♦
97 Example 6 and 79,10; P15 7.89 minutes11; 610.5♦
98 C5412 8.25 – 8.23 (m, 1H), 8.00 (dd, J = 8.5, 1.5 Hz, 1H), 7.89 (d, J = 0.9 Hz, 1H), 7.76 (dd, J = 7.9, 7.6 Hz, 1H), 7.70 (d, J = 8.5 Hz, 1H), 7.64 (dd, J = 10.6, 1.5 Hz, 1H), 7.57 (dd, J = 8.0, 1.5 Hz, 1H), 7.14 (d, J = 0.9 Hz, 1H), 6.78 (dd, the components of ABC mode, J = 7.9, 7.8 Hz, 1H), 6.70 (dd, components of ABC mode,J = 7.8, 1.2 Hz, 1H), 6.66 (br d, components of ABC mode,J = 7.9 Hz, 1H), 5.94 (AB quartet of peaks,J AB = 17.2 Hz, ΔνAB = 6.5 Hz, 2H), 3.96 (s, 2H), 3.02 – 2.92 (m, 2H), 2.74 – 2.63 (m, 1H), 2.31 – 2.21 (m, 2H), 2.05 (br s, 3H), 1.80 – 1.58 (m, 4H); 594.3
99 C5412 8.23 (d, J = 1.4 Hz, 1H), 8.00 (dd, J = 8.5, 1.5 Hz, 1H), 7.89 (d, J = 0.9 Hz, 1H), 7.76 (dd, J = 7.9, 7.6 Hz, 1H), 7.69 (d, J = 8.5 Hz, 1H), 7.64 (dd, J = 10.6, 1.5 Hz, 1H), 7.57 (dd, J = 8.1, 1.5 Hz, 1H), 7.14 (d, J = 0.9 Hz, 1H), 6.78 (dd, the components of ABC mode,J = 7.8, 7.8 Hz, 1H), 6.70 (dd, components of ABC mode,J = 7.8, 1.2 Hz, 1H), 6.66 (br d, components of ABC mode,J = 7.9 Hz, 1H), 5.94 (AB quartet of peaks,J AB = 17.1 Hz, ΔνAB = 6.6 Hz, 2H), 3.96 (s, 2H), 3.01 – 2.92 (m, 2H), 2.74 – 2.63 (m, 1H), 2.30 – 2.20 (m, 2H), 2.05 (br s, 3H), 1.80 – 1.58 (m, 4H); 594.3
100 example 7 game Acid dissociation13; P3, C29 Characteristic peak 7.80 (dd,J = 8.5, 6.6 Hz, 1H), 7.59 (dd, J = 8.3, 8.3 Hz, 1H), 7.51 (d, J = 8.6 Hz, 1H), 7.28 (dd, J = 10.9, 2.0 Hz, 1H), 7.21 (br dd, J = 8.4, 2.0 Hz, 1H), 6.83 – 6.77 (m, 1H), 6.76 – 6.71 (m, 2H), 5.32 – 5.23 (m, 1H), 4.99 (dd, J = 15.5, 7.1 Hz, 1H), 4.79 (dd, J = 15.6, 2.8 Hz, 1H), 4.72 – 4.63 (m, 1H), 4.47 (ddd, J = 9.1, 6.0, 6.0 Hz, 1H), 4.31 (AB quartet,J AB = 14.4 Hz, ΔνAB = 33.3 Hz, 2H), 3.40 (br d, J = 11.9 Hz, 1H), 2.92-2.65 (m, 4H), 2.82 (AB quadruple) The peak(s) of the peak(s),J AB = 15.5 Hz, ΔνAB = 37.5 Hz, 2H), 2.61 – 2.49 (m, 1H), 2.13 – 1.87 (m, 4H), 2.04 (s, 3H); 610.0♦
101 example 5 game Acid dissociation13; C48, C29 Characteristic peak 7.79 (dd,J = 8.5, 6.6 Hz, 1H), 7.57 (dd, J = 8.0, 8.0 Hz, 1H), 7.49 (d, J = 8.5 Hz, 1H), 7.35 (dd, J = 10.2, 1.9 Hz, 1H), 7.30 (br d, J = 8.4 Hz, 1H), 7.22 (s, 1H), 6.88 – 6.82 (m, 1H), 6.82 – 6.74 (m, 2H), 5.30 – 5.21 (m, 1H), 4.95 (dd, J = 15.4, 7.1 Hz, 1H), 4.77 (br d, J = 15.1 Hz, 1H), 4.67 – 4.59 (m, 1H), 4.44 (ddd, J = 9.1, 5.9, 5.9 Hz, 1H), 4.28 (AB quartet,J AB = 14.4 Hz, ΔνAB = 31.7 Hz, 2H), 3.37 (br d, J = 12.3 Hz, 1H^^), 2.92 – 2.61 (m, 4H), 2.82 (AB the peak of the four-fold peak is shown,J AB = 15.6 Hz, ΔνAB = 37.1 Hz, 2H), 2.58 – 2.47 (m, 1H), 2.12 – 1.89 (m, 4H); 596.1 ♦
102 example 4 and 514; P11 8.34 (dd, J = 1.6, 0.7 Hz, 1H), 8.04 (dd, J = 8.5, 1.5 Hz, 1H), 7.80 (dd, J = 8.5, 0.7 Hz, 1H), 7.66 – 7.60 (m, 2H), 7.46 – 7.36 (m, 3H), 6.84 – 6.76 (m, 2H), 6.74 (dd, J = 7.2, 2.0 Hz, 1H), 4.84 (s, 2H), 4.63 (t, J = 4.7 Hz, 2H), 4.01 – 3.91 (m, 4H), 3.76 (dd, J = 5.3, 4.2 Hz, 2H), 3.47 – 3.37 (m, 2H), 3.32 (s, 3H), 3.19 – 3.08 (m, 1H), 2.41 – 2.26 (m, 2H), 2.26 – 2.13 (m, 2H); 544.2

the area is assumed; the peak is partially shielded by the water peak

The area is assumed; the peak is partially masked by the solvent peak

♦ A chlorine isotope pattern was observed.

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]Via SFC [ column: chiral Technologies Chiral cel OD-H, 5 μm; mobile phase: 7: 3 carbon dioxide/(2-propanol containing 0.1% ammonium hydroxide)]Separated into its component enantiomers. The first eluted enantiomer, ENT-1(C76), was used in the synthesis of example 21 and the second eluted enantiomer, ENT-2 (C77), was converted to example 20. C76 retention time: 5.72 minutes (column: Chiral Technologies Chiralpak OD-3, 4.6X 150 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: 2-propanol with 0.05% diethylamine; gradient: 5% to 40% B over 5.5 minutes, then maintained at 40% B for 3.0 minutes; flow rate: 2.5 mL/min). C77 retention time: 6.01 minutes (analytical SFC conditions were the same as those used for C76).

2. Methyl ester derived from 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) via SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65: 35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]Separated into its constituent stereoisomers in dioxolane. The first eluted isomer DIAST-1(C78) was converted to example 26; according to1Examination of the H NMR data shows that this material is the enantiomer of example 15. The second eluted isomer DIAST-2(C79) was used for the synthesis of example 25; according to1Examination of the H NMR data shows that this material is the enantiomer of example 16. C78 retention time: 3.60 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 100 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol with 0.05% diethylamine; gradient: 5% to 40% B over 4.5 minutes, then maintained at 40% B for 2.5 minutes; flow rate: 2.8 mL/min). C79 retention time: 382 minutes (analytical SFC conditions were the same as those used for C78).

4- (4-bromo-1, 3-benzodioxol-2-yl) -3-fluorobenzonitrile is prepared via treatment of 3-fluoro-4-formylbenzonitrile and 3-bromobenzene-1, 2-diol with p-toluenesulfonic acid (in toluene) with removal of water 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 ] -palladium (II) dichloride and copper (I) iodide, followed by ester cleavage using p-toluenesulfonic acid to give the desired 3-fluoro-4- [4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] benzonitrile.

4. Conditions for analytical HPLC. Column: waters XBridge C18, 2.1 × 50 mm, 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.

5. Conditions for analytical HPLC. Column: waters XBridge C18, 2.1 × 50 mm, 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.

6. Synthesis of 4- [ 2-methyl-2- (pyridin-2-yl) -1, 3-benzodioxol-4-yl from 3-bromobenzene-1, 2-diol and 2-ethynylpyridine using the procedure described for the synthesis of C12 in preparation P7]-3, 6-dihydropyridine-1 (2)H) -tert-butyl formate. Followed by hydrogenation on palladium on charcoal and treatment with hydrogen chloride (in ethyl acetate) to give the desired 2- [ 2-methyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl]Pyridine, hydrochloride.

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]Via SFC [ column: chiral Technologies Chiral pak AD, 10 μm; mobile phase: 65: 35 carbon dioxide/(ethanol containing 0.1% ammonium hydroxide)]And separated into its constituent enantiomers. The first eluted enantiomer, ENT-1(C80), was usedThe synthesis of example 90 and the second eluted enantiomer, ENT-2(C81), was converted to example 89. C80 retention time: 4.11 minutes (column: Chiral Technologies Chiralpak AD-3, 4.6X 100 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: ethanol with 0.05% diethylamine; gradient: 5% to 40% B over 4.5 minutes, then maintained at 40% B for 2.5 minutes; flow rate: 2.8 mL/min). C81 retention time: 4.62 minutes (analytical SFC conditions identical to those used for C80).

8. The conversion of P8 and P9 to the corresponding cyano-substituted derivatives was carried out using the procedure described for the synthesis of P4 from P2 in the preparation of P4.

9. Treatment of 1- (4-chloro-2-fluorophenyl) ethanone with trimethyl orthoformate and p-toluenesulfonic acid provides 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 desired tert-butyl 4- [2- (4-chloro-2-fluorophenyl) -7-fluoro-2-methyl-1, 3-benzodioxol-4-yl ] piperidine-1-carboxylate using the procedure described in preparation P1 for the synthesis of P1 from C2.

10.96 and 97 separation of stereoisomers at dioxolane is carried out 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 isomer was designated DIAST-1(96) and the second eluting isomer was designated DIAST-2 (97).

11. Conditions for analytical SFC. Column: chiral Technologies Chiral IG, 4.6 × 100 mm, 5 μm; mobile phase: 7: 3 carbon dioxide/(2-propanol with 0.2% ammonium hydroxide); flow rate: 1.5 mL/min; back pressure: 150 bar.

12. Synthesis of 2- (chloromethyl) -1- (1, 3-oxazol-2-ylmethyl) -1 from tert-butyl 3-fluoro-4-nitrobenzoate and 1- (1, 3-oxazol-2-yl) methylamine using the method described for the synthesis of P11H-benzimidazole-6-carboxylic acid tert-butyl ester. Followed by reaction with C54 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) ]It is separated into its constituent enantiomers. The first eluted enantiomer, ENT-1(C82), was used for the synthesis of 99 and the second eluted enantiomer, ENT-2(C83), was converted to 98. C82 retention time: 1.47 min (column: Chiral Technologies Chiralpak OD-3, 4.6X 50 mm, 3 μm; mobile phase A: carbon dioxide; mobile phase B: methanol with 0.05% diethylamine; gradient: 5% B, 0.2 min, then 5% to 40% B over 1.4 min, then held at 40% B for 1.05 min; flow rate: 4 mL/min). C83 retention time: 1.85 minutes (analytical SFC conditions identical to those used for C82).

Reaction of 1-bromo-2, 3-difluoro-4-nitrobenzene with copper (I) cyanide in 1-methylpyrrolidin-2-one at elevated temperature provides 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 desired 2- (chloromethyl) -7-fluoro-1- [ (2-chloro methyl) -7-fluoro-1- [ (2) via the method described for the synthesis of P11 from methyl 3-fluoro-4-nitrobenzoate in preparation P11 by using C29S) -Oxetazedin-2-ylmethyl]-1H-benzimidazole-6-carboxylic acid methyl ester.

14. The desired [ 2-phenyl-4- (piperidin-4-yl) -1, 3-benzodioxol-2-yl ] methanol was synthesized from 2-oxo-2-phenylethyl acetate by analogy with the procedure described for the synthesis of C13.

CHO GLP-1R clone H6-assay 1

GLP-1R-mediated agonist activity was determined using a HTRF (homogeneous time-resolved fluorescence) cAMP detection Kit (cAMP HI Range Assay Kit; CisBio cat #62AM6PEJ) that measures cAMP levels in cells, as a cell-based functional Assay. The method is a competitive immunoassay between native cAMP produced by the cell and exogenous cAMP labeled with the dye d 2. Tracer binding was visualized by cryptate labeled mAb anti-cAMP. In standard or experimental samples, the specific signal (i.e., energy transfer) is inversely proportional to the cAMP concentration.

The Sequence encoding human GLP-1R (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 recipient was isolated (designated clone H6). Use of125I-GLP-17-36 Saturation binding analysis (filter assay procedure) of (Perkin Elmer) showed that plasma membranes derived from this cell line express high GLP-1R density (K)d: 0.4 nM, Bmax1900 fmol/mg protein).

Cells were removed from cryopreservation, resuspended in 40 mL of Dulbeco's phosphate buffered saline (DPBS-Lonza Cat # 17-512Q) and centrifuged at 800 x g for 5 minutes at 22 ℃. The cell pellet was then resuspended in 10 mL growth medium [ DMEM/F121: 1 mixture containing HEPES, L-Gln, 500 mL (DMEM/F12 Lonza Cat # 12-719F), 10% heat-inactivated fetal bovine serum (Gibco Cat # 16140-071), 5 mL of 100X Pen-Strep (Gibco Cat # 15140-122), 5 mL of 100X L-glutamine (Gibco Cat # 25030-081), and 500. mu.g/mL Geneticin (Geneticin) (G418) (Invitrogen #10131035) ]. Cell suspension samples in 1 mL of growth medium were counted on a Becton Dickinson ViCell to determine cell viability and cell count per mL. The remaining cell suspension was then conditioned with growth media to deliver 2000 viable cells per well using a Matrix Combi Multidrop reagent dispenser and the cells were dispensed into a white 384-well tissue culture treated assay plate (Corning 3570). The assay plates were then incubated for 48 hours at 37 ℃ in a humidified environment of 5% carbon dioxide.

Each test compound (in DMSO) was diluted in assay buffer (calcium/magnesium containing HBSS (Lonza/BioWhittaker cat # 10-527F)/0.1% BSA (Sigma Aldrich cat # A7409-1L)/20 mM HEPES (Lonza/BioWhittaker cat #17-737E)) containing 100. mu.M 3-isobutyl-1-methylxanthine (IBMX; Sigma cat # I5879) at various concentrations. The final DMSO concentration was 1%.

After 48 hours, growth medium was removed from the assay plate wells and serially diluted in 20 μ L of assay buffer at 37 ℃ in a humidified environment of 5% carbon dioxideThe cells were treated with the compound for 30 minutes. After 30 min incubation, 10. mu.L of labeled d2 cAMP and 10. mu.L of anti-cAMP antibody (both diluted 1: 20 in cell lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. Then, the plates were incubated at room temperature and after 60 minutes, changes in HTRF signal were read using excitation at 330 nm and emission at 615 and 665 nm with an Envision 2104 multi-label plate reader. Raw data were converted to nM cAMP from cAMP standard curve by interpolation (as described by the manufacturer's assay protocol) and full agonist GLP-1 versus saturation concentration included in each plate 7-36(1. mu.M) the percent effect was determined. EC was performed from agonist dose-response curves analyzed by curve fitting procedure using the 4-parameter logistic dose-response equation50And (4) determining.

CHO GLP-1R clone C6-assay 2

GLP-1R-mediated agonist activity was determined using a HTRF (homogeneous time-resolved fluorescence) cAMP detection Kit (cAMP HI Range Assay Kit; Cis Bio cat #62AM6PEJ) that measures cAMP levels in cells, in a cell-based functional Assay. The method is a competitive immunoassay between native cAMP produced by the cell and exogenous cAMP labeled with the dye d 2. Tracer binding was visualized by cryptate labeled mAb anti-cAMP. In standard or experimental samples, the specific signal (i.e., energy transfer) is inversely proportional to the cAMP concentration.

The Sequence encoding human GLP-1R (NCBI Reference Sequence NP-002053.3, including the naturally occurring variant Leu260Phe) was subcloned into pcDNA5-FRT-TO and a Flp-In T-Rex system was used TO isolate a clonal CHO cell line stably expressing low receptor densities 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: 0.3 nM, Bmax240 fmol/mg protein) relative to clone H6 cell line.

Cells were removed from cryopreservation and resuspended in 40 mL of Dulbeco's phosphate buffered saline (DPBS-Lonza Cat #)17-512Q) and centrifuged at 800 x g for 5 minutes at 22 ℃. DPBS was aspirated and the cell pellet resuspended in 10 mL of complete growth medium (DMEM: F121: 1 mixture containing HEPES, L-Gln, 500 mL (DMEM/F12 Lonza Cat # 12-719F), 10% heat-inactivated fetal bovine serum (Gibco Cat # 16140-071), 5 mL of 100X Pen-Strep (Gibco Cat # 15140-122), 5 mL of 100X L-glutamine (Gibco Cat # 25030-081), 700. mu.g/mL hygromycin (Invitrogen Cat # 10687010) and 15. mu.g/mL blasticidin (Gibco Cat # R21001)). Cell suspension samples in 1 mL of growth medium were counted on a Becton Dickinson ViCell to determine cell viability and cell count per mL. The remaining cell suspension was then conditioned with growth medium to deliver 1600 viable cells per well using a Matrix Combi Multidrop reagent dispenser and the cells were dispensed into a white 384-well tissue culture treated assay plate (Corning 3570). Then, at 37 ℃ in a humid environment (95% O) 2, 5% CO2) The assay plates were incubated for 48 hours.

Test compounds were diluted in DMSO at various concentrations in assay buffer [ calcium/magnesium containing HBSS (Lonza/BioWhittaker cat # 10-527F)/0.1% BSA (Sigma Aldrich cat # A7409-1L)/20 mM 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, growth medium was removed from assay plate wells and incubated at 37 ℃ in a humid environment (95% O)2, 5% CO2) Cells were treated with 20 μ L of serially diluted compounds in assay buffer for 30 min. After 30 min incubation, 10. mu.L of labeled d2 cAMP and 10. mu.L of anti-cAMP antibody (both diluted 1: 20 in cell lysis buffer; as described in the manufacturer's assay protocol) were added to each well of the assay plate. Then, the plates were incubated at room temperature and after 60 minutes, changes in HTRF signal were read using excitation at 330 nm and emission at 615 and 665 nm with an Envision 2104 multi-label plate reader. Raw data were converted to nM cAMP from cAMP standard curve by interpolation (as described in the manufacturer's assay protocol) and plotted against package The percent effect was determined by the saturation concentration of the full agonist GLP-1 (1. mu.M) included in each plate. EC was performed from agonist dose-response curves analyzed by curve fitting procedure using the 4-parameter logistic dose-response equation50And (4) determining.

In Table 3, the assay data are presented as geometric mean (EC) based on the listed number of repetitions (times) in two (2) significant figures50s) and the arithmetic mean (Emax). Blank space means no data for the embodiment or no Emax is calculated.

Table 3: biological Activity of examples 1 to 102

Test as ammonium and trifluoroacetate salts

Testing as ammonium and 1, 3-dihydroxy-2- (hydroxymethyl) propan-2-aminium (Tris) salts, and as the free acid

Testing as ammonium salt and free acid

Test as formate and free acid.

All patents, patent applications, and references cited herein are hereby incorporated by reference in their entirety.

162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:通过递送抗OSMRβ抗体治疗皮肤疾病或病症

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!