Fluorinated bile acid derivatives

文档序号:620842 发布日期:2021-05-07 浏览:26次 中文

阅读说明:本技术 氟化胆汁酸衍生物 (Fluorinated bile acid derivatives ) 是由 A·C·韦茅斯-威尔逊 G·帕克 B·J·P·林克劳 D·基德-辛克莱 K·A·沃森 于 2019-07-30 设计创作,主要内容包括:通式(I)的化合物:其中R~(2a)、R~(2b)、R~(3a)、R~(3b)、R~5、Y和R~7如本文所定义,该化合物为FXR受体的选择性激动剂,并且用于治疗或预防疾病和病症,包括非酒精性脂肪性肝炎(NASH);原发性胆汁性肝硬化;原发性硬化性胆管炎;胆道闭锁;胆汁郁积性肝病;丙型肝炎感染;酒精性肝病;纤维化;或因纤维化导致的肝损伤。(A compound of the general formula (I): wherein R is 2a 、R 2b 、R 3a 、R 3b 、R 5 Y and R 7 As herein describedAs defined, which are selective agonists of the FXR receptor and are useful for the treatment or prevention of diseases and disorders, including nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis.)

1. A compound of the general formula (I):

wherein

R2a、R2b、R3aAnd R3bEach independently is H or F, with the proviso that R2bAnd R3bAt least one of (a) is F;

R5is CR6aR6bR8、OR8、SR8Or NR6aR8

R6a、R6bAnd R8Each independently is H or methyl;

y is a bond or C1-4Alkylene or C2-4Alkenylene linking groups, one of which is optionally substituted with one or more R10Substitution;

wherein R is10Each independently is halogen or OH;

R7selected from C (O) NR17S(O)2R15、NR17C(O)NR18S(O)2R15、NR17C(S)NR18S(O)2R15And NR17C(NH20)NR18S(O)2R15

R15Is a 5 to 10 membered aryl or heteroaryl ring, optionally substituted with one or more substituents selected from C1-6Alkyl radical, C1-6Haloalkyl, halogen, O (C)1-6Alkyl) and O (C)1-6Haloalkyl);

R17and R18Each independently is H or methyl;

R20is H, methyl or CN;

or a salt or isotopic variation thereof.

2. A compound according to claim 1, wherein R3bIs F, and R3a、R2aAnd R2bAre each H.

3. A compound according to claim 1, wherein R2bIn the case of F, the content of the compound,and R is2a、R3aAnd R3bAre each H.

4. A compound according to claim 1, wherein:

R3bis F, R3aIs H, R2aAnd R2bOne is F, and R2aAnd R2bIs H; or R3aAnd R3bAre both F, and R2aAnd R2bAre all H.

5. A compound according to any one of claims 1-4, wherein R5Is ethyl.

6. A compound according to any one of claims 1 to 5, wherein Y is a bond or C1-3An alkylene linker optionally substituted with one or more OH groups.

7. A compound according to any one of claims 1 to 6, wherein R7Is C (O) NR17S(O)2R15Or NR17C(O)NR18S(O)2R15Wherein R is15、R17And R18As defined in claim 1.

8. The compound according to claim 7, wherein independently or in any combination:

R17and R18Each (if present) of (a) is H; and/or

R15Selected from phenyl and 5-or 6-membered heteroaryl, which may optionally be unsubstituted or substituted by one or more substituents as defined in claim 1.

9. A compound according to claim 8, wherein R15Is phenyl, unsubstituted or substituted by a mono-substituent selected from fluoro, C1-4Alkyl radical, C1-4Fluoroalkyl group, O (C)1-4Alkyl) and O (C)1-4Fluoroalkyl groups).

10. A compound according to claim 1, selected from:

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-toluenesulfonylurea (compound 1);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (compound 2);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (tert-butyl) benzenesulfonylurea (compound 3);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m-toluenesulfonylurea (compound 4);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-toluenesulfonylurea (compound 5);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-fluorobenzenesulfonylurea (compound 6);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m-fluorobenzenesulfonylurea (compound 7);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-fluorobenzenesulfonylurea (compound 8);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p- (trifluoromethyl) benzenesulfonylurea (compound 9);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5B-cholan-23-yl) -m- (trifluoromethyl) benzenesulfonylurea (compound 10);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o- (trifluoromethyl) benzenesulfonylurea (compound 11);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (compound 12);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (compound 13);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -p-trifluoromethoxybenzenesulfonamide (compound 14);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -p-fluorobenzenesulfonamide (compound 15);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -m-fluorophenyl sulfonamide (compound 16);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -o-fluorophenyl sulfonamide (compound 17);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -4-trifluoromethylphenylsulfonamide (compound 18);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -3-trifluoromethylphenylsulfonamide (compound 19);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -2-trifluoromethylphenylsulfonamide (compound 20);

n, N' - (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (Compound 21)

N- (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholan-24-yl) -benzenesulfonamide (compound 22);

and salts and isotopic variations thereof.

11. A compound according to any one of claims 1 to 10 for use in medicine.

12. A compound according to any one of claims 1 to 10 for use in the treatment or prevention of nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis.

13. Use of a compound according to any one of claims 1-10 for the manufacture of a medicament for the treatment or prophylaxis of nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage caused by fibrosis.

14. Treating or preventing nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis, which method comprises administering to a patient in need of such treatment an effective amount of a compound according to any one of claims 1-10.

15. The compound for use according to claim 12, the use according to claim 13 or the method according to claim 14, wherein the fibrosis is selected from the group consisting of liver, kidney and intestinal fibrosis.

16. The compound for use according to claim 15, the use or the method according to claim 15, wherein liver fibrosis is associated with NASH, alcoholic or non-alcoholic fatty liver disease, or with an infection such as hepatitis, especially hepatitis b or c or parasitic liver disease, or is caused by damage induced by congenital diseases, such as Wilson's disease, gaucher's disease, glycogen storage disorders, hemochromatosis, Zellweger's syndrome and congenital liver fibrosis, or by drugs, such as chlorpromazine, tolbutamide, methotrexate, isoniazid and methyldopa; and/or

Renal fibrosis associated with diseases such as diabetic nephropathy, hypertensive nephrosclerosis, glomerulonephritis, interstitial nephritis, transplantation-related glomerulopathy and polycystic kidney disease; and/or

Intestinal fibrosis associated with intestinal diseases.

17. A pharmaceutical composition comprising a compound according to any one of claims 1-10 and a pharmaceutically acceptable excipient or carrier.

18. The pharmaceutical composition according to claim 17, further comprising one or more additional active agents suitable for treating or preventing metabolic syndrome, including non-alcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis.

19. A process for the preparation of a compound according to any one of claims 1 to 10, which process comprises:

A. for the compounds of the general formula (I), wherein R7Is NHC (O) N (R)18)S(O)2R15

Reacting a compound of formula (III):

y, R therein2a、R2b、R3a、R3bAnd R5As defined in claim 1, and R40Is a protected OH group;

with a sulfonamide of formula (IV) in the presence of a catalyst such as 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU):

wherein R is15And R18As defined in claim 1;

to form a compound of formula (II):

y, R therein2a、R2b、R3a、R3bAnd R5As defined in claim 1, and R40Is a protected OH group; and is

Deprotecting a compound of formula (II); or

B. For the compounds of the general formula (I), wherein R7Is NHC (O) N (R)18)S(O)2R15

Reacting a compound of formula (XIII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined in claim 1, and R45And R46Each independently is a protected OH group;

with a sulfonamide of formula (IV) as defined above in the presence of a catalyst such as 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU) to form a compound of formula (XII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined in claim 1, and R45And R46Each independently is a protected OH group; and is

Deprotecting a compound of formula (XII); or

C. For the compounds of the general formula (I), wherein R7Is C (O) N (R)17)S(O)2R15

Reacting a compound of formula (XXIII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined in claim 1, and R46Is a protected OH group;

with sulfonamides of the formula (XXIV)

Wherein R is15And R17As defined in claim 1;

in the presence of a coupling agent such as 1-ethyl-3 (3-dimethylaminopropyl) carbodiimide (EDCI) and a base such as dimethylaminopyridine:

to give a compound of formula (XXII):

y, R therein2a、R2b、R3a、R3b、R5、R15And R17As defined in claim 1, and

R46as defined for the compounds of the general formula (XII); and is

Deprotecting a compound of formula (XXII).

Technical Field

The present invention relates to compounds which are bile acid derivatives and which may be used for the treatment of liver diseases. In particular, the present invention relates to compounds that are selective agonists of the Farnesoid X receptor (Farnesoid X receptor) and are therefore useful in the treatment of diseases such as non-alcoholic steatohepatitis (NASH) and primary cholangitis. The invention also relates to pharmaceutical compositions comprising the compounds of the invention.

Background

Nonalcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver disease worldwide and is rapidly becoming a major indicator of liver transplantation (Bellentani, 2017). NAFLD describes a range of physiological conditions, ranging from simple lipid accumulation in the liver (sebaceous gland disease) to non-alcoholic steatohepatitis (NASH), characterized by lobular inflammation and hepatocellular injury (Haas, frame and stals, 2016). In the west, NAFLD is considered a huge public health burden, estimated to affect 30% of the population in the uk (Dyson, anste and McPherson, 2014). Increased prevalence of NAFLD reflects increased prevalence of obesity and type 2diabetes, and NAFLD is considered a hepatic manifestation of metabolic syndrome (Cave et al, 2016).

In patients with NAFLD, at least 10-20% continue to develop NASH, which is accompanied by a patient's predisposition to hepatic and extrahepatic complications such as fibrosis, cirrhosis, hepatocellular carcinoma (HCC) and cardiovascular disease. Although little is known, the pathological progression from steatosis to NASH is thought to consist of multiple "hits" with lipotoxicity, oxidative stress and endoplasmic reticulum stress, sensitizing the liver to other damaging systems mediated by the innate immune defense system and causing cytokine-induced cell damage (Pacana and Sanyal, 2015). One "hit" involves liver neoadipogenesis, which is activated by hyperinsulinemia and carbohydrate-rich diet. Furthermore, as a result of insulin resistance, lipolysis in dysfunctional adipocytes is not inactivated, leading to the penetration of Free Fatty Acids (FFA) into the circulatory system. These FFAs accumulate in ectopic tissues (e.g. liver) and are stored as triglycerides. However, due to triglyceride-derived toxic metabolites, the excessive accumulation of FFA exceeds the triglyceride storage threshold, eventually leading to lipotoxicity. In addition, a decrease in triglyceride clearance and a decrease in Very Low Density Lipoprotein (VLDL) output also contribute to the accumulation of fat in the liver. These events are offset by mitochondria and peroxisomes, which attempt to oxidize fatty acids, but ultimately result in damage to these organelles, resulting in overproduction of Reactive Oxygen Species (ROS) and destruction of associated molecular pattern molecules (DAMPs). In the second "hit", lipid peroxidation and FFA and ROS activate inflammatory cytokines to trigger inflammation and apoptosis and can activate the natural immune defense system through Toll-like receptors, further prolonging NASH progression by exacerbating insulin resistance and initiating fibrogenesis (cui, 2012). Furthermore, recent evidence suggests that the liver-gut axis is involved in the development of disease. In NAFLD patients, the composition of the intestinal flora is altered and intestinal permeability is increased, and inflammatory body (inflamesome) -mediated dysbiosis is also thought to contribute to the development of NASH (Henao-mejia et al, 2012; Mouzaki et al, 2013). The complex, multifactorial, trans-systemic nature of this metabolic abnormality is highlighted by the lipotoxic hepatic events in combination with the indirect effects of inflammatory mediators of the adipose tissue, gut and immune system, and importantly reflects a number of potential therapeutic targets for NASH (Haas, Francque and stals, 2016).

Currently, there is no effective medical therapy for the treatment or prevention of NASH. Most commonly, weight loss and improved insulin sensitivity through changes in diet and lifestyle are recommended, but since many patients cannot initiate or sustain these changes, long-term pharmacological solutions are needed (Neuschwander-Tetri et al, 2015). Research has focused on potential molecular targets for the treatment of NASH, including several nuclear hormone receptors of the NR1 subfamily. These receptors, in particular the Farnesoid X Receptor (FXR), are attractive targets due to their potential role in several pathways contributing to the etiology of the disease.

Like all nuclear receptors, Farnesoid X Receptors (FXRs) act as ligand-activated transcription factors that can modulate the cellular mechanisms responsible for controlling epigenetic changes. There are two genes encoding FXR: FXR α (NR1H4), which is highly conserved in many species, and FXR β (NR1H5), which exists as a pseudogene in humans. The FXR α gene encodes four different isoforms that arise from different promoter applications and alternative mRNA splicing combinations. Although all four isoforms have highly conserved ligand binding domains, suggesting that FXR ligand will bind to any isoform in a non-selective manner, there is clearly differential expression where 2 isoforms are predominantly expressed in hepatocytes and cells with active steroid metabolism, and others are predominantly expressed in the colon, intestine and other cells involved in the enterohepatic cycle (Huber et al, 2002; Vaquero et al, 2013). Furthermore, certain FXR target genes are more responsive to certain isoforms than others, and the overall pattern of isoform expression is believed to have a significant impact on the sensitivity of specific tissues to FXR ligands and transcriptional response (Zhang, Kast-woelbern and Edwards, 2003; Vaquero et al, 2013).

FXR stands for its structural organization and activationTypical nuclear receptors. Briefly, FXR consists of an N-terminal DNA Binding Domain (DBD) composed of two Zn's responsible for recognition and binding of a common hormonal response element2+Finger components, which are connected by a variable hinge region to the C-terminal Ligand Binding Domain (LBD), which is the hydrophobic pocket necessary for the identification and precipitation of small molecule ligands (Chiang, 2013). Similar to other nuclear receptors, FXR binds to DNA to a heterodimeric Retinoid X Receptor (RXR) with an obligatory partner. FXR is inactive in the absence of ligand. Typically, FXR/RXR heterodimers are pre-associated with AGGTCA reverse heavy response elements of their target genes in the form of complexes with co-repressor peptides (Neuschwander-Tetri, 2012). Upon activation by ligand binding, the receptor undergoes a conformational change, releasing the co-repressor complex, exposing the binding site of the LXXLL co-activation motif to the hydrophobic groove of the ligand binding pocket (Copple and Li, 2016). Hydrogen bonding between the LBD surface and the two ends of coactivators will form "charge clamps" and coactivators are recruited to the site. Eventually, this results in a change in the chromatin structure of the target gene, bringing the universal transcription factor and RNA polymerase close to its promoter and thus initiating its transcription (anathanarayanan et al, 2004).

A typical endogenous ligand for FXR is bile acid. Bile acids are steroid acids found in mammalian bile and include compounds such as cholic acid, chenodeoxycholic acid, lithocholic acid, and deoxycholic acid, all found in humans.

The general numbering system for steroids and the numbering of the carbon atoms in chenodeoxycholic acid are shown below.

FXR acts as a major regulator of bile acid metabolism. The main role of FXR is to promote nutrient and energy transfer along the entero-hepatic-fatty axis in fed and fasted states (Evans and magelsdorf, 2014). After postprandial stimulation, bile acids cause lipid absorption and activate FXR-mediated signal transduction pathways. This promotes nutrient absorption by the gut and stimulates energy metabolism in the liver by the action of FXR target gene fibroblast growth factor 19(FGF 19). In addition to intestinal FGF19, expression of FXR transcription targets (short heterodimer partners (SHPs)) in the liver also leads to down-regulation of de novo bile acid synthesis and tight control of enterohepatic bile acid pool according to metabolic needs. Studies in NASH patients indicate that decreased expression of FXR and bile acid biosynthetic enzymes, cholesterol 7 alpha hydroxylase (CYP7a1), and sterol 27 hydroxylase (CYP27a1) are all directly proportional to the severity of the disease; NAFLD patients also showed impaired response to hepatic FGF19 (Yang, Shen and Sun, 2010; Min et al, 2013; Cave et al, 2016).

Recently, FXR has become a major participant in lipid, glucose and cholesterol homeostasis, regulating genes involved in liver lipogenesis, VLDL synthesis, insulin sensitivity, and partly due to interactions with other nuclear receptors, gluconeogenesis and glycogenesis (Kast et al, 2001; Watanabe et al, 2004; Ma et al, 2006; Zhang et al, 2006). As a result of acting through the SHP signaling cascade, FXR down-regulates the sterol regulatory element binding protein (SREBP1) to reduce fatty acid synthesis, while up-regulating peroxisome proliferator-activated receptor alpha (PPAR α) to increase fatty acid catabolism by mitochondrial β -oxidation, thereby reducing liver fatty acid accumulation. FXR also increases expression levels of apolipoprotein C2(APOC2) and VLDL receptors, which are responsible for VLDL hydrolysis and clearance. Studies using FXR deficient mice show phenotypic similarities to human NASH patients, including significantly elevated hepatic triglyceride levels, elevated circulating FFA, and hepatic steatosis (Maloney et al, 2000; Zhang et al, 2004). Furthermore, FXR activation by natural and synthetic agonists has been able to improve plasma triglyceride levels in rodents (Kast et al, 2001). By reducing the levels of triglycerides and FFA, it is believed that FXR activation may also increase insulin sensitivity in the liver and surrounding tissues, as observed in FXR null mice, which exhibit mild glucose intolerance and insulin signaling oscillations in both liver and muscle (Ma et al, 2006). Consistent with this finding, semi-synthetic FXR agonist obeticholic acid (OCA) may improve insulin sensitivity in human subjects and animal obesity models of NASH (Cipriani et al, 2010; Mudaliar et al, 2013). Also, FXR has been proposed to be primarily mediated in glucose homeostasis by FGF19 signaling; lowering plasma glucose concentrations, lowering the expression and activity of three key gluconeogenic enzymes, phosphoenolpyruvate carboxykinase (PEPCK), fructose-1, 6-bisphosphatase (FBP1) and glucose-6-phosphatase (G6Pase), and inhibiting cAMP regulatory element binding protein (CREB) and downstream PPAR γ coactivator 1- α (PGC1 α) to promote storage of glucose as glycogen play an important role (Zhang et al, 2006). However, contradictory results from animal models suggest that FXR involvement may be only part of the complex network of receptors and pathways (Watanabe et al, 2011).

Notably, FXR is involved in the inhibition of hepatitis, where complex pathways are involved that lead to the negative regulation of specific nuclear factor κ B (NF- κ B) target genes and proinflammatory cytokines (Wang et al, 2008). Furthermore, although the underlying mechanisms are still poorly understood, FXR is thought to play an important role in gut protection and maintenance of the gut barrier of the gut flora (Inagaki et al, 2006). FXR knockout mice fed a high-fat diet exhibit excessive intestinal bacterial growth and elevated levels of proinflammatory and profibrogenic mediators, such as tumor necrosis factor alpha (TNF α), Tissue Inhibitors of Metalloproteinases (TIMP), and transforming growth factor beta (TGF β -1) (Kong et al, 2009). Furthermore, preclinical evidence suggests that the involvement of FXR agonists inhibits NF κ B expression in primary hepatocytes and in mouse models of NASH, thereby improving the inflammatory microenvironment and fibrosis (Kong et al, 2009; Ma et al, 2013).

To further support its role in NASH (and HCC in particular), FXR has been shown to modulate the expression of tumor suppressor genes, and FXR agonists were shown to reduce tumor growth and metastasis in mouse liver xenograft tumor models (Deuschle et al, 2012; Jiang et al, 2013). Thus, this evidence, in addition to playing a central role in the gut-liver-fat axis, supports the concept that FXR dysregulation leads to the development of NASH in terms of maintaining intestinal barrier integrity, inhibiting inflammation, and regulating bile acid, glucose, and lipid metabolism, and thus confirms that FXR is an ideal target for a therapeutic agent for NASH.

Many FXR agonists are known, including various non-steroidal compounds. More recently, bile acid analogs having FXR agonist activity have been developed. They include obeticholic acid (OCA; INT-747) described in WO 02/072598 and EP 1568706. Analogs and medical uses of OCAs and methods of making OCAs and analogs are described in the following references: WO 2005/092925, WO 2005/089316, WO 2006/122977, WO 2007095174, WO 2008/002573, WO 2008/091540, WO 2010/014836, WO 2010/059853, WO 2010/059859, WO 2013/192097, WO 2014/066819, WO 2015/085474, WO 2014/184271, WO 2016/127019, WO 2016/144946, WO 2016/164413, WO 2016/176208, WO 2016/205475, WO 2017/019524, WO 2017/027396, WO 2017/053428, WO 2017/053826, WO 2017/062763, WO 2017/079062, WO 2017/111979 and WO 2017/156024 (all inclusive Pharmaceuticals).

Additional 6-alkylcholytic acid analogs with modified side chains are described in WO 2016/073767, WO 2016/086115, WO 2016/086134, WO 2016/086169, WO 2016/086218, WO 2016/130809, WO 2016/161003, WO2017/147137, WO 2017/147159 and WO 2017/147174 (all from Enanta Pharmaceuticals, Inc).

Other references relating to similar compounds include CN105646634, WO 2016/173524, WO 2016/173397, CN105348365, US 2014/0206657.

Our earlier applications WO 2016/079518, WO 2016/079518, WO 2016/079519, WO 2016/079520, WO 2017/199036, WO 2017/199039 and WO 2017/199033 all relate to methods of preparing these bile acid analogues and intermediates in their synthesis.

One of the problems with the bile acid analogues described in the prior art is that, in addition to their activity as FXR agonists, they are also modulators of the G protein-coupled receptor TGR 5. It is a member of the rhodopsin-like superfamily of G protein-coupled receptors and plays an important role in the bile acid signaling network. For example, one of the adverse effects associated with the use of OCA is pruritus and is believed to result from OCA activation of off-target receptors such as TGR5 (Alemi et al, 2013).

Xiao et al, 2017, relates to the synthesis and biological evaluation of OCA and its series of OCA derivatives as FXR agonists. In the derivatives, the carboxylic acid groups are replaced by various substitutes. The authors indicate that all tested compounds showed low to moderate TGR5 potency and the best selectivity obtained was 30 times the selectivity towards FXR, which was achieved by the tetrazole derivative. With the highest liver: compound 18, having CH, at a plasma concentration ratio2CH2C(O)NH-S(O)2CH3Side chains.

The present invention relates to novel compounds which maintain FXR agonist activity and have enhanced selectivity for FXR over TGR 5.

Furthermore, bile acid derivatives fluorinated at the 2 and/or 4 position and having arylsulfonamide or sulfonylurea side chains have increased agonist activity towards the FXR receptor compared to known bile acid derivatives. The inventors postulate that this increased agonist activity is due to the binding of the molecule into the canonical and allosteric pocket of the FXR ligand binding domain. However, the effectiveness of the compounds of the present invention is not affected by the correctness of this presumption or otherwise.

Summary of The Invention

Accordingly, the present invention provides a compound of general formula (I):

wherein

R2a、R2b、R3aAnd R3bAre each independently H or F, with the proviso that R2bAnd R3bAt least one of (a) is F;

R5is CR6aR6bR8、OR8、SR8Or NR6aR8

R6a、R6bAnd R8Each independently is H or methyl

Y is a bond or C1-4Alkylene or C2-4Alkenylene linking group, among themOptionally substituted by one or more R10Substitution;

wherein R is10Each independently is halogen or OH;

R7selected from C (O) NR17S(O)2R15、NR17C(O)NR18S(O)2R15、NR17C(S)NR18S(O)2R15And NR17C(NR20)NR18S(O)2R15

R15Is a 5 to 10 membered aryl or heteroaryl ring, optionally substituted with one or more substituents selected from C1-6Alkyl radical, Ci-6Haloalkyl, halogen, O (C)1-6Alkyl) and O (C)1-6Haloalkyl);

R17and R18Each independently is H or methyl;

R20is H, methyl or CN;

or a salt or isotopic variation thereof.

Some fluorinated bile acid derivatives are known, for example, Roda et al 1995, Honorio et al 2006, US 5,175,320 and WO 97/44043 all relate to 6-analogues of bile acid analogues, while WO 2014/160441 describes 6, 6-biliac acid analogues. Sato et al 2008 relates to 7-fluorolithocholic acid derivatives, and sieven et al 2008 and Cushman et al 1995 disclose 3, 3-difluorocholane-24-acid and its methyl ester, respectively. EP 3290429 discloses bile acid derivatives, which are believed to be cited for the treatment of FXR-mediated diseases. The disclosed compounds include some 4-fluoro bile acid derivatives. Both Clerici et al 2006 and Macchiarulo et al 2008 relate to 3 α -6 α -dihydroxy-7 α -fluoro-5 β -cholanic acid ester, which is believed to be cited for treatment of liver disease. WO2016/154216 relates to 3-and 7-fluorinated derivatives of UDCA, which are cited for the treatment of neurodegenerative diseases. WO2016/173493 relates to bile acid derivatives with modified side chains and which are modulators of FXR and/or TGR 5. US 2018/0148470 relates to 4 β -fluorinated bile acid derivatives which are believed to be useful in the treatment of FXR-mediated diseases. However, no compounds of the general formula (I) are taught in the prior art.

The compounds of formula (I) are selective FXR agonists and are therefore useful in the treatment of diseases and disorders such as nonalcoholic steatohepatitis (NASH); primary Biliary Cirrhosis (PBC); primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; and liver damage due to fibrosis.

While not wishing to be bound by this theory, the present inventors have speculated that the presence of fluorine in the ring alters the hydrogen bonding capability of the 3 α hydroxyl group, thereby affecting the receptor activity and selectivity of the compound towards FXR and TGR 5. Furthermore, it is clear that compounds having at position 2 or 4 of the steroid ring system may have increased metabolic stability.

The present inventors have also found that, as shown in the following examples, a side chain group (-Y-R)7) Has a significant effect on FXR agonist activity.

In this specification, unless the context requires otherwise due to express language or necessary implication, the word "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention.

In the present specification, reference to "pharmaceutical use" is to be taken to a human or animal, in particular a human or a mammal, such as a domestic or domestic animal mammal, for the purpose of treating or preventing a disease or medical condition. The term "pharmaceutical composition" refers to a composition suitable for pharmaceutical use, and "pharmaceutically acceptable" refers to an active agent suitable for use in a pharmaceutical composition. Other similar terms should be construed accordingly.

In the context of this specification, the term "plurality" means two or more.

All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.

Drawings

In the figure, p is < 0.05, p is < 0.01, and p is < 0.001.

FIG. 1 shows the human hepatoma cell line Huh7 in comparison with the control, OCA (in EC)50And EC90) And Compound 2 (in EC)50And EC90) Change in SHP expression after 24 hours incubation together.

FIG. 2 shows the human hepatoma cell line Huh7 in comparison with the control, OCA (in EC)50And EC90) And Compound 2 (in EC)50And EC90) Change in the expression of OST α after 24 hours incubation together.

FIG. 3 shows the human hepatoma cell line HepG2 together with the control, OCA (in EC)50And EC90) And Compound 2 (in EC)50And EC90) Change in CYP7a1 expression after 24 hours incubation together.

FIG. 4 shows the human hepatocellular carcinoma cell line HepG2 and control, OCA (in EC)50And EC90) And Compound 2 (in EC)50And EC90) Change in TGF-beta 1 expression after 24 hours incubation together.

FIG. 5 shows human hepatocytesTumor cell line Huh7 and control, OCA (in EC)50And EC90) And compound 14 (in EC)50And EC90) Change in SHP expression after 24 hours incubation together.

FIG. 6 shows the human hepatoma cell line Huh7 in comparison with control, OCA (in EC)50And EC90) And compound 14 (in EC)50And EC90) Change in the expression of OST α after 24 hours incubation together.

FIG. 7 shows the human hepatocellular carcinoma cell line HepG2 and control, OCA (in EC)50And EC90) And compound 14 (in EC)50And EC90) Change in CYP7a1 expression after 24 hours incubation together.

FIG. 8 shows the human hepatocellular carcinoma cell line HepG2 and control, OCA (in EC)50And EC90) And compound 14 (in EC)50And EC90) Change in TGF-beta 1 expression after 24 hours incubation together.

Examples

In the examples, the following abbreviations are used.

Ac2O acetic anhydride

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

DMAP dimethylaminopyridine

EDCI 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide

Equiv equivalent

Et3N-Triethylamine

EtOAc ethyl acetate

IPA isopropyl alcohol

h hours

HDCA hyodeoxycholic acid

HPLC high performance liquid chromatography

LDA lithium diisopropylamide

MeOH methanol

n-BuLi n-butyl lithium

OCA obeticholic acid

PE Petroleum Ether

PTFE Polytetraene

pTSA p-toluenesulfonic acid

RT Room temperature

sat saturation

TBAF tetrabutylammonium fluoride

TBDMS-OTf triflic acid tert-butyldimethylsilyl ester

TEMPO (2, 2, 6, 6-tetramethylpiperidin-1-yl) oxidinyl

THF tetrahydrofuran

TMS-Cl trimethylsilyl chloride

TMS-OTf Trimethylsilyl triflate

TLC thin layer chromatography

Example 1-3 α -hydroxy-4 β -fluoro-6 α -ethyl-7 α -hydroxy-5 β -cholane with sulfonylurea-substituted side chain Synthesis of acid analogs

A.6 alpha-ethyl-3 alpha, 7 alpha-dihydroxy-5 beta-cholane-24-oic acid methyl ester

To a solution of OCA (23.5g, 55.87mmol) in MeOH (540mL) at RT was added p-toluenesulfonic acid (1.02mg, 5.59mmol,. about.0.1 equiv.) and sonicated at 30 ℃ for 3 h. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in chloroform (500mL) and saturated NaHCO was used3(500mL)、H2O (500mL) and brine (500mL), MgSO4Drying, filtration and concentration in vacuo gave the title compound as a white solid in quantitative yield. The resulting solid was used without further purification.

1H NMR(400MHz,CDCl3):δ3.70(1H,s),3.67(3H,s,),3.44-3.37(1H,m),2.40-2.32(1H,m),2.26-2.18(1H,m),1.96(1H,dt,J=12.0,2.6Hz),1.92-1.76(6H,m),1.69-1.59(3H,m),1.58-1.12(14H,m),1.00(1H,td,14.2,3.3Hz),0.93(3H,d,J=6.3Hz),0.90(3H,s),0.90(3H,t,J=7.4Hz),0.66(3H,s)ppm。

LRMS(ESI+)m/z:452.4[M+NH4]+,100%。

B.6 alpha-Ethyl-7 alpha-hydroxy-3-oxo-5 beta-cholan-24-oic acid methyl ester

Stirred methyl 6 α -ethyl-3 α, 7 α -dihydroxy-5 β -cholan-24-oate (9.53g, 21.9mmol) from step A at RT in H2To a solution of O (22mL) and t-butanol (88mL) was added KBr (5.22g, 43.9mmol,. about.2.0 equiv.), KHCO3(22.0g, 219mmol,. about.10 equivalents) and TEMPO (4.45g, 28.5mmol,. about.1.3 equivalents). The reaction mixture was cooled to 0 deg.C and received dropwise addition of NaClO (28mL, 32.9mmol,. about.1.5 equiv.) at a rate of 4 mL/hr over a period of 7 hours. On completion, by slow addition of 1: 1 saturated Na2S2O3The reaction was quenched (250mL) and diluted with EtOAc (200 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X150 mL). The organic phases were combined and MgSO4Drying, filtration and concentration in vacuo gave 14.2g of crude material as an orange oil. The resulting oil was purified by column chromatography (acetone 40-60, 0-20% gradient in PE) to give the title compound as a white solid (8.48g, 89%).

1H NMR(400MHz,CDCl3):δ3.78(1H,d,J=2.2Hz),3.67(3H,s),3.07(1H,dd,J=15.2,13.5Hz),2.46-2.33(2H,m),2.29-1.91(7H,m),1.84-1.77(1H,m)。1.74-1.15(18H,m),1.00(3H,s),0.94(3H,d,J=6.5Hz),0.91(3H,t,J=7.4Hz),0.70(3H,s)ppm。

LRMS(ESI+)m/z:450.3[M+NH4]+,100%。

C.6 alpha-Ethyl-4 beta-fluoro-7 alpha-hydroxy-3-oxo-5 beta-cholan-24-oic acid methyl ester

To a stirred, pre-cooled solution of diisopropylamine (0.78mL, 5.54mmol,. about.12 equivalents) in dry THF (6.9mL) was added dropwise a solution of n-BuLi in hexane (1.44mL, 2.31mmol,. about.5.0 equivalents) at-78 deg.C under an argon atmosphere over 0.25 h. After addition, trimethylsilyl chloride (0.29mL, 2.31mmol,. about.5.0 equiv.) was added and stirred for 1 h. A solution of methyl 6 α -ethyl-7 α -hydroxy-3-oxo-5 β -cholan-24-oate (200mg, 0.46mmol) from step B in dry THF (3mL) and triethylamine (1.16mL, 8.32mmol, 18 equivalents) was then added. After the addition was complete, the reaction was gradually warmed to-20 ℃ and stirred for 2 h. Upon completion, saturated NaHCO was added dropwise3The reaction was quenched (5mL) and warmed to RT for 2 h. The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X 10 mL). The combined organic phases were washed with brine (30mL) and MgSO4Drying, filtration and concentration in vacuo gave 271mg of crude material as a yellow residue.

To a stirred solution of the crude material obtained in MeCN (13mL) was addedThe mixture was stirred for 16 h. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in EtOAC (20mL) and acidified with 2M HCl (30 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3 × 15 mL). The combined organic phases were washed with brine (100mL) and MgSO4Drying, filtration and concentration in vacuo gave 196mg of crude material as a green solid. Purification by HPLC using hexane/acetone (90/10) as eluent gave the title compound and a mixture of methyl-2 β -3-oxo-6 α -ethyl-7 α -hydroxy-5 β -cholan-24-oate which could not be separated as a colorless oil (79mg, 0.18mmol, 37% title compound, accounting for 1% methyl-2 β -fluoro-3-oxo-6 α -ethyl-7 α -hydroxy-5 β -cholan-24-oate contamination by1H NMR)。

1H NMR(400MHz,CDCl3):δ5.94(1H,dd,J=46.5,10.9Hz),3.88(1H,s),3.65(3H,s),2.49(1H,td,J=14.6,5.0Hz),2.38-2.09(4H,m),2.01-1.30(18H,m),1.25-1.14(3H,m),1.04(3H,s),0.93-0.89(6H,m),0.68(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):-194.3(1F,dd,J=46.8,13.9Hz)ppm。

LRMS(ESI+)m/z:468.4[M+NH4]+,100%。

D.6 alpha-ethyl-4 beta-fluoro- (3 alpha, 7 alpha) -dihydroxy-5 beta-cholan-24-oic acid methyl ester

To a stirred solution of methyl 6 α -ethyl-4 β -fluoro-7 α -hydroxy-3-oxo-5 β -cholan-24-oate (75mg, 0.17mmol) from step C in dry THF (6.7mL) at RT under argon was added NaBH4(19mg, 0.50mmol,. about.3.0 equiv.) and stirred for 16 h. On completion, by dropwise addition of H2The reaction was quenched with O (8mL) and diluted with EtOAc (10 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3 × 50 mL). The organic phases are combined with H2O (100mL) wash, MgSO4Drying, filtration and concentration in vacuo gave 77mg of crude material as a white residue. Purification by HPLC using hexane/acetone (90/10) as eluent gave the title compound as a colorless oil (55mg, 0.12mmol, 74%).

1H NMR(400MHz,CDCl3):δ5.31(1H,ddd,J=50.0,10.4,8.9Hz),3.82(1H,s),3.67(3H,s),3.57-3.50(1H,m),2.36(1H,ddd,J=15.4,10.1,5.7Hz),2.27-2.18(1H,m),1.96-1.92(2H,m),1.83-1.07(23H,m),0.97(3H,s),0.94-0.92(6H,m),0.66(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-189.0(1F,d,J=50.3Hz)ppm。

LRMS(ESI+)m/z:470.4[M+NH4]+,100%。

E.3 alpha, 7 alpha-dihydroxy-4 beta-fluoro-6 alpha-ethyl-5 beta-cholanic acid

To a stirred solution of methyl 6 α -ethyl-4 β -fluoro- (3 α, 7 α) -dihydroxy-5 β -cholan-24-oate (58mg, 0.13mmol) in MeOH (5mL) was added NaOH (250mg, 5% solution) at RT and stirred for 18 h. Upon completion, the reaction mixture was concentrated in vacuo, and the residue was acidified to pH 2 with 1M HCl and diluted with EtOAc (20 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3 × 50 mL). The combined organic phases were washed with brine (100mL) and MgSO4Drying, filtration and concentration in vacuo gave 76mg of crude material as a yellow oil. Activation by HPLC using hexane/acetone (70/30) as eluent gave the title compound as a colorless oil (41mg, 0.09mmol, 72%).

1H NMR(400MHz,CDCl3):δ5.31(1H,dt,J=49.9,9.5Hz),3.83(1H,s),3.60-3.50(1H,m),2.36(1H,ddd,J=15.5,10.4,5.3Hz),2.26(1H,ddd,J=15.8,9.5,6.6Hz),1.97-1.91(2H,m),1.85-1.08(21H,m),0.97(3H,s),0.95-0.91(6H,m),0.67(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.7(1F,d,J=48.6Hz)ppm。

LRMS(ESI+)m/z:456.2,[M+NH4]+,100%。

Synthesis of Compounds with sulfonylurea-substituted side chains

The following method exemplifies 4 β -fluoro derivatives, but may also be used for 2 β -fluorinated, 4-difluoroor 2, 4-difluoro compounds.

F.3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-5 beta-cholanic acid

Stirring of 3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -chola from step E under argon atmosphere at RTA solution of alkanoic acid (2.08g, 4.74mmol) in dry THF (160mL) was added dropwise with NaHCO3(2.04g, 23.7mmol,. about.5.0 equivalents) and Ac2O (2.29mL, 23.7mmol,. about.5.0 equiv.) over 5 min. After addition, the reaction mixture was heated at 70 ℃ for 16 h. On completion, the reaction mixture was cooled to RT and H was added dropwise2O (100mL) was quenched, acidified with 1M HCl (20mL), and diluted with EtOAc (100 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3 × 150 mL). The combined organic phases were washed with brine (400mL) and MgSO4Dried, filtered and concentrated in vacuo to give a yellow oil. The resulting oil (MeOH CH) was purified by column chromatography2Cl2Gradient elution of the solution 0-3%) gave the title compound as a white solid (660mg, 29%).

1H NMR(400MHz,CDCl3):δ5.47(1H,dt,J=49.4,9.4Hz),4.78(1H,dddd,J=14.1,11.9,9.3,5.0Hz),3.84(1H,s),2.41(1H,ddd,J=15.5,10.2,5.3Hz),2.27(1H,ddd,J=15.8,9.7,6.6Hz),2.06(3H,s),1.97-1.89(2H,m),1.86-1.80(3H,m),1.70-1.14(19H,m),0.99(3H,s),0.94(3H,d,J=6.2Hz),0.92(3H,t,J=7.1Hz),0.67(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.6(1F,dt,J=50.3,12.1Hz)ppm。

LRMS(ESI+)m/z:498.2,[M+NH4]+,100%。

G.3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-5 beta-cholane-24-acyl azide

To a solution of 3 α -acetoxy-4 β -fluoro-6 α -ethyl-7 α -hydroxy-5 β -cholanic acid (200mg, 0.42mmol) from step F in dry THF (4mL) was added Et3N (0.12mL, 0.83mmol,. about.2.0 equiv) dropwise at RT under an argon atmosphere. After addition, the reaction mixture was cooled to 0 ℃ and diphenylphosphoryl azide was added dropwiseSubstance (0.13mL, 0.62mmol,. about.1.5 equiv). After addition, the reaction mixture was stirred for 3h after a strong gas flow hood. Upon completion, the reaction was quenched with brine (5mL) and CH2Cl2(5mL) dilution. Removing the organic phase with CH2Cl2Back extract the aqueous phase (3 × 5 mL). The organic phases were combined and MgSO4Drying, filtration and concentration in vacuo at 0 ℃ gave a yellow oil. The resulting oil was used without further purification.

1H NMR-characteristic peaks (400MHz, CDCl)3):δ5.47(1H,ddd,J=49.4,10.4,9.2Hz),4.82-4.74(1H,m),3.83(1H,s),2.38(1H,ddd,J=15.8,10.0,5.3Hz),2.29-2.23(1H,m),2.06(3H,s),0.98(3H,s),0.920(3H,d,J=6.5Hz),0.919(3H,t,J=7.2Hz),0.67(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.8(1F,dt,J=50.3Hz)ppm。

H. 3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl isocyanate

A stirred solution of the crude oily 3 α -acetoxy-4 β -fluoro-6 α -ethyl-7 α -hydroxy-5 β -cholane-24-acyl azide from step G (105mg putative, 0.42mmol) in dry toluene (3.1mL) was heated to 125 ℃ under an argon atmosphere. After 5h, the reaction was cooled to RT. The resulting solution was used without further purification.

1H NMR-characteristic peaks (400MHz, CDCl)3):δ5.47(1H,ddd,J=49.4,10.4,9.2Hz),4.81-4.73(1H,m),3.83(1H,s),3.35(1H,ddd,J=12.9,7.8,4.5Hz),3.30-3.24(1H,m),2.05(3H,s),0.98(3H,s),0.94(3H,d,J=6.6Hz),0.92(3H,t,J=7.2Hz),0.68(3H,s)ppm。

19F NMR(1H non-uncoupled, 376MHz, CDCl 3): delta-186.5 (1F, dt, J ═ 49.3, 12.7)Hz)ppm。

General procedure 1 for the conversion of isocyanates into sulfonylureas

To a stirred crude solution of 3 α -acetoxy-4 β -fluoro-6 α -ethyl-7 α -hydroxy-24-nor-5 β -cholan-23-yl isocyanate from step H in toluene was added sulfonamide (-1.5 equivalents) and DBU (-1.5 equivalents) and stirred for 16H. Upon completion, the reaction was quenched by dropwise addition of 1M HCl (2mL) and diluted with EtOAc (5 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X5 mL). The organic phases were combined and MgSO4Drying, filtering and vacuum concentrating. The resulting product was purified by column chromatography (acetone gradient elution in PE, 40-60, 5-20%) to give the desired sulfonylurea.

N, N' - (3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -p-toluenesulfonylurea (intermediate 1)

Prepared according to general procedure 1 using 53.4mg p-toluenesulfonamide to give intermediate 1 as a yellow oil (51.7mg, 38%).

1H NMR(400MHz,CDCl3):δ7.78(2H,d,J=8.2Hz),7.32(2H,d,J=8.3Hz),6.50(1H,t,J=4.8Hz),5.47(1H,dt,J=49.4,9.8Hz),4.83-4.73(1H,m),3.83(1H,s),3.33-3.25(1H,m),3.19-3.11(1H,m),2.44(3H,s),2.06(3H,s),1.96-1.80(5H,m),1.72-1.38(15H,m),1.23-1.14(5H,m),0.99(3H,s),0.93(3H,d,J=6.6Hz),0.92(3H,t,J=7.5Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.6(1F,dt,J=49.0,12.8Hz)ppm。

LRMS(ESI+)m/z:666.4,[M+NH4]+,100%。

N, N' - (3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -benzenesulfonylurea (intermediate 2)

Prepared according to general procedure 1 using 49.0mg benzenesulfonamide to give intermediate 2 as a yellow oil (49.9mg, 38%).

1H NMR(400MHz,CDCl3):δ7.90(2H,d,J=7.6Hz),7.63(1H,t,J=7.0Hz),7.50(2H,t,J=7.7Hz),6.60(1H,s),5.48(1H,dt,J=49.2,9.8Hz),4.84-4.74(1H,m),3.83(1H,s),3.31-3.25(1H,m),3.18-3.10(1H,m),2.06(3H,s),1.95-1.80(5H,m),1.69-1.38(13H,m),1.29-1.12(7H,m),0.99(3H,s),0.93(3H,d,J=6.7Hz),0.92(3H,t,J=7.0Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.5(1F,dt,J=48.6,12.1Hz)ppm。

LRMS(ESI+)m/z:652.3,[M+NH4]+,100%。

N, N' - (3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -4- (tert-butyl) benzenesulfonylurea (intermediate 3)

Prepared according to general procedure 1 using 53.2mg 4- (tert-butyl) benzenesulfonamide to give intermediate 3 as a colorless oil (81.6mg, 71%).

1H NMR(400MHz,CDCl3):δ7.81(2H,d,J=8.7Hz),7.54(2H,d,J=8.7Hz),6.56(1H,s),5.48(1H,ddd,J=49.4,10.4,9.3Hz),4.83-4.73(1H),3.84(1H,s),3.33-3.29(1H,m),3.21-3.14(1H,m),2.07(3H,s),1.97-1.80(4H,m),1.74-1.44(13H,m),1.36(9H,s),1.29-1.16(7H,m),0.99(3H,s),0.95(3H,d,J=6.6Hz),0.93(3H,t,J=7.0Hz),0.67(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.6(1F,dt,J=49.9,13.2Hz)ppm。

LRMS(ESI+)m/z:708.4,[M+NH4]+,100%。

N, N' - (3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -m-toluenesulfonylurea (intermediate 4)

Prepared according to general procedure 1 using 42.6mg of m-toluenesulfonamide to give intermediate 4 as a colorless oil (85.7mg, 80%).

1H NMR(400MHz,CDCl3):δ7.71-7.68(2H,m),7.45-7.38(2H,m),6.54(1H,s),5.47(1H,ddd,J=49.4,10.3,9.4Hz),4.83-4.73(1H,m),3.83(1H,s),3.33-3.26(1H,m),3.19-3.12(1H,m),2.42(3H,s),2.06(3H,s),1.96-1.80(4H,m),1.72-1.11(21H,m),0.99(3H,s),0.94(3H,d,J=6.6Hz),0.93(3H,t,J=7.3Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.6(1F,dt,J=49.4,13.4Hz)ppm。

LRMS(ESI+)m/z:666.3,[M+NH4]+,100%。

N, N' - (3 alpha-acetoxy-4 beta-fluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -o-toluenesulfonylurea (intermediate 5)

Prepared according to general procedure 1 using 42.6mg o-toluenesulfonamide to give intermediate 5 as a colorless oil (55.4mg, 51%).

1H NMR(400MHz,CDCl3):δ7.94(1H,dd,J=8.3,1.0Hz),7.51(1H,td,J=7.6,1.2Hz),7.34(2H,d,J=7.3Hz),6.47(1H,s),5.47(1H,dt,J=49.3,9.5Hz),4.83-4.73(1H,m),3.83(1H,s),3.28-3.22(1H,m),3.15-3.08(1H,m),2.65(3H,s),2.21-2.17(1H,m),2.06(3H,s),1.94-1.80(4H,m),1.68-1.10(20H,m),0.98(3H,s),0.92(3H,d,J=6.7Hz),0.90(3H,t,J=7.3Hz),0.63(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.6(1F,dt,J=49.9,12.4Hz)ppm。

LRMS(ESI+)m/z:666.3,[M+NH4]+,100%。

General procedure for deprotection of 3 alpha-acetate sulfonylureas 2

To the flask to which the protected sulfonylurea was added, NaOH in MeOH (5% solution, 10mL) was added and stirred for 16 h. Upon completion, the reaction was acidified to pH 7.0 with 1M HCl and diluted with EtOAc (10 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X 10 mL). The organic phases are combined and washed with NaHCO3The solution (50mL) was washed with MgSO4Drying, filtering and vacuum concentrating. The resulting product (in CH) was purified by column chromatography2Cl2MeOH in gradient elution, 0-5%) to give the deprotected sulfonylurea.

N, N' - (3 alpha, 7 alpha-dihydroxy-4 beta-fluoro-6 alpha-ethyl-24-nor-5 beta-cholan-23-yl) -p-toluenesulfonylurea (Compound 1)

Prepared according to general method 2 using 49.7mg of intermediate 1 to give compound 1 as a colorless residue (18.6mg, 40%).

1H NMR(400MHz,CDCl3):δ7.78(2H,d,J=8.3Hz),7.34(2H,d,J=8.1Hz),6.50(1H,t,J=4.8Hz),5.32(1H,ddd,J=50.0,10.0,9.2Hz),3.83(1H,s),3.59-3.51(1H,m),3.33-3.26(1H,m),3.21-3.12(1H,m),2.46(3H,s),1.96-1.06(26H,m),0.98(3H,s),0.94(3H,t,J=6.2Hz),0.93(3H,t,J=6.4Hz),0.66(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.8(1F,dt,J=50.3,10.4Hz)ppm。

LRMS(ESI+)m/z:624.4,[M+NH4]+,100%。

N, N' - (3 alpha, 7 alpha-dihydroxy-4 beta-fluoro-6 alpha-ethyl-24-nor-5 beta-cholan-23-yl) -benzenesulfonylurea (Compound 2)

Prepared according to general procedure 2 using 44.8mg of intermediate 2 to give compound 2 as a colorless residue (28.5mg, 64%).

1H NMR(400MHz,CDCl3):δ7.91(2H,d,J=7.5Hz),7.65(1H,t,J=7.3Hz),7.54(2H,t,J=7.8Hz),6.51(1H,s),5.31(1H,ddd,J=50.1,10.3,9.1Hz),3.82(1H,s),3.60-3.50(1H,m),3.35-3.26(1H,m),3.20-3.12(1H,m),1.95-1.36(17H,m),1.27-1.11(9H,m),0.97(3H,s),0.94(3H,d,J=6.2Hz),0.93(3H,t,J=6.7Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-189.0(1F,dt,J=50.3,12.1Hz)ppm。

LRMS(ESI+)m/z:610.2,[M+NH4]+,100%。

N, N' - (3 alpha, 7 alpha-dihydroxy-4 beta-fluoro-6 alpha-ethyl-24-nor-5 beta-cholan-23-yl) -4- (tert-butyl) benzenesulfonylurea (Compound 3)

Prepared according to general procedure 2 using 79.6mg of intermediate 3 to give compound 3 as a colorless residue (50.7mg, 65%).

1H NMR(400MHz,CDCl3):δ7.82(2H,d,J=8.6Hz),7.53(2H,t,J=8.4Hz),6.54(1H,s),5.32(1H,ddd,J=49.9,10.3,9.1Hz),3.82(1H,s),3.60-3.50(1H,m),3.35-3.25(1H,m),3.19-3.11(1H,m),1.95-1.41(16H,m),1.34(9H,s),1.28-1.08(10H,m),0.97(3H,s),0.930(3H,t,J=6.9Hz),0.927(3H,d,J=6.2Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.8(1F,dt,J=50.3,12.1Hz)ppm。

LRMS(ESI+)m/z:666.4,[M+NH4]+,100%。

N, N' - (3 alpha, 7 alpha-dihydroxy-4 beta-fluoro-6 alpha-ethyl-24-nor-5 beta-cholan-23-yl) -m-toluenesulfonylurea (Compound 4)

Prepared according to general method 2 using 83.7mg of intermediate 4 to give compound 4 as a colorless residue (29.0mg, 37%).

1H NMR(400MHz,CDCl3):δ7.71-7.69(2H,m),7.45-7.38(2H,m),6.52(1H,s),5.32(1H,ddd,J=49.9,10.4,8.9Hz),3.82(1H,s),3.60-3.50(1H,m),3.35-3.26(1H,m),3.20-3.10(1H,m),2.43(3H,s),1.95-1.39(17H,m),1.28-1.11(10H,m),0.97(3H,s),0.94(3H,d,J=6.2Hz),0.93(3H,t,J=6.5Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.9(1F,dt,J=48.6,10.4Hz)ppm。

LRMS(ESI+)m/z:624.3,[M+NH4]+,100%。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-toluenesulfonylurea (Compound 5)

Prepared according to general procedure 2 using 53.4mg of intermediate 5 to give compound 5 as a colorless residue (24.2mg, 48%).

1H NMR(400MHz,CDCl3):δ7.94(1H,d,J=8.1Hz),7.52(1H,td,J=7.6,1.1Hz),7.35(2H,d,J=7.7Hz),6.47(1H,t,J=4.6Hz),5.31(1H,ddd,J=49.9,10.4,8.9Hz),3.82(1H,s),3.60-3.50(1H,m),3.30-3.22(1H,m),3.17-3.08(1H,m),2.67(3H,s),1.94-1.06(25H,m),0.97(3H,s),0.91(3H,t,J=7.5Hz),0.90(3H,d,J=6.6Hz),0.63(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-189.0(1F,dt,J=48.6,12.1Hz)ppm。

LRMS(ESI+)m/z:624.3,[M+NH4]+,100%。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-fluorobenzenesulfonylurea (compound 6)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,MeOD):δ7.93-7.(2H,m),7.20-7.17(2H,m),5.19(1H,dq,J=49.3,10.5,8.9Hz),3.65(1H,s),3.31(1H,m),3.06(1H,m),2.95(1H,m),1.94-1.06(21H,m),0.84-0.76(9H,m),0.63(3H,s)ppm。

19F NMR(1H non-uncoupled, 376MHz, MeOD): delta-107.29 (1F, m), -186.6(1F, m) ppm.

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m-fluorobenzenesulfonyl urea (compound 7)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,CDCl3)δ7.71(ddd,J=7.8,1.6,1.0Hz,1H),7.61(br dt,J=8.0,2.1Hz,1H),7.54(td,J=8.1,5.3Hz,1H),7.35(tdd,J=8.3,2.6,0.7Hz,1H),6.50(br t,J=5.1Hz,1H),5.32(ddd,J=49.9,10.5,8.8Hz,1H),3.83(s,1H),3.55(dddd,J=14.2,12.0,8.8,5.1Hz,1H),3.32(ddt,J=13.1,9.5,5.1Hz,1H),3.18(dtd,J=13.0,8.0,6.1Hz,1H),1.96-1.86(m,2H),1.85-1.74(m,2H),1.71-1.58(m,6H),1.54-1.41(m,7H),1.31-1.08(m,9H),0.98(s,3H),0.95(d,J=6.5Hz,3H),0.93(t,J=7.0Hz,3H),0.66(s,3H)ppm。

19F NMR(376MHz,CDCl3)δ-108.9(br s,1F),-188.9(br d,J=50.3Hz,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)δ-108.9(s,1F),-188.9(s,1F)ppm。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-fluorobenzenesulfonyl urea (compound 8)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,CDCl3)δ7.91(ddd,J=7.8,7.2,1.7Hz,1H),7.67(dddd,J=8.3,7.5,5.0,1.7Hz,1H),7.33(td,J=7.7,1.1Hz,1H),7.28(ddd,J=10.0,8.4,0.9Hz,1H),6.45(br t,J=5.1Hz,1H),5.31(ddd,J=49.9,10.5,8.7Hz,1H),3.83(br s,1H),3.55(dddd,J=13.8,11.9,8.9,5.4Hz,1H),3.29(ddt,J=12.8,9.4,5.3Hz,1H),3.16(dtd,J=13.5,7.8,5.6Hz,1H),1.96-1.74(m,4H),1.71-1.39(m,14H),1.25-1.09(m,8H),0.98(s,3H),0.94(t,J=7.3Hz,3H),0.93(d,J=6.6Hz,3H),0.65(s,3H)ppm。

19F NMR(376MHz,CDCl3)δ-109.0(ddd,J=10.4,6.9,5.2Hz,1F),-189.2(dt,J=50.3,11.3Hz,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)δ-109.0(s,1F),-189.2(s,1F)ppm。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p- (trifluoromethyl) benzenesulfonylurea (Compound 9)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,CDCl3)δ8.07(d,J=8.2Hz,2H),7.80(d,J=8.1Hz,2H),6.46(br t,J=4.8Hz,1H),5.32(ddd,J=49.8,10.4,8.8Hz,1H),3.82(s,1H),3.55(dddd,J=14.2,11.9,8.8,5.4Hz,1H),3.28(ddt,J=13.6,7.7,4.9Hz,1H),3.15(dtd,J=13.5,7.6,6.0Hz,1H),1.96-1.75(m,4H),1.72-1.55(m,7H),1.53-1.37(m,7H),1.25-1.06(m,8H),0.97(s,3H),0.933(d,J=6.1Hz,3H),0.927(t,J=6.5Hz,3H),0.63(s,3H)ppm。

19F NMR(376MHz,CDCl3)δ-63.5(s,3F),-188.5(br d,J=48.6Hz,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)δ-63.4(s,3F),-188.6(br s,1F)ppm。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m- (trifluoromethyl) benzenesulfonylurea (Compound 10)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,CDCl3)δ8.17(s,1H),8.11(d,J=8.0Hz,1H),7.91(br d,J=7.7Hz,1H),7.71(t,J=7.9Hz,1H),6.49(br t,J=4.7Hz,1H),5.32(ddd,J=49.9,10.2,9.1Hz,1H),3.83(s,1H),3.56(dddd,J=14.1,11.7,8.7,5.1Hz,1H),3.32(ddt,J=13.5,9.4,5.3Hz,1H),3.17(dtd,J=12.6,7.8,6.2Hz,1H),1.96-1.87(m,2H),1.84-1.74(m,2H),1.72-1.58(m,6H),1.53-1.41(m,7H),1.29-1.09(m,9H),0.98(s,3H),0.95(d,J=6.5Hz,3H),0.93(t,J=6.9Hz,3H),0.66(s,3H)ppm。

19F NMR(376MHz,CDCl3)δ-63.1(s,3F),-189.0(br s,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)δ-63.1(s,3F),-189.0(br s,1F)ppm。

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o- (trifluoromethyl) benzenesulfonylurea (Compound 11)

This compound was prepared by a method similar to that described above for compounds 1-5.

1H NMR(400MHz,CDCl3)δ8.26(dd,J=6.3,2.3Hz,1H),7.92(dd,J=6.6,2,3Hz,1H),7.77(m,2H),6.35(br t,J=4.8Hz,1H),5.31(ddd,J=49.9,10.4,8.9Hz,1H),3.82(s,1H),3.56(dddd,J=14.2,12.1,8.7,5.1Hz,1H),3.29(ddt,J=13.5,9.1,5.1Hz,1H),3.13(dtd,J=13.5,7.7,6.2Hz,1H),1.96-1.72(m,5H),1.70-1.35(m,14H),1.25-1.06(m,7H),0.97(s,3H),0.93(d,J=7.0Hz,3H),0.92(t,J=6.5Hz,3H),0.63(s,3H)ppm。

19F NMR(376MHz,CDCl3)δ-58.0(s,3F),-188.9(br d,J=48.6Hz,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)δ-58.0(s,3F),-188.9(s,1F)ppm。

N, N' - (3 α, 7 α -dihydroxy-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (comparative compound A)

This compound was prepared by a similar method as described for compounds 5-9 above.

1H NMR(400MHz,MeOD):δ7.86-7.81(2H,m),7.53(1H,m),7.47-7.41(2H,m),3.52(1H,br.s),3.22(1H,m),3.04(1H,m),2.93(1H,m),1.87-0.83(25H,m),0.81-0.76(9H,m),0.52(3H,s)ppm。

Example 2 alternative Synthesis of Compounds with sulfonylurea-substituted side chains

The following methods are exemplified for 4 β -fluoro derivatives, but may also be used for 2 β -fluorinated, 4-difluorinated or 2, 4-difluorinated compounds.

A.6 alpha-ethyl-4 beta-fluoro-7 alpha-trimethylsilyloxy-3-oxo-5 beta-cholan-24-oic acid methyl ester

To a stirred pre-cooled diisopropylamine (0.78mL,5.54mmol,. about.12 equivalents) in dry THF (6.9mL) was added n-BuLi in hexane (1.44mL, 2.31mmol,. about.5.0 equivalents) dropwise over 0.25 h. After addition, trimethylsilyl chloride (0.29mL, 2.31mmol,. about.5.0 equiv.) was added and stirred for 1 h. A solution of methyl 6 α -ethyl-7 α -hydroxy-3-oxo-5 β -cholane-24-oate (200mg, 0.46mmol) from example 1, step B, in dry THF (3mL) and triethylamine (1.16mL, 8.32mmol, 18 equiv.) was then added. After the addition, the reaction was gradually warmed to-20 ℃ and stirred for 2 h. Upon completion, saturated NaHCO was added dropwise3The reaction was quenched (5mL) and warmed to RT for 2 h. The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X 10 mL). The combined organic phases were washed with brine (30mL) and MgSO4Drying, filtration and concentration in vacuo gave 271mg of crude material as a yellow residue.

To a solution of the crude material (1.16g, 2.3mmol) in dry MeCN (55mL) was added(1.23g, 3.47 mmol). After stirring at RT for 14.5h, the mixture was diluted with ethyl acetate (100mL) and 5% NaHCO3(100mL) and 10% NaCl (50 mL). The aqueous phase was extracted with ethyl acetate (3X 100mL) and MgSO4The combined organic phases were dried, filtered and concentrated in vacuo to give an orange/yellow oil. Purification of the crude material (SiO) by column chromatography20-40% EtOAc in heptane) to give the title compound as a colorless oil (319.5 mg).

B.6 alpha-Ethyl-4 beta-fluoro-7 alpha-trimethylsilyloxy-3 alpha-hydroxy-5 beta-cholan-24-oic acid methyl ester

Crude methyl 6 α -ethyl-4 β -fluoro-7 α -trimethylsiloxy-3-oxo-5 β -cholan-24-oate (319.5mg, 0.71mmol) from step a was dissolved in THF (28mL) under an argon atmosphere while stirring. Adding NaBH4(80.5mg, 2.13mmol), the reaction was stirred at RT for 16.5h, after which time NaBH was added4(0.24g, 6.38 mmol). The mixture was stirred for a further 4.5h, then water (20 μ L) was added and the mixture was stirred for-60 h. After this time, the reaction was quenched by addition of water (15mL) and diluted with EtOAc (50 mL). The phases were separated and the aqueous phase was extracted with EtOAc (3 × 50 mL). With MgSO4The combined extracts were dried, filtered and concentrated in vacuo to give a clear syrup (0.34 g). Purification of the crude material (SiO) by column chromatography20-40% EtOAc in heptane) to give the title compound as a clear oil (162.3 mg).

C.6 alpha-Ethyl-4 beta-fluoro-7 alpha-trimethylsilyloxy-3 alpha-o-tert-butyldimethylsilyl-5 beta-cholan-24-oic acid methyl ester

Methyl 6 α -ethyl-4 β -fluoro-7 α -trimethylsilyloxy-3 α -hydroxy-5 β -cholan-24-oate (0.48g, 0.92mmol) from step B was dissolved in dry DCM (12mL) and cooled to 0 ℃ under an argon atmosphere while stirring. 2, 6-lutidine (1.1mL, 9.17mmol) was added followed by TBMDS-OTf (0.32mL, 1.38mmol) dropwise. The reaction was warmed to RT, stirred for 24h, then cooled to 0 ℃ and quenched by the dropwise addition of 10% citric acid (5 mL). The phases were separated and the aqueous phase was extracted with DCM (3 × 5 mL). With 10% citric acid (5mL), NaHCO3The combined extracts were washed with aqueous (5mL) and water (5mL) and MgSO4Dried, filtered and concentrated in vacuo to give a yellow oil (0.69 g). Purification of the crude material (SiO) by column chromatography20-20% EtOAc in heptane) to give the title compound as a clear oil (0.58 g).

6 α -Ethyl-4 β -fluoro-7 α -trimethylsiloxy-3 α -o-tert-butyldimethylsilyl-5 β -cholanic acid

Stirring while stirring 6 alpha-ethyl-4 beta-fluoro-7 alpha-trimethylsiloxy-3 alpha-o-tert-butyl ether from step CButyldimethylsilyl-5 β -cholan-24-oic acid methyl ester (0.58g) was dissolved in IPA (5.8 mL). 0.5M NaOH (5.8mL) was added and the reaction was stirred at RT for 15 h. The reaction mixture was concentrated under reduced pressure to-half volume, then water (5mL) was added by the addition of 2M H2SO4The solution was neutralized and diluted with EtOAc (10 mL). With 2M H2SO4The mixture was acidified to pH1, the phases were separated and the aqueous phase was extracted with EtOAc (10 mL). The combined extracts were washed with water (5mL) and brine (5mL), MgSO4Drying, filtration and concentration in vacuo gave a white foam (0.52 g). Purification of the crude material (SiO) by column chromatography20-50% acetone in toluene) to give the title compound as a white solid (0.41g, 72%).

E.6 alpha-Ethyl-4 beta-fluoro-7 alpha-trimethylsiloxy-3 alpha-o-tert-butyldimethylsilyl-5 beta-cholan-24-acyl azide

To a stirred solution of 6 α -ethyl-4 β -fluoro-7 α -trimethylsiloxy-3 α -o-tert-butyldimethylsilyl-5 β -cholanic acid from step D (197mg, 0.32mmol) in dry THF (3.2mL) was added dropwise Et under argon atmosphere at RT3N (0.09mL, 0.64mmol,. about.2.0 equiv). After addition, the reaction mixture was cooled to 0 ℃ and diphenylphosphoryl azide (0.1mL, 0.48mmol,. about.1.5 equivalents) was added dropwise. After addition, the reaction mixture was stirred for 2.5h behind a strong gas flow hood. Upon completion, the reaction was quenched with brine (3mL) and extracted with DCM (3 × 5 mL). With MgSO4The combined organic phases were dried, filtered and concentrated in vacuo at 0 ℃. The resulting oil was used without further purification.

General procedure 3 for the formation of sulfonylureas

Stirring in an argon atmosphere from example 2A solution of the crude 6 α -ethyl-4 β -fluoro-7 α -trimethylsilyloxy-3 α -o-tert-butyldimethylsilyl-5 β -cholan-24-acyl azide from step E (69mg) in dry toluene (2.1mL) was heated to 125 ℃. After 5h, the reaction was cooled to RT. The resulting solution was used without further purification. The solution was stirred under an argon atmosphere and sulfonamide (1.5 equiv.) and DBU (1.5 equiv.) were added. Upon completion, the reaction was quenched by dropwise addition of 1M HCl (1mL) and diluted with EtOAc (5 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X5 mL). The combined organic phases were washed with brine (3mL) and MgSO4Dried, filtered and concentrated in vacuo (231.7 mg). The resulting product is purified by column chromatography to give the desired sulfonylurea as a crude inseparable mixture.

N, N '- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trimethoxy) benzenesulfonylurea (Compound 12) and N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (Compound 13)

Compounds 12 and 13 were prepared according to general procedure 3 above by reacting the crude isocyanate product with 4- (trifluoromethoxy) benzenesulfonamide and 4- (methoxy) benzenesulfonamide, respectively.

To obtain pure product, crude compounds 12 and 13 were converted to protected species (intermediates 12 and 13), purified, and then deprotected to regenerate compounds 12 and 13. The method is as follows.

N, N' - (3 α, 7 α -di-o-tert-butyldimethylsilyl-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (intermediate 12)

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (24.3mg, 0.036mmol) was dissolved in dry DCM (1mL) and cooled to 0 ℃ under an argon atmosphere while stirring. Adding 2, 6-lutidine (0.04mL, 0.36mmol), and adding TBMDS-OTf (0.02 mmol) dropwisemL, 0.108 mmol). The reaction was warmed to RT, stirred for 1.5h, then cooled to 0 ℃ and quenched by the dropwise addition of 10% citric acid (1 mL). The phases were separated and the aqueous phase was extracted with DCM (3 × 1 mL). With 10% citric acid (1mL), NaHCO3The combined extracts were washed with aqueous (1mL) and water (1mL) over MgSO4Dried, filtered and concentrated in vacuo to give a yellow oil. Purification of the crude material (SiO) by column chromatography20-50% EtOAc in heptane) to give the title compound as a clear oil (9.4mg, 33%).

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (Compound 12)

N, N' - (3 α, 7 α -di-o-tert-butyldimethylsilyl-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (9.4mg) was dissolved in dry THF (1mL) under an argon atmosphere while stirring. A1M solution of TBAF in THF (31. mu.L, 0.03mmol) was added and the reaction stirred at RT for 6 days. The crude solution was dried on silica gel and purified by column chromatography (SiO)250-100% EtOAc in heptane) to give the title compound (1 mg).

N, N' - (3 α, 7 α -di-o-tert-butyldimethylsilyl-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (intermediate 13)

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (26.1mg, 0.042mmol) was dissolved in dry DCM (1mL) and cooled to 0 ℃ under an argon atmosphere while stirring. 2, 6-lutidine (0.05mL, 0.419mmol) was added followed by TBMDS-OTf (0.03mL, 0.126mmol) dropwise. The reaction was warmed to RT, stirred for 16h, then cooled to 0 ℃ by dropwise addition of 10% citric acid (1 mL). The phases were separated and the aqueous phase was extracted with DCM (3 × 1 mL). With 10% citric acid (1mL), NaHCO3The combined extracts were washed with aqueous (1mL) and water (1mL) over MgSO4Dried, filtered and concentrated in vacuo to give a yellow oil (28.1 mg). Purification of the crude material (SiO) by column chromatography20-80% MeOH in DCM) and then repurified by column chromatography (SiO)20-50% acetone in toluene) to give the title compound (7.8 mg).

N, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxy and sulfonylurea (Compound 13)

N, N' - (3 α, 7 α -di-o-tert-butyldimethylsilyl-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (7.8mg) was dissolved in dry THF (1mL) under an argon atmosphere while stirring. A1M THF solution of TBAF (28. mu.L, 0.03mm0L) was added and the reaction was stirred at RT for 17 h. The crude solution was dried on silica gel and purified by column chromatography (SiO)20-80% acetone in toluene) to give the title compound (3.4mg, 59.6%).

1H NMR(400MHz,CDCl3):δ7.97-7.81(2H,m),7.11-7.02(2H,m),5.30(1H,ddd,J=49.6,10.3,8.9Hz),3.88(3H,s),3.76(1H,br.s),3.42(1H,m),3.15(1H,m),3.06(1H,m),1.99-0.96(26H,m),0.95-0.80(6H,m),0.64(3H,s)ppm。

EXAMPLE 3 Synthesis of Compounds with sulfonamide-substituted side chains

The following method exemplifies 4 β -fluoro derivatives, but may also be used for 2 β -fluorinated, 4-difluorinated or 2, 4-difluorinated compounds. Steps A and B are as described, for example, in FIG. 2.

C.6 alpha-ethyl-4 beta-fluoro-7 alpha-trimethylsiloxy-3 alpha-hydroxy-5 beta-cholanic acid

Methyl 6 α -ethyl-4 β -fluoro-7 α -trimethylsilyloxy-3 α -hydroxy-5 β -cholan-24-oate (162.3mg) from step B was dissolved in IPA (1.6mL) with stirring. 0.5M NaOH (1.6mL) was added and the reaction was stirred at RT for 16 h. The reaction mixture was concentrated under reduced pressure to-half volume, then water (5mL) was added by the addition of 2M H2SO4The solution was neutralized and diluted with EtOAc (10 mL). With 2M H2SO4The mixture was acidified to pH1, the phases were separated and the aqueous phase was extracted with EtOAc (10 mL). The combined extracts were washed with water (3mL) and brine (5mL), MgSO4Dried, filtered and concentrated in vacuo to give a white foam (151.1 mg). Purification of the crude material (SiO) by column chromatography20-80% EtOAc in heptane) to give the title compound as a clear oil (164.1 mg).

General procedure for acyl sulfonamide side chain formation 4

6 α -Ethyl-4 β -fluoro-7 α -trimethylsiloxy-3 α -hydroxy-5 β -cholanic acid (50mg, 0.11mmol) was dissolved in dry DCM (2 mL). EDCI (43.7mg, 0.23mmol) and DMAP (27.8mg, 0.23mmol) were added followed by the appropriate sulfonamide (3 equiv.). After stirring overnight at RT, water (5mL) was added, the phases separated and the aqueous phase extracted with DCM (2 × 5 mL). The combined extracts were washed with 1M HCl (2mL) and brine (2mL), MgSO4Drying, filtering, and vacuum concentrating to obtain crude material as yellow-white solid.

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -p-trifluoromethoxybenzenesulfonamide (compound 14)

Stirring while under argonCrude N- (6 α -ethyl-4 β -fluoro-7 α -trimethylsilyloxy-3 α -hydroxy-5 β -cholan-24-yl) -trifluoromethoxy benzenesulfonamide (81.6mg), obtained using trimethoxybenzenesulfonamide according to general procedure 4, was dissolved in dry THF (5mL) under an atmosphere. A1M solution of TBAF in THF (0.48mL, 0.48mmol) was added and the reaction stirred at RT for 17.5 h. The crude solution was dried on silica gel and purified by column chromatography (SiO)20-100% EtOAc in heptane). Fractions containing the desired product were combined, concentrated in vacuo, dissolved in EtOAc (5mL) and washed with 2M HCl (5 mL). The aqueous phase was extracted with EtOAc (2X 5mL) over MgSO4The combined extracts were dried, filtered and concentrated in vacuo to give a white solid which was purified by column chromatography (SiO)20-25% acetone in toluene) to give the title compound as a clear residue (4.9 mg).

1H NMR(400MHz,CDCl3):δ8.21-8.10(2H,m),7.38-7.36(2H,dd,J=8.9,0.8Hz),5.31(1H,ddd,J=49.8,10.4,9.0Hz),3.82(1H,br.s),3.56(1H,m),2.31(1H,ddd,J=15.6,10.1,5.0Hz),2.17(1H,m),1.92-1.07(23H,m),0.97(3H,s),0.93(3H,t,J=6.9Hz),0.86(3H,d,J=6.4Hz),0.62(3H,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -p-fluorobenzenesulfonamide (compound 15)

This compound was prepared by a route analogous to that described above for compound 14.

1H NM[R(400MHz,CDCl3)δ8.11(m,2H),7.23(m,2H),5.31(ddd,J=49.9,10.3,9.1Hz,1H),3.81(br s,1H),3.57(dddd,J=13.8,11.6,8.6,5.1Hz,1H),2.29(ddd,J=15.4,10.0,5.3Hz,1H),2.17(ddd,J=15.7,9.2,6.4Hz,1H),1.90(dt,J=12.3,2.9Hz,1H),1.87-1.57(m,9H),1.53-1.29(m,8H),1.24-1.08(m,7H),0.97(s,3H),0.93(t,J=7.0Hz,3H),0.86(d,J=6.4Hz,3H),0.61(s,3H)ppm。

19F NMR(376MHz,CDCl3)-103.1(br s,1F),-188.8(br s,1F)ppm。

19F{1H}NMR(376MHz,CDCl3)-103.1(br s,1F),-188.8(br s,1F)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholane-24-acyl) -m-fluorophenyl sulfonamide (Compound 16)

This compound was prepared by a route analogous to that described above for compound 14.

1H NMR(400MHz,CDCl3)8.39(1H,br s),7.89(1H,br d,J=7.8Hz),7.78(1H,br d,J=7.7Hz),7.55(1H,td,J=8.1,5.4Hz),7.36(1H,td,J=8.3,1.3Hz),5.31(1H,dt,J=50.0,9.8Hz),3.82(1H,br s),3.56(1H,dddd,J=14.2,11.0,8.7,5.1Hz),2.31(1H,ddd,J=15.0,9.9,5.1Hz),2.18(1H,ddd,J=15.3,9.5,7.2Hz),1.99-1.03(25H,m),0.97(3H,s),0.94(3H,t,J=6.6Hz),0.87(3H,d,J=8.1Hz),0.62(3H,s)ppm。

19F NMR(376MHz,CDCl3)-109.52(1F,br d,J=5.2Hz),-189.0(1F,dt,J=50.1,13.9Hz)ppm。

19F{1H}NMR(376MHz,CDCl3)-109.52(1F,s),-189.00(1F,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholane-24-acyl) -o-fluorophenyl sulfonamide (Compound 17)

This compound was prepared by a route analogous to that described above for compound 14.

1H NMR(400MHz,CDCl3)δ9.06(1H,br s),8.10(1H,td,J=7.5,1.5Hz),7.65(1H,m),7.35(1H,t,J=7.6Hz),7.22(1H,t,J=9.2Hz),5.31(1H,dt,J=50.1,9.5Hz),3.81(1H,br s),3.57(1H,dddd,J=14.1,11.3,8.6,5.1Hz),2.34(1H,ddd,J=15.4,10.0,5.0Hz),2.21(1H,ddd,J=15.9,9.3,6.6Hz),2.08-1.02(25H,m),0.96(3H,s),0.92(3H,br t,J=6.9Hz),0.86(3H,d,J=6.2Hz),0.61(3H,s)ppm。

19F NMR(376MHz,CDCl3)-110.0(1F,br s),-189.1(1F,d,J=46.8Hz)ppm。

19F{1H}NMR(376MHz,CDCl3)-109.8(1F,s),-188.9(1F,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -4-trifluoromethylphenylsulfonamide (Compound 18)

This compound was prepared by a route analogous to that described above for compound 14.

1H NMR(400MHz,CDCl3)8.22(2H,d,J=8.3Hz),8.00(1H,br s)7.83(2H,d,J=8.4Hz),5.30(1H,dtd,J=50.1,10.2,1.6Hz),3.81(1H,br s),3.56(1H,dddd,J=13.9,12.0,8.6,5.0Hz),2.31(1H,ddd,J=15.8,10.3,5.1Hz),2.17(1H,ddd,J=15.8,10.0,6.6Hz),1.94-1.05(25H,m),0.97(3H,s),0.93(3H,t,J=5.8Hz),0.87(3H,d,J=6.1Hz),0.61(3H,s)ppm。

19F NMR(376MHz,CDCl3)-63.5(3F,br s),-189.2(1F,br d,J=48.6Hz)ppm。

19F{1H}NMR(376MHz,CDCl3)-63.3(3F,s),-189.0(1F,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -3-trifluoromethylphenylsulfonamide (Compound 19)

This compound was prepared by a route analogous to that described above for compound 14.

1H NMR(400MHz,CDCl3)8.32(2H,m),8.22(1H,br s),7.92(1H,br d,J=7.8Hz),7.72(1H,t,J=7.9Hz),5.30(1H,dtd,J=49.5,9.3,1.2Hz),3.81(1H,br s),3-54(1H,m),2.31(1H,ddd,J=15.6,10.1,5.1Hz),2.18(1H,ddd,J=15.9,9.2,6.4Hz),1.95-1.04(25H,m),0.97(3H,s),0.93(3H,t,J=6.9Hz),0.86(3H,d,J=6.2Hz),0.61(3H,s)ppm。

19F NMR(376MHz,CDCl3)-63.02(3F,s),-189.11(1F,br d,J=48.6Hz)ppm。

19F{1H}NMR(376MHz,CDCl3)-63.02(3F,s,CF3),-189.10(1F,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -2-trifluoromethylphenylsulfonamide (Compound 20)

This compound was prepared by a route analogous to that described above for compound 14.

1H NMR(400MHz,CDCl3)8.53(1H,m),8.09(1H,br s),7.90(1H,m),7.79(2H,m),5.3(1H,dtd,J=50.0,9.8,1.7Hz),3.81(1H,br s),3.55(1H,dddd,J=14.2,11.7,8.8,5.0Hz),2.31(1H,ddd,J=15.3,9.7,4.8Hz),2.17(1H,m),1.91(1H,m),1.87-1.05(24H,m),0.97(3H,s),0.93(3H,t,J=6.9Hz),0.86(3H,d,J=6.2Hz),0.61(3H,s)ppm。

19F NMR(376MHz,CDCl3)-63.02(3F,s),-189.11(1F,br d,J=48.6Hz)ppm。

19F{1H}NMR(376MHz,CDCl3)-63.02(3F,s),-189.10(1F,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -cyclopropylsulfonamide (comparative compound B)

N- (3 α -hydroxy-4 β -fluoro-6 α -ethyl-7 α -trimethylsilyloxy-5 β -cholan-24-yl) -cyclopropylsulfonamide (50mg) was dissolved in dry THF under an argon atmosphere while stirring. A1M solution of TBAF in THF (0.3mL, 0.3mmol) was added and the reaction stirred at RT for 23 h. The reaction was diluted with EtOAc (20mL) and washed with water (10mL) and 10% aqueous NaCl. The crude solution was dried on silica gel and purified by column chromatography (SiO)20-50% acetone in toluene) to give the title compound (5.4 mg).

Rf 0.65 (EtOAc/heptane, 50: 50).

1H NMR(400MHz,CDCl3):δ5.31(1H,ddd,J=49.8,10.7,8.9Hz),3.83(1H,br.s),3.55(1H,m),2.95(1H,tt,J=8.1,4.8Hz),2.39(1H,m),2.25(1H,m),2.01-1.07(27H,m),0.98(3H,s),0.95(3H,d,J=6.5Hz),0.94(3H,t,J=7.1Hz),0.67(3H,s)ppm。

N- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -methylsulphonamide (comparative compound C)

N- (3 α -hydroxy-4 β -fluoro-6 α -ethyl-7 α -trimethylsilyloxy-5 β -cholan-24-yl) -methanesulfonamide (50mg) was dissolved in dry THF under an argon atmosphere while stirring. A1M solution of TBAF in THF (0.3mL, 0.3mmol) was added and the reaction was stirred at RT for 16 h. The reaction was diluted with EtOAc (5mL) and washed with brine (3 mL). The crude solution was dried on silica gel and purified by column chromatography (SiO)20-50% acetone in toluene). The fractions containing the desired product were combined, concentrated under reduced pressure, dissolved in CDCl3Washed with 2M HCl and water, filtered through a PTFE filter and concentrated in vacuo to give the title compound (7.1 mg).

1H NMR(400M[Hz,CDCl3):δ5.30(1H,ddd,J=49.8,10.4,8.8Hz),3.83(1H,br.s),3.56(1H,m),3.30(3H,s),2.39(1H,ddd,J=15.6,10.3,5.1Hz),2.25(1H,m),2.00-1.08(23H,m),0.97(3H,s),0.94(3H,d,J=6.5Hz),0.93(3H,t,J=6.8Hz),0.67(3H,s)ppm。

Example 4-4, 4-difluoro-3 α, 7 α -dihydroxy-6 α -ethyl-5 β - - Synthesis of cholanic acid analogs

A.6 alpha-ethyl-4, 4-difluoro-7 alpha-hydroxy-3-oxo-5 beta-cholan-24-oic acid methyl ester

To a pre-cooled solution of methyl 6 α -ethyl-4 β -7 α -hydroxy-3-oxo-5 β -cholan-24-oate (product of example 1C; 7.30g, 16.0mmol) in dry THF (300mL) was added dropwise a solution of LDA in hexane (21.1mL, 21.1mmol,. about.1.3 equiv) at-78 ℃ under an argon atmosphere over 0.25 h. After addition, trimethylsilyl chloride (2.70mL, 21.1mmol,. about.1.3 equiv.) was added as a solution in dry THF (150mL) and stirred for 1 h. Upon completion, saturated NaHCO was added dropwise3The reaction was stopped by solution (300mL) and warmed to RT for 0.25 h. The organic phase was removed and the aqueous phase was back-extracted with dichloromethane (2X150 mL). The combined organic phases were washed with brine (300mL) and MgSO4Drying, filtration and concentration in vacuo afforded the crude material as a yellow residue (3% contamination with 6 α -ethyl-4 β -fluoro-7 α -hydroxy-3-oxo-5 β -cholan-24-oic acid methyl ester by19F NMR). The resulting residue was used in the next reaction without further purification.

19F NMR (1H non-uncoupled, 376 MHz): delta-135.2 (1F),s)。

To a stirred solution of the resulting crude material in MeCN (360mL) was added (11.4g, 32.0mmol,. about.2.0 eq.) and stirred for 16 h. Upon completion, the reaction mixture was concentrated in vacuo. The residue was dissolved in dichloromethane (500mL) and H2O (500 mL). The organic phase was removed and the aqueous phase was back-extracted with dichloromethane (2 × 250 mL). The combined organic phases were washed with brine (250mL) and MgSO4Drying, filtration and concentration in vacuo gave the crude material as a yellow residue. The resulting residue was used in the next reaction without further purification.

1H NMR(400MHz,CDCl3):δ3.70(1H,dq,J=7.7,3.2Hz),3.66(3H,s),2.70(1H,tdd,J=14.2,5.1,3.4Hz),2.44(1H,dq,J=15.2,3.8Hz),2.35(1H,ddd,J=15.5,10.2,5.4Hz,2.24(1H,dd,J=9.6,6.5Hz),2.19(1H,dd,J=10.2,2.9Hz),2.14(1H,dt,J=17.0,5.6Hz),2.08(1H,td,J=14.6,5.8Hz),2.00-1.86(3H,m),1.84-1.75(3H,m),1.73-1.63(3H,m),1.58(1H,dd,J=13.9,4.8Hz),1.54-1.28(6H,m),1.25-1.11(3H,m),1.10(3H,s),0.98(3H,t,J=7.3Hz),0.93(3H,d,J=6.4Hz),0.67(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-99.2(1F,dd,J=263.6,17.3Hz),-100.7(1F,ddd,J=263.6,29.5,15.6Hz)ppm。

LRMS(ESI+)m/z:486.6,[M+NH4]+,100%。

B.4, 4-difluoro- (3 α, 7 α) -dihydroxy-6 α -ethyl-5 β -cholan-24-oic acid methyl ester

To a stirred solution of crude methyl 6 α -ethyl-4, 4-difluoro-7 α -hydroxy-3-oxo-5 β -cholan-24-oate (7.51g of approved, 16.0mmol) from step A in dry methanol (500mL) at RT was added NaBH4(3.03g, 80.1mmol,. about.5.0 equivalents), and stirred under an argon atmosphere for 72 h. Upon completion, the reaction was concentrated in vacuo. The residue was dissolved in dichloromethane (500mL) and H2O (500 mL). The organic phase was removed and the aqueous phase was back-extracted with dichloromethane (2 × 250 mL). The combined organic phases were washed with brine (250mL) and MgSO4Drying with SiO2Filtration and concentration in vacuo gave 7.63g of crude material as a colorless residue. Purification by flash chromatography (Biotage SNAP KP-Sil 100g column) using hexane/acetone (100/0-80/20) as eluent gave the title compound 4, 4-difluoro- (3 α, 7 α) -dihydroxy-6 α -ethyl-5 β -cholan-24-oic acid methyl ester as a colorless residue (3.09g, 6.57mmol, 41%, 3 steps).

1H NMR(400MHz,CDCl3):δ3.76-3.65(2H,m),3.67(3H,s),2.36(1H,ddd,J=15.5,10.3,5.4Hz),2.31-2.19(2H,m),2.11(1H,d,J=5.4Hz),2.00-1.92(3H,m),1.91-1.30(16H,m),1.22-1.10(4H,m),1.04(3H,s),0.97(3H,t,J=7.3Hz),0.92(3H,d,J=6.5Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-99.3(1F,d,J=239.3Hz),-111.4(1F,dtd,J=239.3,34.7,22.5Hz)ppm。

LRMS(ESI+)m/z:488.6,[M+NH4]+,100%。

3 alpha, 7 alpha-dihydroxy-4, 4-difluoro-6 alpha-ethyl-5 beta-cholanic acid

To a stirred solution of methyl 4, 4-difluoro- (3 α, 7 α) -dihydroxy-6 α -ethyl-5 β -cholan-24-oate (1.77g, 3.75mmol, 1.0 equiv.) from step B in 1, 4-dioxane (95mL) and water (35mL) was added at RT a concentrated (37%) saltAcid (11mL, 9: 3: 1 ratio). After refluxing for 1h, the reaction mixture was cooled to RT and taken up with saturated NaHCO3The solution (50mL) was neutralized. The organic phase was removed and the aqueous phase was back-extracted with dichloromethane (3 × 50 mL). The combined organic phases were washed with brine (200mL) and MgSO4Drying, filtration and concentration in vacuo gave 1.84g of crude material as a brown oil. Purification by flash chromatography (Biotage SNAP KP-Sil 50g column) using hexane/acetone (100/0-90/10) as eluent gave the title compound 3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholanic acid as a colorless oil (1.51g, 3.30mmol, 88%).

1H NMR(400MHz,CDCl3):δ3.78-3.64(2H,m),2.39(1H,ddd,J=15.8,10.3,5.3Hz),2.25(1H,ddd,J=15.7,9.6,6.4Hz),,1.98-1.93(3H,m),1.86-1.65(7H,m),1.60(1H,d,J=13.0Hz),1.49-1.41(5H,m),1.40-1.29(3H,m),1.25-1.16(6H,m),1.04(3H,s),0.96(3H,t,J=7.3Hz),0.93(3H,d,J=6.4Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-99.1(1F,d,J=239.3Hz),-111.1(1F,dtd,J=241.0,38.2,19.1Hz)ppm。

LRMS(ESI+)m/z:474.6,[M+NH4]+,100%。

3 alpha-acetoxy-4, 4-difluoro-6 alpha-ethyl-7 alpha-hydroxy-5 beta-cholanic acid

To a stirred solution of 3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholanic acid (400mg, 0.88mmol, 1.0 equiv.) from step C in dry THF (50mL) was added sodium bicarbonate (370mg, 4.38mmol,. about.5.0 equiv.) and acetic anhydride (0.41mL, 4.38mmol,. about.5.0 equiv.) at HRT. After 16H at 70 ℃ the reaction mixture was cooled to RT and the reaction mixture was cooled by slow addition of H2O (50mL) quench. The organic phase was removed and the aqueous phase was back-extracted with EtOAc (2X50 mL). Combining the organic phases and usingAnd NaHCO3The solution (100mL) was washed with MgSO4Drying, filtration and concentration in vacuo gave 462mg of crude material as a yellow oil. Purification by flash chromatography (Biotage SNAP KP-Sil Ultra 25g column) using hexane/acetone (100/0-80/20) as eluent gave the title compound 3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-5 β -cholanic acid as a colorless oil (270mg, 0.54mmol, 62%).

1H NMR(400MHz,CDCl3):δ4.97(1H,ddd,J=27.5,10.2,6.0Hz),3.67(1H,s),2.54-2.33(2H,m),2.30-2.26(1H,m),2.24-2.18(1H,m),2.13(3H,s),2.02-1.79(6H,m),1.77-1.62(3H,m),1.56-1.42(6H,m),1.41-1.21(5H,m),1.20-1.13(3H,m),1.05(3H,s),0.95(3H,t,J=7.3Hz),0.94(3H,d,J=6.5Hz),0.66(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-98.2(1F,d,J=244.5Hz),-107.1(1F,ddd,J=242.8,36.4,22.5Hz)ppm。

LRMS(ESI+)m/z:516.5,[M+NH4]+,100%。

3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-5 β -cholan-24-acyl-azides

To a pre-cooled solution of 3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-5 β -cholanic acid (240mg, 0.48mmol, 1.0 equiv.) from step D in dry THF (5.3mL) at 0 ℃ were added triethylamine (0.14mL, 0.96mmol,. about.2.0 equiv.) and diphenylphosphoryl azide (0.16mL, 0.72mmol,. about.1.5 equiv.). After 3h, the reaction mixture was quenched by slow addition of brine (10 mL). The organic phase was removed and the aqueous phase was back-extracted with dichloromethane (3X20 mL). The organic phases were combined and MgSO4Drying, filtration and concentration in vacuo at 0 ℃ gave the crude material as a yellowish white oil. The resulting oil was used without further purificationIn the next reaction.

1H NMR-characteristic peaks (400MHz, CDCl)3):δ4.94-4.83(1H,m),3.59(1H,s),2.30(1H,ddd,J=15.7,10.0,5.4Hz),2.04(3H,s),0.97(3H,s),0.87(3H,t,J=7.3Hz),0.84(3H,d,J=6.4Hz),0.57(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-98.2(1F,d,J=242.8Hz),-107.2(1F,dtd,J=242.8,33.0,26.0Hz)ppm。

F.3 alpha-acetoxy-4, 4-difluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl isocyanate

A stirred solution of the crude oily 3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-5 β -cholan-24-ylazide (252mg putative, 0.48mmol) from step E in dry toluene (7.5mL) was heated to 125 ℃ under an argon atmosphere. After 4h, the reaction was cooled to RT. The resulting solution was used without further purification.

1H NMR-characteristic peaks (400MHz, CDCl)3):δ4.89(1H,ddd,J=27.510.2,6.5Hz),3.59(1H,q,J=3.2Hz),3.27(1H,ddd,J=13.1,7.8,4.5Hz),3.22-3.15(1H,m),2.04(3H,s),0.97(3H,s),0.88(3H,t,J=7.1Hz),0.86(3H,d,J=6.4Hz),0.59(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-98.2(1F,d,J=242.8Hz),-107.2(1F,dtd,J=243.2,37.7,22.5Hz)ppm。

G.N, N' - (3 alpha-acetoxy-4, 4-difluoro-6 alpha-ethyl-7 alpha-hydroxy-24-nor-5 beta-cholan-23-yl) -benzenesulfonylureas

Prepared according to general method 1 using 113mg benzenesulfonamide to give the title compound N, N' - (3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea as a white residue (235mg, 0.36mmol, 75%).

1H NMR(400MHz,CDCl3):δ7.90(2H,dd,J=8.4,1.1Hz),7.63(1H,tt,J=7.6,1.1Hz),7.49(2H,t,J=8.1Hz),6.53(1H,s),4.98(1H,ddd,J=27.1,11.3,4.5Hz),3.67(1H,s),3.27(1H,ddd,J=13.5,8.9,4.5Hz),3.18-3.11(1H,m),2.13(3H,s),2.02-1.79(6H,m),1.75-1.64(4H,m),1.62-1.43(5H,m),1.42-1.36(1H,m),1.28-1.10(7H,m),1.05(3H,s),0.95(3H,t,J=7.3Hz),0.92(3H,d,J=6.5Hz),0.63(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-98.2(1F,d,J=242.8Hz),-107.1(1F,dtd,J=243.2,37.7,22.5Hz)ppm。

LRMS(ESI+)m/z:635.8,[M+NH4]+,100%。

N, N' - (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (Compound 21)

Prepared according to general method 2 using 210mg of N, N '- (3 α -acetoxy-4, 4-difluoro-6 α -ethyl-7 α -hydroxy-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea to give the title compound N, N' - (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea as a white solid (102mg, 0.17mmol, 52%).

1H NMR(400MHz,CDCl3):δ8.19(1H,s),7.90(2H,dt,J=7.3,1.3Hz),7.65(1H,tt,J=7.5,1.1Hz),7.54(2H,tt,J=7.3,1.6 Hz),6.51(1H,t,J=5.2Hz),3.78-3.67(2H,m),3.29(1H,ddt,J=13.9,10.2,5.1Hz),3.18(1H,ddd,J=13.6,8.0,6.1Hz),2.30(1H,dd,J=31.4,11.0Hz),1.99-1.94(3H,m),1.88-1.57(9H,m),1.52-1.37(5H,m),1.23-1.11(7H,m),1.04(3H,s),0.97(3H,t,J=7.3Hz),0.93(3H,d,J=6.5Hz),0.64(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-99.3(1F,d,J=239.3H[z),-111.3(1F,dtd,J=239.3Hz)ppm。

LRMS(ESI+)m/z:593.6,[M+NH4]+,100%。

N- (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholan-24-yl) -benzenesulfonamide (compound 22)

Prepared according to general method 4 using 400mg of 3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholanic acid from step C to give the title compound N- (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholan-24-yl) -benzenesulfonamide as a white solid (161mg, 0.27mmol, 31%).

1H NMR(400MHz,CDCl3):δ8.06(2H,dd,J=7.6,1.3Hz),7.64(1H,tt,J=7.6,0.8Hz),7.54(2H,t,J=7.3Hz),3.85-3.69(2H,m),2.45(1H,dd,J=32.1,11.0Hz),2.27(1H,ddd,J=15.3,10.2,5.0Hz),2.16-2.10(1H,m),1.97-1.89(3H,m),1.86-1.64(8H,m),1.61-1.54(1H,m),1.47-1.43(4H,m),1.37-1.29(2H,m),1.22-1.06(6H,m),1.03(3H,s),0.97(3H,t,J=7.3Hz),0.84(3H,d,J=6.2Hz),0.59(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-99.0(1F,d,J=239.3Hz),-111.0(1F,dtd,J=239.3,33.0,26.0Hz)ppm。

LRMS(ESI+)m/z:613.6,[M+NH4]+,100%。

Example 56 α -Ethyl-2 α/β, 4 β -difluoro- (3 α, 7 α) -dihydroxy-5 with sulfonylurea and sulfonamide side chains Synthesis of beta-cholanic acid analogs

6 α -Ethyl-2 α/β, 4 β -difluoro-7 α -hydroxy-3-oxo-5 β -cholan-24-oic acid methyl ester

To a stirred, pre-cooled solution of 1M LDA in THF/hexane (1.63mL, 1.625mmol) and TMS-Cl (0.21mL, 1.626mmol) in dry THF (6mL) was added dropwise a solution of 6 α -ethyl-4 β -fluoro-7 α -hydroxy-3-oxo-5 β -cholan-24-oic acid methyl ester (product of example 1C; 170mg, 0.325mmol) in dry THF (2.5mL) at-78 deg.C over a period of 10 min. After addition, the reaction was gradually warmed to RT and stirred for 20 h. Upon completion, the reaction mixture was cooled to 0 ℃ by dropwise addition of saturated NaHCO3(5mL) quench with H2Dilution with O (5 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X5 mL). With NaHCO3(5mL)、H2The combined organic phases were washed with O (5mL) and brine (5 mL). The organic phases were combined and MgSO4Dried, filtered and concentrated in vacuo to give a yellow oil. The resulting syrup was used in the next reaction without further purification.

19F NMR(1H non-uncoupled, 376 MHz): delta-169.9 (1F, s);

to a stirred solution of methyl-3-trimethylsilyl-6 α -ethyl-4 β -fluoro-7 α -hydroxy-5 β -cholan-2-en-24-oate (putatively 0.19g, 0.325mmol) in dry MeCN (10mL) was added stepwise(0.17g, 0.488mmol) and the reaction was stirred at RT for 16 h. Upon completion, with EtOAc (5mL) and saturated NaHCO3The reaction was diluted (3 mL). The organic phase was removed and the aqueous phase was back-extracted with EtOAc (3X5 mL). The organic phases were combined and MgSO4Drying, filtering, and vacuum concentrating to obtainTo a yellow oil.

The process described in examples 2D and 2E above can be carried out on the product of step a to give methyl 6 α -ethyl-2 α/β, 4 β -difluoro- (3 α, 7 α) -dihydroxy-5 β -cholan-24-oate.

The compound can be converted to the equivalent sulfonylurea or sulfonamide analog using the general procedures 1-4 as described above in examples 1-3.

Synthesis of 2 beta-fluorocompounds

The 2 α -fluoro and 4 α -fluoro derivatives of obeticholic acid were prepared as follows.

Example 6-2 beta-fluoro-3 alpha, 7 alpha-dihydroxy-6 alpha-ethyl-5 beta-cholanic acid with sulfonylurea and sulfonamide side chains Synthesis of analogs

A. Methyl-3, 7-dioxo-6 alpha-ethyl-5 beta-cholan-24-oic acid ester

To a solution of (6 α, 5 β, 7 α) -6-ethyl-7-hydroxy-3, 7-dioxo-cholan-24-oic acid (36.0g, 87.7mmol, 1.0 eq) prepared as described in WO 2016/079520 in methanol (800mL) was added p-toluenesulfonic acid (1.67g, 8.78mmol,. about.0.1 eq) at RT and sonicated at 30 ℃ for 4 hours. Completion of the reaction was confirmed by TLC and the reaction mixture was concentrated in vacuo. The residue was dissolved in chloroform (400mL) and saturated NaHCO was used3The solution (400mL) and brine (400mL) were washed with Na2SO4Drying, filtration and concentration in vacuo gave 37.6g of crude material as a white solid which was used without further purification (87.3mmol, 99%).

1H NMR(400MHz,CDCl3):δ3.66(3H,s),2.74(1H,m),2.47(1H,t,J=11.3Hz),2.35(1H,ddd,J=15.4,10.0,5.3Hz),2.26-2.14(6H,m),2.10-1.77(6H,m),1.74-1.35(7H,m),1.33(3H,s),1.31-1.26(1H,m),1.21-0.96(4H,m),0.93(3H,d,J=6.5Hz),0.80(3H,t,J=7.4Hz),0.69(3H,s)ppm。

LRMS(ESI+)m/z:448.3[M+NH4]+,100%。

B. Methyl-3 beta-hydroxy-6 alpha-ethyl-7-oxo-5 beta-cholane-24-oic acid ester and methyl-3 alpha-hydroxy-6 alpha-ethyl-7-oxo-5 beta-cholane-24-oic acid ester

To a solution of step A of dry methyl-3, 7-dioxo-6 α -ethyl-5 β -cholan-24-oic acid ester (10.0g, 23.2mmol, 1.0 eq) in tetrahydrofuran (340mL) was added dropwise L-selectride (35.0mL, 34.8mmol,. about.2.5 eq) at-78 ℃ for 15 minutes under an argon atmosphere. After 10 minutes, the reaction mixture received a solution of hydrogen peroxide (40mL, 30% v/v) and 2M sodium hydroxide (40mL) in water (400mL) at 0 ℃. After an additional 10 minutes, the reaction mixture received 2M hydrochloric acid (130mL) at RT. The aqueous phase was separated, extracted with ethyl acetate (2 × 250mL), the combined organic fractions were washed with water (500mL), and Na was used2SO4Drying, filtration and concentration in vacuo gave 11.0g of crude material as a colorless oil. Purification by flash chromatography (Biotage SNAP KP-Sil 100g column) using PE 40-60/acetone (90/10-80/20) as eluent gave the inseparable 65% of the compounds methyl-3 β -hydroxy-6 α -ethyl-7-oxo-5 β -cholan-24-oic acid ester and methyl-3 α -hydroxy-6 α -ethyl-7-oxo-5 β -cholan-24-oic acid ester: 35 as a white residue (7.83g, 18.1mmol, 78%). For this mixture, the two compounds are not separated, and the same mixture of olefins is produced in the next step.

A compound A:1H NMR(400MHz,CDCl3):δ4.05(1H,t,J=2.5Hz),3.66(3H,s),2.77-2.73(1H,m),2.41-2.31(2H,m),2.26-2.14(3H,m),2.00-1.88(2H,m),1.84-1.58(6H,m),1.55-1.29(10H,m),1.25(3H,s),1.15-1.07(4H,m),0.92(3H,d,J=6.5Hz),0.81(3H,t,J=7.4Hz),0.66(3H,s)。

LRMS(ESI+)m/z:450.3[M+NH4]+,100%。

compound B:1H NMR(400MHz,CDCl3):δ3.66(3H,s),3.57-3.48(1H,m),2.76-2.67(1H,m),2.41-2.32(2H,m),2.26-2.14(3H,m),2.00-1.88(2H,m),1.84-1.59(6H,m),1.55-1.29(10H,m),1.22(3H,s),1.17-1.07(4H,m),0.92(3H,d,J=6.4Hz),0.80(3H,t,J=7.4Hz),0.65(3H,s)ppm。

LRMS(ESI+)m/z:450.4[M+NH4]+,100%。

C. methyl-6 alpha-ethyl-7 oxo-5 beta-chole-2-en-24-oic ester and methyl-6 alpha-ethyl-7-oxo-5 beta-chole-3-en-24-oic ester

To a solution of methyl-3 β -hydroxy-6 α -ethyl-7-oxo-5 β -cholan-24-oate and methyl-3 α -hydroxy-6 α -ethyl-7-oxo-5 β -cholan-24-oate (6.31g, 14.6mmol, 1.0 equiv) in dichloromethane (120mL) of step B was added dimethylaminopyridine (3.56g, 29.2mmol,. about.2.0 equiv) at RT. The reaction mixture was cooled to 0 ℃ and received dropwise triflic anhydride (2.57mL, 15.3mmol,. about.1.05 eq.) over 5 minutes. After 2 hours, warm to 12 ℃, confirm completion of the reaction by TLC and quench the reaction mixture with 2M hydrochloric acid (100 mL). The aqueous phase was separated, extracted with dichloromethane (3X 100mL), the combined organic fractions were washed with brine (200mL), Na2SO4Drying, filtration and concentration in vacuo gave 7.56g of crude material as an orange oil. Purification by flash chromatography (Biotage SNAP KP-Sil 100g column) using PE 40-60/acetone (90/10) as eluent gave methyl-6 α -ethyl-7 oxo-5 β -chol-2-en-24-oate and methyl-6 α -ethyl-7-oxo-5 β -chol-3-en-24-oate inseparable 80: 20 as a colorless oil (2.70g, 6.51mmol, 45%).

1H NMR(400MHz,CDCl3):δ5.63-5.40(2H,m),3.66(3H,s,),2.74(1H,dd,J=12.0,6.6Hz),2.34(2H,tt,J=10.3,5.1Hz),2.27-1.29(19H,m),1.27(2H,s),1.26(1H,s),1.18-0.94(3H,m),0.91(3H,d,J=6.5Hz),0.83(3H,t,J=7.5Hz),0.664(1H,s),0.657(2H,s)ppm。

LRMS(ESI+)m/z:432.20[M+NH4]+,100%。

D. Methyl-2 beta, 3 beta-epoxy-6 alpha-ethyl-7-oxo-5 beta-cholane-24-carboxylate and methyl-3 beta, 4 beta-epoxy-6 alpha-ethyl-7-oxo-5 beta-cholane-24-carboxylate

To a solution of methyl-6 α -ethyl-7 oxo-5 β -chol-2-en-24-oate and methyl-6 α -ethyl-7-oxo-5 β -chol-3-en-24-oate (5.00g, 12.1mmol,. about.1.0 equiv.) of step C in dichloromethane (100mL) at RT to 80: 20 ratio was added meta-perchlorobenzoic acid (3.12g, 18.1mmol,. about.1.5 equiv.). After 3 hours at RT, completion of the reaction was confirmed by TLC, saturated Na2S2O3The reaction mixture was quenched with a solution (150 mL). After 10min stirring, the aqueous phase was separated, extracted with dichloromethane (3X 100mL), Na2SO4The combined organic fractions were dried, filtered and concentrated in vacuo to give 5.28g of crude material as a pale yellow product. Purification by flash chromatography (Biotage SNAP KP-Sil 100g column) using PE 40-60/acetone (95/5-90/10) as eluent gave a mixture of methyl-2 β,3 β -epoxy-6 β 0-ethyl-7-oxo-5 β 1-cholan-24-oate and methyl-3 β 2, 4 β 3-epoxy-6 β 4-ethyl.7-oxo-5 β 5-cholan-24-oate inseparable as a colorless oil (4.94g, 11.5mmol, 95%). Further purification by flash chromatography (Biotage SNAP KP-Sil 340g column) using PE 40-60/acetone (95/5-90/10) as eluent gave the compound methyl-2 β,3 β -epoxy-6 α -ethyl-7-oxo-5 β -cholane-24-oate as a colorless oil (3.71g, 8.62mmol, 72%) and the compound methyl-3 β, 4 β -epoxy-6 α -ethyl-7-oxo-5 β -cholane-24-oate as a colorless oil (1.18g, 2.74mmol, 23%).

A compound A:1H NMR(400MHz,CDCl3):δ3.66(3H,s),3.13(1H,t,J=2.6Hz),3.01(1H,dd,J=5.5,4.2Hz),2.67(1H,dd,J=11.5,6.6Hz),2.35(1H,ddd,J=15.4,10.2,5.1Hz),2.30-2.18(3H,m),2.01-1.65(7H,m),1.55-1.20(10H,m),1.17(3H,s),1.15-0.95(3H,m),0.92(3H,d,J=6.4Hz),0.81(3H,t,J=7.4Hz),0.65(3H,s)ppm。

LRMS(ESI+)m/z:448.30[M+NH4]+,100%。

compound B:1H NMR(400MHz,CDCl3):δ3.66(3H,s),3.10-3.09(1H,m),2.79-2.74(2H,m),2.41-2.31(2H,m),2.26-2.04(3H,m),2.00-1.88(4H,m),1.84-1.58(2H,m),1.53-1.20(11H,m),1.17(3H,s),1.14-0.94(2H,m),0.91(3H,d,J=6.3Hz),0.90(3H,t,J=7.5Hz),0.66(3H,s)ppm。

LRMS(ESI+)m/z:448.26[M+NH4]+,100%。

E. methyl-2 alpha-fluoro-3 alpha-hydroxy-6 alpha-ethyl-7-oxo-5 beta-cholan-24-oic acid ester

To a dry solution of methyl-2 β,3 β -epoxy-6 α -ethyl-7-oxo-5 β -cholan-24-oate (3.33g, 7.73mmol, 1.0 equiv.) of step D in dichloromethane (100mL) was added hf pyridine (70%) complex (100mL, 3.86mol,. about.500 equiv.) at 0 ℃ under an argon atmosphere by pouring a freshly opened 100mL vial into a cooled reaction flask through a glass funnel under a steady argon gas flow. After addition of reagents, the vial and funnel were rinsed with dichloromethane (20 mL). After 3 hours at 0 deg.C, the reaction mixture was diluted with dichloromethane (200mL) by slow addition of saturated NaHCO3The solution (500mL) was quenched and stirred at RT for 1 hour while receiving 5.0g of NaHCO3This was carried out in 100mg portions. The aqueous phase was then separated, extracted with dichloromethane (3 × 250mL), and Na2SO4The combined organic fractions were dried, filtered and concentrated in vacuo to give 3.61g of crude material as a colorless oil. By passingFlash chromatography purification (Biotage SNAP KP-Sil 100g column) using PE 40-60/acetone (95/5-90/150 as eluent gave the compound methyl-2 α -fluoro-3 α -hydroxy-6 α -ethyl-7-oxo-5 β -cholane-24-oate as a white residue (2.39g, 5.30mmol, 69%).

1H NMR(400MHz,CDCl3):δ4.53(1H,dq,J=47.0,2.3Hz),4.01(1H,dq,J=6.6,2.9Hz),3.66(3H,s),2.75(1H,dd,J=13.3,5.8Hz),2.38-2.30(2H,m),2.25-2.11(4H,m),2.01(1H,dd,J=12.0,3.8Hz),1.93-1.36(14H,m),1.25(3H,d,J=4.0Hz),1.21-1.10(3H,m),0.91(3H,d,J=6.5Hz),0.82(3H,t,J=7.4Hz),0.65(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-184.3(1F,tt,J=50.3,8.7Hz)ppm。

LRMS(ESI+)m/z:468.28[M+NH4]+,100%。

F. Methyl-2 alpha-fluoro-3, 7-dioxo-6 alpha-ethyl-5 beta-cholan-24-oic acid ester

To a solution of methyl-2 α -fluoro-3 α -hydroxy-6 α -ethyl-7-oxo-5 β -cholan-24-oate (1.00g, 2.26mmol, 1.0 equiv.) from step E in dichloromethane (20mL) at RT was added dess-Martin reagent (1.92g, 4.52mmol,. about.2.0 equiv.) and H2O (0.25 mL). After 3 hours at RT, completion of the reaction was confirmed by TLC, saturated NaHCO3The reaction mixture was quenched with solution (25mL), filtered with celite, and washed with dichloromethane (90 mL). The aqueous phase was then separated, extracted with dichloromethane (2X50 mL), saturated Na2S2O3The combined organic layers were washed with Na solution (150mL)2SO4Drying, filtration and concentration in vacuo gave 1.21g of crude material as a pale yellow oil. Purification by flash chromatography (Biotage SNAP KP-Sil 25g column) using PE 40-60/acetone (95/5-90/10) as eluent gave the compound methyl-2 α -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholan-24-oic acid ester as a white residue (568mg, 1.27mmol, 56%).

1H NMR(400MHz,CDCl3):δ4.69(1H,ddd,J=50.7,4.8,3.4Hz),3.68(3H,s),2.75(1H,ddd,J=7.7,5.0,4.3Hz),2.51-1.94(12H,m),1.83-1.40(9H,m),1.37(3H,s),1.34-1.08(5H,m),0.94(3H,d,J=6.5Hz),0.84(3H,t,J=7.4Hz),0.70(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.2(1F,ddd,J=51.6,42.1,12.1Hz)ppm。

LRMS(ESI+)m/z:466.55[M+NH4]+,100%。

G.2 alpha-fluoro-3, 7-dioxo-6 alpha-ethyl-5 beta-cholanic acid and 2 beta-fluoro-3, 7-dioxo-6 alpha-ethyl-5 beta-cholanic acid

To a solution of methyl-2 α -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholan-24-oate (product of step F, 878mg, 1.95mmol, 1.0 eq) in methanol (20mL) was added sodium hydroxide (1.0g) at RT. After 19 h at RT, completion of the reaction was confirmed by TLC, the reaction mixture was acidified to pH 4.0 and concentrated in vacuo. The residue was dissolved in ethyl acetate (50mL), washed with 1M hydrochloric acid (50mL), and washed with Na2SO4Drying, filtration and concentration in vacuo gave 968mg of crude material as a colorless oil. Purification by flash chromatography (Biotage SNAP KP-Sil 25g column) using dichloromethane/methanol (98/2-90/10) as eluent gave 2 α -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholanic acid and 2 β -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholanic acid inseparable 40: 60% as a white residue (772mg, 1.77mmol, 91%).

2 α -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholanic acid:1h NMR-characteristic peaks (400MHz, CDCl)3):δ4.68(1H,ddd,J=50.7,5.1,3.6Hz),2.75(1H,ddd,J=7.7,5.0,4.3Hz),1.36(3H,s),0.94(3H,d,J=6.5Hz),0.82(3H,t,J=7.3Hz),0.69(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-188.2(1F,ddd,J=51.6,42.1,12.1Hz)ppm。

LRMS(ESI+)m/z:452.51[M+NH4]+,100%。

2 β -3, 7-dioxo-6 α -ethyl-5 β -cholanic acid:1h NMR-characteristic peaks (400MHz, CDCl)3):δ4.90(0.6H,ddd,J=48.7,13.2,6.1Hz),2.75(1H,ddd,J=7.7,5.0,4.3Hz),1.39(2H,s),0.95(2H,d,J=6.5Hz),0.82(3H,t,J=7.3Hz),0.70(2H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-195.1(1F,ddd,J=48.6,10.4,5.2Hz)ppm。

LRMS(ESI+)m/z:452.51[M+NH4]+,100%。

H. Methyl-2 beta-fluoro-3, 7-dioxo-6 alpha-ethyl-5 beta-cholan-24-oic acid ester

Cesium carbonate (840mg, 2.58mmol, 1.5 equiv.) is added at RT to a 40: 60 ratio solution of 2 α -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholanic acid and 2 β -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholanic acid (750mg, 1.72mmol, 1.0 equiv.) in dimethylformamide (17mL) of step G. After RT 20min, iodomethane (0.54mL, 8.59mmol,. about.5.0 equiv.) was added dropwise. After 19 hours at RT, completion of the reaction was confirmed by TLC and the reaction mixture was concentrated in vacuo. The residue was dissolved in ethyl acetate (25mL) and H2O (20 mL). The aqueous layer was separated, extracted with ethyl acetate (3 × 25mL), the combined organic layers were washed with brine (100mL), and Na2SO4Drying, filtration and concentration in vacuo gave 917mg of crude material as a pale yellow oil. Purification by flash chromatography (Biotage SNA)P Ultra KP-Sil 25g column) using PE 40-60/acetone (95/5-90/10) as eluent gave the compound methyl-2 β -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholan-24-oate as a white residue (416mg, 0.95mmol, 54%). The corresponding 2 α -fluoro derivative is not isolated.

1H NMR(400MHz,CDCl3):δ4.69(1H,ddd,J=48.8,13.3,6.4Hz),3.67(3H,s),2.75(1H,dd,J=13.1,5.0Hz),2.54-2.44(2H,m),2.40-2.32(2H,m),2.29-2.13(3H,m),2.09(1H,dt,J=13.0,3.3Hz),2.01-1.91(2H,m),1.85-1.65(5H,m),1.54-1.42(2H,m),1.39(3H,s),1.38-1.05(7H,m),0.94(3H,d,J=6.5Hz),0.82(3H,t,J=7.4Hz),0.70(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-195.1(1F,ddt,J=48.6,10.4,5.2Hz)ppm。

LRMS(ESI+)m/z:466.59[M+NH4]+,100%。

I. Methyl-2 beta-fluoro-3 beta, 7 alpha-dihydroxy-6 alpha-ethyl-5 beta-cholane-24-oic acid ester and methyl-2 beta-fluoro-3 alpha, 7 alpha-dihydroxy-6 alpha-ethyl-5 beta-cholane-24-oic acid ester

Sodium borohydride (164mg, 4.44mmol, 5.0 equiv.) is added to a dry solution of methyl-2 β -fluoro-3, 7-dioxo-6 α -ethyl-5 β -cholan-24-oate (390mg, 0.87mmol, 1.0 equiv.) from step H at RT in methanol (20mL) under an argon atmosphere. After 1 hour at RT, the reaction mixture was concentrated in vacuo. The residue was dissolved in dichloromethane (20mL) and H2O (20mL), the aqueous phase was separated and extracted with dichloromethane (3 × 20 mL). With Na2SO4The combined organic fractions were dried, filtered, and concentrated in vacuo to give 446mg of crude material as a pale yellow oil. Purification by flash chromatography (Biotage SNAP Ultra KP-Sil 25g column) using PE 40-60/acetone (95/5-90/10) as eluent gave the compound methyl-2 β -fluoro-3 β, 7 α -dihydroxy-6 α -ethyl-5 β -cholan-24-oate as colorless oil (161mg, 0.36mmol, 41%) and methyl-2 β -fluoro-3 α, 7 α -dihydroxy-6 α -ethyl-5 β -cholan-24-oate as colorless oil (146mg, 0.32mmol, 37%).

Methyl-2 β -fluoro-3 β, 7 α -dihydroxy-6 α -ethyl-5 β -cholan-24-oic acid ester:1H NMR(400MHz,CDCl3):δ4.63(1H,dddd,J=47.4,12.5,4.4,3.1Hz,),4.15(1H,q,3.6Hz),3.71(1H,s),3.67(3H,s),2.36(1H,ddd,J=15.5,10.2,5.3Hz),2.33(1H,ddd,J=16.0,9.7,6.6Hz),2.12(1H,td,J=13.2,2.2Hz),2.00(1H,dt,J=12.5,3.1Hz),1.94-1.78(5H,m),1.77-1.54(6H,m),1.52-1.25(8H,m),1.20-1.12(4H,m),1.00(3H,s),0.93(3H,d,J=6.8Hz),0.92(3H,t,J=7.1Hz),0.67(3H,s)ppm。

19F NMR(1h non-decoupled, 376MHz, CDCl3):δ-187.1(1F,dquin,J=46.9,7.8Hz)ppm。

LRMS(ESI+)m/z:470.64[M+NH4]+,100%。

Methyl-2 β -fluoro-3 α, 7 α -dihydroxy-6 α -ethyl-5 β -cholan-24-oic acid ester:1H NMR(400MHz,CDCl3):δ4.42(1H,dddd,J=52.7,12.5,8.7,4.5Hz),3.70(1H,s),3.67(3H,s),3.52(1H,ddt,13.8,12.5,6.0Hz),2.36(1H,ddd,J=15.4,10.2,5.3Hz),2.27-2.17(3H,m),2.09-1.97(2H,m),1.95-1.86(2H,m),1.84-1.76(1H,m),1.66-1.29(14H,m),1.22-1.13(4H,m),0.98(3H,s),0.93(3H,d,J=6.5Hz),0.92(3H,t,J=7.2Hz),0.67(3H,s)ppm。

19F NMR(1h non-decoupled, 376MHz, CDCl3):δ-188.2(1F,ddd,J=51.6,42.1,12.1Hz)ppm。

LRMS(ESI+)m/z:470.64[M+NH4]+,100%。

J.2 beta-fluoro-3 alpha, 7 alpha-dihydroxy-6 alpha-ethyl-5 beta-cholanic acid

To a solution of methyl-2 β -fluoro-3 α, 7 α -dihydroxy-6 α -ethyl-5 β -cholan-24-oate (119mg, 0.26mmol, 1.0 equiv) of step I in 1, 4-dioxane (9.8mL) and water (3.6mL) was added concentrated (37%) hydrochloric acid (1.2mL, 9: 3: 1 ratio) at RT. After 1 hour of reflux, the reaction was confirmed to be complete by TLC, using saturated NaHCO3The reaction mixture was neutralized with solution (20 mL). The aqueous phase was extracted with ethyl acetate (3 × 15mL) and Na2SO4The combined organic fractions were dried, filtered, and concentrated in vacuo to give 141mg of crude material as a colorless oil. Purification by flash chromatography (Biotage SNAP Ultra KP-Sil 10g column) using dichloromethane/methanol (95/5-90/10) as eluent gave compound 2 β -fluoro-3 α, 7 α -dihydroxy-6 α -ethyl-5 β -cholanic acid as a white residue (92mg, 0.21mmol, 80%).

1H NMR(400MHz,CDCl3):δ4.42(1H,dddd,J=52.6,12.5,8.7,4.3Hz),3.69(1H,s),3.52(1H,tdd,12.0,8.8,5.4Hz),2.39(1H,ddd,J=15.5,10.3,5.1Hz),2.29-2.19(3H,m),2.01(2H,t,J=13.0Hz),1.93-1.77(3H,m),1.65-1.54(3H,m),1.53-1.26(12H,m),1.23-1.10(4H,m),0.97(3H,s),0.94(3H,d,J=6.5Hz),0.91(3H,t,J=7.1Hz),0.66(3H,s)ppm。

19F NMR(1H non-decoupled, 376MHz, CDCl3):δ-186.8(1F,ddq,J=52.9,13.0,7.5Hz,)ppm。

LRMS(ESI+)m/z:456.60[M+NH4]+,100%。

The compound can be converted to the equivalent sulfonylurea or sulfonamide analog using the general procedures 1-4 as described above in examples 1-3.

Biological examples

For the following biological examples 8 and 9, all work has been done by professor Kim Watson and professor Dannielle Kydd-Sinclair of University of Reading, UK and the data is provided friendly.

Example 7 determination of EC50 and efficacy at FXR receptor

Agonist activity of the compounds of the invention at the FXR receptor was determined. Table 1 shows the EC of the example compounds of the invention compared to the values of the comparative example compound obeticholic acid and the known FXR agonist GW4064 having the following structure50Values and efficacy values:

efficacy was defined as the highest point on the dose-response curve and the efficacy value for GW4064 in table 1 was designated as 100%.

Mixing EC in Table 150EC against GW4064 designated 25nM50And (6) calibrating.

Obeticholic acid may be prepared as described in WO 02/072598 or our applications WO 2016/079518, WO 2016/079518, WO 2016/079519 and WO 2016/079520.

FXR EC50Efficacy protocol

Dose response assays were performed as described in the handbook of Human Farnesoid X Receptor (NR1H4, FXR) Reporter gene detection System technology (Indigo Biosciences Human Farnesoid X Receptor (NR1H4, FXR) Reporter Assay System, Technical Manual (7.1b edition), www.indigobiosciences.com).

FXR receptor cells consisting of the FXR-responsive promoter gene functionally linked to the luciferase gene were thawed and seeded into 96-well plates, and different concentrations (10-0.05 μ M) of test compound were immediately administered to these cells according to the manufacturer's protocol. After incubation for 24h in the presence of test compound or solvent (DMSO), the Cell viability of these treated/untreated reporter cells was measured using a fluorescence-based Living Cell Multiplex (LCM) Assay to eliminate false negative results (Indigo Biosciences Live Cell Multiplex Assay, Technical Manual (version 3.1), www.indigobiosciences.com). Fluorescence of live cells was measured using a plate reader in combination with a filter of [485nmEx |535nmEm ]. Thereafter, the induction of luciferase activity, which is a measure of agonist activity, was quantified using a luminometer (TECAN) according to the manufacturer's protocol.

Positive control tests were carried out in each case, in which EC of GW4064 was measured50The value was assigned as 25nM and the efficacy of GW4064 was assigned as 100% efficacy value. Efficacy and EC of each test compound50Efficacy and EC with GW406450A comparison is made.

The results are shown in table 1.

TABLE 1

The compounds of the invention all have FXR agonist activity. All compounds except compound 11 had significantly improved EC compared to obeticholic acid50The value is obtained. Furthermore, the efficacy values of all compounds of the invention are at least as good as those of GW4064 and in most cases better than GW 4064.

Comparison of the results for Compound 2 and its unfluorinated analog versus Compound A shows that fluorination improves EC50And efficacy. Comparison of the results for the aromatic sulfonamide compounds 14-20 with the carbocyclic sulfonamide comparative compound B and the methylsulfonamide comparative compound C indicates the importance of the aromatic substituent on the side chain.

50Example 8 determination of EC and efficacy at the TGR5 receptor

Provided using DiscoverX in agonist modecAMP assay to monitor the activity of compounds 2 and 14, TGR5 (control) and taurine and glycine conjugates of obeticholic acid at TGR5 receptor to monitor activation of TGR5(GPBAR1) receptor through Gi and Gs secondary messenger signaling. Data were normalized to the maximum and minimum response observed in the presence of control ligand (TGR5) and vehicle.

And (3) experimental design: GPCR cAMP modulation

Cell manipulation

1. cAMP Hunter cell line was expanded from refrigerator stocks according to standard methods.

2. Cells were seeded in white-walled 384-well microplates in a total volume of 20 μ Ι _ and incubated at 37 ℃ for an appropriate time prior to testing.

3. Using DiscoverXThe cAMP XS + assay measures cAMP modulation.

Gs agonist forms

1. For agonist assays, cells are incubated with the sample to induce a response.

2. The medium was aspirated from the cells and incubated with 15. mu.L of 2: 1HBSS/10mM Hepes: cAMP XS + Ab reagent.

3. Intermediate dilutions of the sample stock were performed to generate 4X samples in assay buffer.

4. Add 5 μ L of 4x sample to the cells and incubate at 37 ℃ or room temperature for 30 or 60 minutes. Vehicle concentration was 1%.

Signal detection

1. After incubation with the appropriate compounds, assay signals were generated by incubation with 20 μ LcAMP XS + ED/CL lysis mix for 1 hour, followed by incubation with 20 μ LcAMP XS + EA reagent for 3 hours at room temperature.

2. After the signal has occurred, a Perkinelmer Envision for chemiluminescent signal detection is usedTMThe instrument reads the microplate.

Data analysis

1. Compound activity was analyzed using CBIS data analysis kit (chemlinnovation, CA).

2. For the Gs agonist mode assay, the percent activity is calculated using the following formula, where RLU is an abbreviation for relative light units:

% activity 100% x ((average RLU of test samples-average RLU of vehicle control)/(average RLU of maximum control-average RLU of vehicle control))

5. For Gi agonist mode assays, the percent activity is calculated using the following formula:

% activity 100% x ((1- (mean RLU of test sample-mean RLU of maximum control))/(mean RLU of vehicle control-mean RLU of maximum control))

Results

The results are shown in table 2.

TABLE 2

Compound (I) EC50(μM) Maximum response
TGR5 (control) 0.445 101.26
Compound 2 >100 0
Compound 14 >100 0
Obeticholic acidTaurine conjugates 0.979 97.70
Obeticholic acid glycine conjugates 1.904 110.9

The results indicate that, unlike obeticholic acid conjugates, neither compound 2 nor compound 14 have agonist activity at TGR5 receptor. Thus, these compounds are selective FXR agonists.

Example 9 quantitative analysis of ligand-induced Gene expression

The effect of compound 2 and obeticholic acid on the expression of a number of FXR target genes was tested. This example describes cell-based assays and gene expression analysis to observe functional activation of FXR at the cellular level by the compounds of the invention and obeticholic acid.

To assess specific changes in gene expression in response to test compounds, precise quantification and analysis was performed by quantitative real-time pcr (qpcr). Mammalian tissue culture experiments involved hepatocellular carcinoma (Huh7) cells at 1 × 106The concentration of cells/well was seeded in 6-well plates and incubated at 37 ℃ for 24 hours to allow attachment. Exposing the cells to the EC in the cells50Or EC90The corresponding compound or vehicle (DMSO) below was for 24 hours.

Compound 2 and OCA were also tested in the human hepatocellular carcinoma HepG2 cell line. HepG2 cells were also conjugated with inclusion of EC50Or EC90Media of OCA or compound 2 at concentration were incubated together for 24 hours.

Compound 14 was also tested for Huh7 cells and HepG2 cells in the same manner as compound 2.

Isolation of Total RNA from cultured cells

Using RNAqueousTMTotal RNA isolation kit (Ambion)) Total RNA was extracted and all reagents were provided in the kit or prepared according to the manufacturer's instructions. For the cultured cells, the medium was removed, and the cells were washed with 1 × PBS to remove cell debris and residual medium. Total RNA was extracted from fresh cells. For 1X 106Cells were harvested by adding 350. mu.l lysis buffer directly to the wells and scraping with a pipette tip. Lysed cells were combined with an equal volume of 64% ethanol and mixed thoroughly by pipetting. The ethanol lysate mixture was transferred to the column and spun at 12,000XG for 1 minute and the flow-through was discarded. The membrane was washed by adding wash buffer 1 to the column and centrifuging at 12,000XG for 1 minute, then discarding the flow-through and repeating this step twice with wash buffer 2. Additionally included was spinning with an empty column to completely dry the ethanol membrane. Finally, total RNA was eluted in 2 sequential aliquots of 50. mu.l pre-heated elution buffer (nuclease free water with traces of EDTA).

RNA quantity, purity and integrity analysis

RNA concentration was quantified by measuring absorbance at 260nm using a Nanodrop Lite spectrophotometer (Thermo Scientific). Purity of RNA by analysis A260∶280Wherein values between 1.8 and 2.1 are considered free of protein contamination and acceptable for downstream applications. The integrity of the RNA was determined by running the sample on a denaturing formaldehyde agarose gel. By adding 1 × SYBRTMSafe DNA dye (Invitrogen) A1% agarose (Sigma Aldrich), 1X MOPS (Sigma Aldrich), 6.6% formaldehyde (Fisher Scientific) gel was made to visualize nucleic acids. Before loading, an equal volume of formaldehyde staining dye (Ambion) was added to 1 μ g of RNA, and the sample was heated at 70 ℃ for 10 minutes, then immediately cooled rapidly on ice for 2-3 minutes. The gel was run at 90V for 1 hour 30 minutes and developed under UV light using the NuGenius gel doe system (Syngene). The bands of 28S and 18S rRNA were carefully examined, and clear, intense bands were observed at approximately 5kb and 1.9kb, respectively, where the intensity of the upper band of 28S was expected to be twice that of the lower band of intact RNA 18S, respectively. Smears below the 18S rRNA band were considered to represent degraded RNA, while smears above the 28S rRNA band were considered to represent degraded RNAThe patch and/or strip then indicates DNA contamination. Immediately after analysis, the RNA was used for reverse transcription.

Reverse transcription

Using iScript for RT-qPCRTMReverse transcription was performed using the Advanced cDNA Synthesis kit (Biorad). Mu.l of 5 XScript was added to 1. mu.g of DNase-treated RNATMAdvanced reaction mixture and 1. mu.l of iScriptTMAdvanced reverse transcriptase. Nuclease-free water was added to a final volume of 20. mu.l and the reaction was incubated at 46 ℃ for 20 minutes and then inactivated at 95 ℃ for 1 minute. The newly synthesized cDNA was diluted 10-fold in TE buffer (10mM Tris pH 8, 1mM EDTA), aliquoted and stored at-20 ℃ until used in the qPCR experiment.

Qualitative real-time PCR analysis

Reference genes were selected based on data from the existing literature. The selected target genes are shown in table 3.

TABLE 3 selected target genes for qualitative real-time PCR analysis

Optimization of qPCR primers

For the above target genesGreen pre-designed primers (selected according to best grade) were purchased from Sigma Life Science. Nuclease-free water was added to the lyophilized primers at a stock concentration of 100. mu.M. The primers were diluted with nuclease-free water to a working concentration of 10. mu.M. To test the efficiency, reproducibility and dynamic range of the assay, ten-fold serial dilutions were performed; consisting of 5 concentrations of cDNA (as described above) produced from human reference RNA (Agilent). After qPCR was performed on these samples, a standard curve was constructed by threshold cycle (Ct) values (y-axis) versus log cDNA concentration (x-axis). The amplification efficiency (E) of the primer in one cycle of the exponential phase is given by the equation E-10(-1/slope)1(Pfaff1, 2001). The accuracy of these qPCR reactions was determined from R of the standard curve2Values are determined, where values > 0.98 are suitable. The specificity of each primer was determined by melting curve analysis, which was performed at the end of each experiment, wherein the generation of one peak at one melting temperature indicates the amplification of only one product and, therefore, is a highly specific primer. The amplification products were confirmed by agarose gel electrophoresis (2% agarose, 1 × TAE, run at 100V for 30 minutes) to check whether the amplicon size was as expected, and only one product was observed.

Quantitative PCR

Ready-to-use reaction standard mixture iTaqTM UniversalGreen Supermix (Biorad) was used for all qPCR reactions. A typical reaction for each gene contained 5. mu.l of 2 XiTaqTM UniversalGreen Supermix, 500nm forward and 500nm reverse primers, about 15ng cDNA and nuclease-free water to a final volume of 10. mu.l. Each target gene, reference gene and no template control were run in triplicate on Optical MicroAmp 96-well plates (Applied Biosystems). The plates were sealed with an optical adhesive sealer (Applied biosystems) and briefly placed on a plate shaker to mix the ingredients and centrifuged. An Applied Biosystem Step One Plus real-time PCR system was used to perform the reaction under the following cycling conditions; the initial denaturation step at 95 ℃ lasted 15 minutes, 40 amplification cycles consisting of a denaturation step at 94 ℃ lasted 15 seconds, combined annealing and a1 minute extension step at 60 ℃, where a single fluorescence measurement was taken. Immediately after each run, melting curve analysis was performed by increasing the temperature from 60 ℃ to 95 ℃ in 0.3 ℃ increments and measuring fluorescence dissociation.

Data analysis

To quantify gene expression, baseline corrected Ct values were automatically determined by qPCR system software (Applied Biosystems). Determination of relative Gene expression by Livak or Δ Δ Ct methodsA variation wherein the Ct values of the target genes for the vehicle treated/untreated sample (control) and the test compound treated sample (test sample) are calibrated to the Ct value of the reference gene. The Δ Ct values for the test samples were then calibrated to the Δ Ct values for the control samples. And finally, using equation 2-ΔAC tAnd calculating the expression rate. A total of 3 biological replicates were taken and all data are expressed as mean ± SE. Treatments were compared by one-way analysis of variance, followed by Tukey and Dunnet's post hoc tests.

Results

Upon treatment with compound 2, Huh7, the direct target genes nr0b2(SHP) and slc51a (OST α) involved in bile acid homeostasis and known to be positively regulated by FXR (Goodwin et al, 2000; Landrier et al, 2006) displayed significant increases. SHP expression levels increased by 1.5 (EC) after Compound 2 treatment50Concentration) -2.5 times (EC)90Concentration) (fig. 1). Ost alpha mRNA is also significantly up-regulated, in its EC50And EC90Treatment at concentration showed an 11-fold and 18-fold increase in expression, respectively (figure 2).

FXR activation leads to inhibition of CYP7a1 through SHP-mediated and FGF 19-mediated pathways. CYP7a1 was down-regulated with increasing concentration of compound 2 (figure 3).

As the counter compound 2 in its EC50In response to treatment at concentrations, HepG2 cells exhibited significant down-regulation of TGF β 1 expression levels (fig. 4).

SHP showed a modest increase, so Compound 14 was used in its EC90The expression level of the treatment at concentration was increased more than doubled (FIG. 5). Compound 14 induced a significant increase in FXR-target Ost α, with mRNA expression levels of the treated cells reaching 20-40 fold higher than vehicle-treated cells (fig. 6).

At EC50At concentrations, compound 14 significantly reduced CYP7a1 expression to more than half of the level observed for vehicle control cells (fig. 7).

Similar to compound 2, compound 14 induced a decrease in TGF β 1 expression with increasing compound concentration (figure 8).

The present inventors thanks to Dannielle Kydd-Sinclair of University of Reading, who provided a number of background sections and methods and data for the biological examples described above, and to David Evans, Simon Holland and Lawrence Tam of University of Southampton, who performed the synthesis of several example compounds.

Reference to the literature

Alemi, F. et al (2013) 'The TGR5 receiver media acids-induced bit and analytical analysis', The Journal of Clinical Investigation, 123(4), pp.1513-1530. doi: 10.1172/JCI64551. station.

Anthanarayanan, M. et al (2004) 'Ligand-dependent activation of the farnesoid X-receptor orientation of hormone H3 by CARM 1', Journal of Biological Chemistry, 279(52), pp.54348-54357. doi: 10.1074/jbc. M410021200.

Bellentani,S.(2017)‘The epidemiology of non-alcoholic fatty liver disease’,37(October 2016),pp.81-84.doi:10.1111/liv.13299.

Cave, M.C. et al (2016) 'Biochimica et Biophysica Acta Nuclear receptors and nonalcoholic facial skin disease 1', BBA-Gene Regulatory machinery, Elsevier B.V., 1859(9), pp.1083-1099. doi: 10.1016/j.bbagrm.2016.03.002.

Using Combinatorial Peptide Libraries:Discovery of Peptide Antagonists of Estrogen Receptorsαand β.Molecular and Cellular Biology,19(12),8226-8239.https://doi.org/10.1128/mcb.19.12.8226

Chiang,J.Y.L.(2013)‘Bile Acid Metabolism and Signalling’,

Comprehensive Physiology,3(3),pp.1191-1212.doi:10.1002/cphy.c120023.Bile.

Ciprini, S. et al (2010) 'FXR activation reactions in Steel resistance and Lipid abnormalities an d protects against Lipid boiler in Zucker (fa/fa) reactors', Journal of Lipid Research, 51, pp.771-784. doi: 10.1194/jlr. M001602.

Clerici et al, biology and Applied Pharmacology, 214, 199-208(2006) Copple, B.L.and Li, T. (2016)' Pharmacology of two acid receptors:

Evolution of bile acidsfrom simple detergents to complex signaling molecules’,PharmacologicaI Research.Elsevier Ltd,104,pp.9-21.doi:10.1016/j.phrs.2015.12.007.

cushman et al J.Med.chem. (1995), Cosalane analogues with enhanced loci as inhibitors of HIV-1protease and integrin ', 38(3), 443. 452. Cusi, Gastroenterology (2012),' Role of organism and lipotoxicity in the course of the differentiation of nociceptive stephanism: pathophysiology and clinical modalities', 142(4), 711, 725-

Deuschle, U.S. (2012) 'FXR Controls the turbine NDRG2 and FXR Agonists Reduce turbine Growth and Metastasis in an organic Mouse xenon Model', PLoS ONE, 7(10). Doi: 10.1371/journal.pone.0043044.

Dyson, j.k., anste, q.m., and McPherson, S. (2014)' Non-alcoholic facial skin disease: a practical approach to diagnosis and starting, Frontline gastroenterology, 5(3), pp.211-218. doi: 10.1136/flgastro-2013-.

Evans,R.M.and Mangelsdorf,D.J.(2014)‘Nuclear Receptors,RXR and the Big Bang’,Cell,157(1),pp.255-266.doi:10.1016/j.cell.2014.03.012.Nuclear.

Goodwin,B.,Jones,S.A.,Price,R.R.,Watson,M.A.,McKee,D.D.,Moore,L. B.,…KIiewer,S.A.(2000).A regulatory cascade of the nuclear receptors FXR,SHP-1,and LRH-1represses bile acid biosynthesis.Molecular Cell,6(3),517-526.

https://doi.org/10.1016/S1097-2765(00)00051-4.

Haas,J.T.,Francque,S.and Staels,B.(2016)‘Pathophysiology and Mechanisms of Nonalcoholic Fatty Liver Disease’,Annual Review 0f Physiology,78,pp.181-205.doi:10.1146/annurev-physiol-021115-105331.

Henao-mejia, J. et al (2012) 'Inflamamame-media regulations of NAFLD and ease.', Nature, 482(7384), pp.179-185. doi: 10.1038/nature10809. inflmamame-media.

Honorio et al 'Hologram QSAR Studies on Farnesoid X receptors Activators', Letters in Drug Design & Discovery, (2006), 3(4), 261-.

Inagaki, T. et al (2006) 'Regulation of antibiotic damage in the antibiotic by the nuclear acid receiver', Proc Natl Acad Sci U S A, 103(10), pp.3920-3925. doi: 10.1073/pnas.0509592103.

Jiang, y. et al (2013) 'Farnesoid X reeeptor inhibitors in mouse livers and previous depths of driver cancer.', Hepatology (Baltimore, Md.), (57), (3), pp.1098-106. doi: 10.1002/hep.26146.

Kast, H.R. et al (2001)' Farnesoid X-activated receptors inductor C-II transfer: a Molecular mechanism linking plasma molecules to biological acids', Molecular endicerology (Baltimore, Md.), 15(10), pp.1720-1728. doi: 10.1210/me.15.10.1720.

Kong, B. et al (2009) 'Farnesoid X Receptor discovery indexes in Low-sensitivity Lipoprotein Receptor-Knockout Mice Fed a High-Fat Diet', Journal of pharmaceutical and Experimental Therapeutics, 328(1), pp.116-122. doi: 10.1124/jpeg.108.144600.

Landrier, J-F et al (2006) 'The nuclear receptor for biological acids, FXR, transactivates human organic reagents transporter-alpha and-beta genes', am.J.Physiol-gateway.Liver Physiol.290, pp G476-G485.

Ma, k. et al (2006) 'Farnesoid X receiver is accession for normal glucose hometasis', Journal of Clinical Investigation, 116(4), p.1102. doi: 10.1172/JCI25604.1102.

Ma, Y, et al (2013) 'Synthetic FXR Agonist GW4064 previous Diet-Induced Pharmaceutical Processes and Insulin Resistance', Pharmaceutical Research, 30, pp.1447-1457. doi: 10.1007/s11095-013-0986-7.

Macchiarulo et al Journal of Chemical Information and Modeling, 48, 1792-

Maloney, p.r. et al (2000) 'Identification of a chemical tool for the alpha nuclear reactor FXR', Journal of medical Chemistry, 43(16), pp.2971-2974. doi: 10.1021/jm0002127.

Min, H.et al (2013) 'incorporated regenerative synthesis and dystraction of cholesterol Metabolism is associated with the selectivity of non-alcoholic fatty lift disease', Cell Metabolism, 15(5), pp.665-674. doi: 10.1016/j.cmet.2012.04.004.

Mouzaki, m. et al (2013) 'intellectual Microbiota in Patients With Nonalcoholic Fatty Liver Disease', Hepatology, 1, pp.120-127. doi: 10.1002/hep.26319.

Mudaliar, S. et al (2013) 'efficiency and Safety of the Farnesoid X Receptor Agonist Objective Acid in Patients With Type 2Diabetes and Nonalcoholic fat reagent', gastroenterology.Elsevier, Inc, 145(3), p.574-582.e1. doi: 10.1053/j.gastro.2013.05.042.

Neuschwander-Tetri,B.A.(2012)‘Farnesoid X receptor agonists:What they are and how they might be used in treating liver disease’,Current Gastroenterology Reports,14(1),pp.55-62.doi:10.1007/s11894-011-0232-6.

neuschwanderer-Tetri, b.a. et al (2015)' Farnesoid X nuclear receptor ligand for non-cyclic acid, non-cyclic stearic acid (FLINT): a multicentre, randomised, placebo-controlled trim prof.', Lancet, 385(9972), pp.956-965. doi: 10.1016/S0140-6736(14)61933-4 Farnesoid.

Pacan, T.and Sanyal, A.J. (2015) 'registration advances in understating/management of non-alcoholic stephanities', F1000 Prime Reports, 7(28), pp.1-8. doi: 10.12703/P7-28.

Paulekuhn et al (2007) 'Trends in Active Pharmaceutical Ingredient Selection based on Analysis of the Orange Book Database', J.Med.chem.50, 6665-

Pfafffl,M.W.(2001).A new mathematical model for relative quantification in real-time RT-PCR.Nucleic Acids Research,29(9),16-21.

Roda et al Gastroenterology (1995) 'Metabolism, pharmacologic, and activity of a new 6-fluoro analog of ursodeoxycholic acid in rates and hamsters', 108(4), 1204-.

Sato et al' Novel Power and Selective double Acid Derivatives as TGR5 Agonites: biological Screening, Structure-Activity Relationships, and Molecular Modeling students', J.Med.chem. (2008), 51(15), 4831-4849.

Sieven E. et al'1H,13C,19F NMR,and ESI mass spectral characterization of two geminal difluorosteroids′,Magnetic Resonance in Chemistry,(2008),46(4),392-397.

Uekawa et al (2004) 'Short-step Synthesis of chemodiol from StigmastateFol', biosci, Biotechnol, biochem., 68, 1332-

Vaquero, J.et al (2013) 'Differential activation of the human faulted X-receiver depends on the pattern of the expressed iso forms and the binary acid pool composition', Biochemical pharmacy, 86(7), pp.926-939. doi: 10.1016/j.bcp.2013.07.022.

Wang, Y. et al (2008) 'Farnesoid X receiver antagonists NF-kB in a fibrous infllamation response', Hepatology, 48(5), pp.1632-1643. doi: 10.1002/hep.22519.Farnesoid.

Watanabe, m., Houten, s.m., Wang, l., moshetta, a., magelsdorf, d.j., Heyman, r.a., Auwerx, j. (2004). Bile acids lower triglyceriders levels via a path accounting FXR, SHP, and SREBP-1 c.journal of Clinical investment, 113(10), 1408-1418. https: org/10.1172/JCI200421025Watanabe, M. et al (2011) 'Power double acid pool size with a synthetic Facade X Receiver (FXR) inductive and diabetes through reduced energy extension', Journal of Biological Chemistry 286(30), pp.26913-26920. doi: 10.1074/jbc.M111.248203.

Wuts, PGM and Greene, TW (2006) "Greene's Protective Groups in Organic Synthesis", 4 th edition, John Wiley & Sons, Inc., Hoboken, NJ, USA, Xiao, H. et al (2017) ' Synthesis and biological evaluation of a series of biological derivatives as R aggregations for FXeatment of NASH ', Med.chem.Lett.8, 1246-.

Yang, Z., Shen, W., and Sun, H. (2010) 'Effects of nuclear receiver FXR on the alignment of real metallic in substrates with non-alcoholic magnetic efficiency disease', Hepatology International, 4(4), pp.741-748. doi: 10.1007/s12072-010-9202-6.

Zhang, y. et al (2004) 'Peroxisome promoter-activated ligands trigyceride by activation of the nuclear receptor FXR', Genes & Development, 18, pp.157-169. doi: 10.1101/gad.1138104.

Zhang, y, et al (2006) 'Activation of the nuclear receptor FXR improves hyperglycemics and hyperlipidemia in diabetes mellitus', Proceedings of the National Academy of Sciences of the United States of America, 103(4), pp.1006-1011. doi: 10.1073/pnas.0506982103.

Zhang, y., Kast-woelbern, h.r. and Edwards, p.a. (2003) 'Natural Structural Variants of the Nuclear Receptor Farnesoid X Receptor Affect transformational Activation', Journal of Biological Chemistry 278(1), pp.104-110. doi: 10.1074/jbc. M209505200.

Detailed Description

In the present application, the techniqueThe word "C1-6"alkyl" means a straight or branched chain fully saturated hydrocarbon group having 1 to 6 carbon atoms. The term includes methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl. Other alkyl radicals, e.g. C1-4Alkyl radical, C1-3Alkyl or C1-2Alkyl groups are as defined above and contain varying numbers of carbon atoms.

The term "alkylene" refers to a straight or branched chain hydrocarbon chain that is fully saturated. Suitable alkylene groups are C1-4Alkylene radical, C1-3Alkylene or C1-2An alkylene group. Examples of alkylene groups include-CH2-、-CH2CH2-、-CH(CH3)-CH2-、-CH2CH(CH3)-、-CH2CH2CH2-and-CH2CH(CH2CH3)-。

The term "alkenylene" refers to a straight or branched hydrocarbon chain containing at least one carbon-carbon double bond. Suitable alkenylene radicals are C2-4Alkenylene or C2-3An alkenylene group. Examples of alkenylene include-CH ═ CH-, -CH ═ C (CH)3)-、-CH2CH=CH-、-CH=CHCH2-、-CH2CH2CH-and-CH2CH=C(CH3)-。

The terms "aryl" and "aromatic group" refer to cyclic groups that are aromatic, have 6 to 14 carbon atoms (unless otherwise specified, e.g., 6 to 10 ring carbon atoms) and contain up to 3 rings. If the aryl group contains more than one ring, not all rings need be aromatic. Examples include phenyl, naphthyl and anthracenyl and partially saturated systems such as tetrahydronaphthyl, indanyl and indenyl. A further example of an aryl group is 1, 2, 3, 4-tetrahydronaphthalene.

The terms "heteroaryl" and "heteroaromatic group" refer to cyclic groups that are aromatic and have 5 to 14 ring atoms (unless otherwise specified, e.g., 5 to 10 ring atoms), at least one of which is a heteroatom selected from N, O and S and contains up to 3 rings. If the heteroaryl group contains more than one ring, not all rings need be aromatic. Examples of heteroaryl groups include pyridine, pyrimidine, indole, benzofuran, benzimidazole, and indolylene. Additional examples of heteroaryl groups include quinoline and isoquinoline.

The term "halogen" refers to fluorine, chlorine, bromine or iodine, and the term "halo" refers to fluoro, chloro, bromo or iodo groups.

The term "C1-6Haloalkyl "means a straight or branched chain alkyl group as defined above having 1 to 6 carbon atoms and substituted, up to perhalogenated, with one or more halogen atoms. Examples include trifluoromethyl, chloroethyl and 1, 1-difluoroethyl. Other haloalkyl radicals, e.g. C1-5Haloalkyl, C1-4Haloalkyl, C1-3Haloalkyl or C1-2Haloalkyl groups are as defined above, but contain a different number of carbon atoms.

The term "side chain" means-Y-R7And (4) partial. In UDCA, -YR7is-CH2CH2-C (O) OH, and the variable side chain involved is-YR7The other part.

The term "isotopic variation" refers to isotopically-labeled compounds identical to those recited in formula (I) but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature, or wherein the proportion of atoms having atomic masses or mass numbers less commonly found in nature has increased (the latter concept is referred to as "isotopic enrichment"). Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, iodine and chlorine. For example2H (deuterium),3H、11C、13C、14C、18F、123I or125I (e.g.3H、11C、14C、18F、123I or125I) They may be naturally occurring isotopes or non-naturally occurring isotopes.

The salt of a compound of formula (I) is suitably a pharmaceutically acceptable surface salt. However, non-pharmaceutically acceptable salts may also be used, for example, in the preparation of intermediates in the preparation of compounds of formula (I) or in reactions to prepare compounds of formula (I) having alternative side chains.

Suitable salts of the compounds described herein include base addition salts such as sodium, potassium, calcium, aluminum, zinc, magnesium and other metal salts as well as choline, diethanolamine, ethanolamine, ethylenediamine, meglumine and other well known base addition salts, as outlined in Paulekuhn et al, j.med.chem.2007, 50, 6665-.

As mentioned above, in the compounds of formula (I), R2a、R2b、R3aAnd R3bEach independently is H or F, with the proviso that R2bAnd R3bAt least one of (a) is F.

In some compounds of formula (I), R3bIs F, and R2a、R2bAnd R3aEach of which is H or F. In a further compound of the formula (I), R2bIs F, and R2a、R3aAnd R3bEach of which is H or F.

Some suitable compounds of the invention are monofluorinated.

In some monofluorinated compounds, R3bIs F, and R3a、R2aAnd R2bI.e. the compound of formula (I) has a 4 β -fluoro substituent.

In some other monofluorinated compounds, R2bIs F, and R2a、R3aAnd R3bI.e. the compound of formula (I) has a 2 β -fluoro substituent.

Some suitable compounds of formula (I) are difluorinated. In some suitable difluorinated compounds of formula (I), R3bIs F, R3aIs H, R2aAnd R2bOne is F, and R2aAnd R2bI.e. the compound of formula (I) has a 2-fluoro substituent and a 4 β -fluoro substituent.

In another suitable compound of the formula (I) difluorinated, R3aAnd R3bAre both F, and R2aAnd R2bAre all H, so that the compounds of the general formula (I) are 4, 4-difluoro-substituted compounds。

The compound of formula (I) may be a trifluoride compound, wherein R is2a、R2b、R3aAnd R3bThree of F and another is H, or a tetrafluoride compound, wherein R is2a、R2b、R3aAnd R3bAll are F.

In the compounds of formula (I), suitably R6aAnd R6bAre both hydrogen, and/or R8Is methyl.

In some suitable compounds, R5Is ethyl, OH or methoxy, especially ethyl or methoxy, and especially ethyl.

In some suitable compounds, R5Is methoxy.

In particularly suitable compounds, R5Is ethyl.

In some suitable compounds of formula (I), Y is a bond.

In another suitable compound of formula (I), Y is C1-4Alkylene or C2-4Alkenylene linking groups, one of which is optionally substituted by one or more radicals R as defined above10And (4) substitution.

More suitably, in these compounds, Y is C1-4、C1-3Or C1-2An alkylene linker optionally substituted by one or more groups R as defined above10And (4) substitution.

In some compounds of formula (I), Y is unsubstituted.

In further compounds of the formula (I), Y is substituted by one or more substituents R10And (4) substitution.

In some cases, R10Is halogen.

In other cases, R10Is OH.

Examples of suitable linking groups Y include a bond, -CH2-、-CH2CH2-、-CH(OH)-CH2-, -CH ═ CH-or-CH ═ C (CH)3) -, in particular the valence bond, -CH2-、-CH2CH2-, -CH ═ CH-or-CH ═ C (CH)3) -, especially-CH2-、-CH2CH2-, -CH ═ CH-or-CH ═ C (CH)3)-。

More suitably, Y is a bond or C1-3An alkylene linker optionally substituted with one or more OH groups. Still more suitably, Y is C1-3An alkylene linker optionally substituted with one or more OH groups.

In some particularly suitable compounds, Y is unsubstituted C1-3An alkylene linker.

In particular, suitable compounds of the formula (I) are those in which Y is-CH2-or-CH2-CH2-, especially-CH2-CH2-。

In the compounds of the invention, R7More suitably C (O) NR17S(O)2R15Or NR17C(O)NR18S(O)2R15Wherein R is15、R17And R18As defined above for the compounds of general formula (I).

More suitably, R17And R18Each (if present) is H.

In some suitable compounds of formula (I), R15Selected from phenyl and 5 or 6 membered heteroaryl, for example pyridyl, pyrimidinyl or pyrrolyl, any of which may be unsubstituted or substituted by one or more substituents as defined herein, especially 1 to 3 substituents, more usually 1 or 2 substituents, and especially 1 substituent. More suitably, R15Is phenyl or 6-membered heteroaryl, any of which is unsubstituted or substituted by one or more substituents as defined above, especially 1-3 substituents, more usually 1 or 2 substituents, and especially 1 substituent.

R15Particularly suitable substituents for moieties are selected from fluorine, C1-4Alkyl radical, C1-4Fluoroalkyl group, O (C)1-4Alkyl) and O (C)1-4Fluoroalkyl groups).

In the compounds of the general formula (I) R15Examples of groups include phenyl, which is unsubstituted or substituted with a single substituent, said substituentSelected from fluorine, C1-4Alkyl radical, C1-4Fluoroalkyl group, O (C)1-4Alkyl) and O (C)1-4Fluoroalkyl) especially fluorine, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, trimethyl, methoxy and trifluoromethoxy.

When R is7Is NR17C(O)NR18S(O)2R15And R is15In the case of a phenyl group with a single substituent, the substituent may be at any position on the ring, but is most suitably an intero-para substituent, especially a para substituent. In particular, when R7Is NR17C(O)NR18S(O)2R15And R is15Is CF3When substituted phenyl, CF is preferred3The substituents are located in the meta or para position.

Examples of compounds of formula (I) include:

n, N' - (3 α, 7 α dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-toluenesulfonylurea (compound 1);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (compound 2);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (tert-butyl) benzenesulfonylurea (compound 3);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m-toluenesulfonylurea (compound 4);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-toluenesulfonylurea (compound 5);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-fluorobenzenesulfonylurea (compound 6);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m-fluorobenzenesulfonylurea (compound 7);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o-fluorobenzenesulfonylurea (compound 8);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p- (trifluoromethyl) benzenesulfonylurea (compound 9);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -m- (trifluoromethyl) benzenesulfonylurea (compound 10);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -o- (trifluoromethyl) benzenesulfonylurea (compound 11);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -4- (trifluoromethoxy) benzenesulfonylurea (compound 12);

n, N' - (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -p-methoxybenzenesulfonylurea (compound 13);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -p-trifluoromethoxybenzenesulfonamide (compound 14);

n- (3 α, 7 α -bis.hydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-oyl) -p-benzenesulfonamide (compound 15);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -3-fluorophenylsulfonamide (compound 16);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholane-24-acyl) -2-fluorophenylsulfonamide (compound 17);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -4-trifluoromethylphenylsulfonamide (compound 18);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -3-trifluoromethylphenylsulfonamide (compound 19);

n- (3 α, 7 α -dihydroxy-4 β -fluoro-6 α -ethyl-5 β -cholan-24-yl) -2-trifluoromethylphenylsulfonamide (compound 20);

n, N' - (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-24-nor-5 β -cholan-23-yl) -benzenesulfonylurea (Compound 21)

N- (3 α, 7 α -dihydroxy-4, 4-difluoro-6 α -ethyl-5 β -cholan-24-yl) -benzenesulfonamide (compound 22); and salts and isotopic variations thereof.

General formula (VII)(I) Wherein R is7Is NHC (O) N (R)18)S(O)2R15: deprotecting a compound of formula (II):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R40Is a protected OH group.

Suitably, the protecting group is one which can be removed by treatment with a base. Examples of protected OH groups of this type include R41C (O) O, wherein R41Is C1-6Alkyl or benzyl, especially methyl. Silyl ether protecting groups may also be used. Other suitable protecting groups for OH are well known to those skilled in the art (see Wuts, PGM and Greene, TW (2006)).

The compound of formula (II) may be prepared from a compound of formula (III):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R40As defined for formula (II);

with sulfonamides of the general formula (IV)

In the presence of a catalyst such as 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), and suitably in an organic solvent such as toluene,

wherein R is15And R18As defined for the compounds of general formula (I).

Sulfonamides of the general formula (IV) are known and readily available or can be prepared by known methods.

The compound of formula (III) may be prepared by heating the compound of formula (V) in an inert atmosphere, for example in an argon atmosphere:

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R40As defined for formula (II).

Suitably, the compound of formula (V) is heated to about 100-150 deg.C, typically about 125 deg.C.

The compound of formula (V) may be prepared from a compound of formula (VI):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R40As defined for formula (II);

prepared by reaction with diphenylphosphoryl azide under basic conditions, for example in the presence of triethylamine.

The reaction is suitably carried out in an inert atmosphere, for example in an argon atmosphere.

The compound of formula (VI) may be prepared by deprotecting a compound of formula (VII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of general formula (I).

For example, when the OH group R is protected40In the case of an acetoxy group, the compound is,the compound of formula (VII) may be reacted with acetic anhydride under mildly basic conditions, for example in the presence of sodium bicarbonate.

The reaction is suitably carried out in an inert atmosphere, for example in an argon atmosphere.

The compounds of formula (VII) may be prepared by hydrolysis of a compound of formula (VIII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R47Is C1-6Alkyl or benzyl.

The hydrolysis may be base catalysed, for example using an alkali metal hydroxide, for example sodium hydroxide or lithium hydroxide. Suitably, the reaction is carried out in an alcoholic solvent such as methanol.

A compound of the general formula (VIII), wherein R3bTo F, may be prepared by reduction of a suitable compound of formula (XX):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII).

Suitable reducing agents include hydrides, such as sodium borohydride. The reaction is suitably carried out under an inert atmosphere, for example argon, and in an organic solvent, for example tetrahydrofuran.

The process for the preparation of the compounds of the general formula (XX) and of the general formula (VIII), in which R is as follows2bIs F.

In an alternative process, the compound of formula (I) may be prepared by deprotecting a compound of formula (XII), wherein R is7Is NHC (O) N (R)18)S(O)2R15

Y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R45And R46Each is independently a protected OH group.

Suitably, R45And R46Each independently a silyl ether, such as tert-butyldimethylsiloxy or trimethylsiloxy. When R is45And R46In the case of silyl ethers, they may be the same or different, and deprotection may be carried out by treatment with a reagent such as tetrabutylammonium fluoride (TBAF). Alternative protecting groups may also be used, for example the group R as in formula (II) above41C (O) O. Other protecting groups are well known in the art (see Wuts, PGM and Greene, TW (2006)).

The compound of formula (XII) may be prepared from a compound of formula (XIII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R45And R46As defined for the compounds of the general formula (XII);

prepared by reaction with a sulphonamide of general formula (IV) as defined above in the presence of a catalyst such as 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU) and suitably in an organic solvent such as toluene.

The compound of formula (XIII) may be prepared by heating the compound of formula (XIV) in an inert atmosphere, for example in an argon atmosphere:

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R45And R46As defined for the compounds of the general formula (XII).

The compound of formula (XIV) may be prepared from a compound of formula (XV):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R45And R46As defined for the compounds of the general formula (XII);

prepared by reaction with diphenylphosphoryl azide under basic conditions, for example in the presence of triethylamine.

The compounds of formula (XV) can be prepared by hydrolysis of a compound of formula (XVI) as defined:

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), R45And R46As defined for the compounds of the general formula (XII), and R47As defined for formula (VIII).

Suitably, the hydrolysis is carried out under basic conditions, for example by treatment with an alkali metal hydroxide, such as sodium hydroxide or lithium hydroxide. Suitably, the reaction is carried out in an alcoholic solvent.

The compound of formula (XVI) can be prepared by protecting a compound of formula (XVII):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), R46As defined for formula (XII), and R47As defined for formula (VIII).

When protected OH group R45In the case of silyl ethers, protection may be effected by reacting a compound of formula (XVII) with a compound of formula (XVIII):

(R50)3Si-R48

(XVIII)

wherein R is48Is a leaving group, typically a halide, such as fluoride, chloride or bromide, or a sulfonyl group, such as trifluoromethanesulfonyl, tosyl or mesyl; and wherein R50Each independently is, for example C1-6Alkyl or phenyl, so that the radical R in the compound of the formula (XVI)45Is (R)50)3Si-O。

The reaction may be carried out in an inert atmosphere in the presence of a base such as 2, 6-lution, and at a temperature of about-5-5 ℃, typically 0 ℃. Suitable solvents include organic solvents such as methylene chloride.

The compounds of formula (XVII) can be prepared by reducing a compound of formula (XIX):

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), R46As defined for formula (XII), and R47As defined for formula (VIII).

Suitable reducing agents include hydrides, such as sodium borohydride. The reduction is suitably carried out in an inert atmosphere, such as argon, at a temperature of about 15-25 ℃, typically at room temperature, and in a solvent such as tetrahydrofuran.

Compounds of formula (XIX) may be prepared by protecting compounds of formula (XX), wherein R3b is F:

y, R therein2a、R2b、R3a、R3bAnd R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII).

When protected OH group R46In the case of silyl ethers, protection may be effected by reaction with a suitable silyl halide or sulfonate (e.g. triflate, mesylate or tosylate) in the presence of a base such as n-butyllithium. For example, when R is46In the case of trimethylsiloxy, the compound of formula (XX) may be reacted with trimethylsilylchloride.

Compounds of formula (I) can be prepared by deprotecting a compound of formula (XXII) wherein R is7Is C (O) N (R)17)S(O)2R15

Y, R therein2a、R2b、R3a、R3b、R5、R15And R17As defined for the compounds of the general formula (I), and R46As defined for formula (XII).

When protected OH group R46In the case of silyl ethers, deprotection can be achieved by treatment with a base such as TBAF. The reaction is suitably carried out in an inert atmosphere, for example an argon atmosphere, and in a dry organic solvent, for example tetrahydrofuran.

The compound of formula (XXII) may be represented by formula (XXIII):

wherein Y is,R2a、R2b、R3a、F3bAnd R5As defined for the compounds of the general formula (I), and R46As defined for formula (XII);

by reaction with sulfonamides of the formula (XXIV)

Wherein R is15And R17As defined for the compounds of general formula (I);

in the presence of a coupling agent such as 1-ethyl-3 (3-dimethylaminopropyl) carbodiimide (EDCI) and a base such as dimethylaminopyridine.

The compounds of formula (XXIII) may be prepared by hydrolysis of compounds of formula (XVII), suitably under basic conditions, for example by treatment with an alkali metal hydroxide, for example sodium hydroxide or lithium hydroxide. Suitably, the reaction is carried out in an alcoholic solvent.

For the compounds of the general formula (I), wherein R2bIs F, R3aAnd R3bIs H, and R7Is C (O) N (R)17)S(O)2R15An analogous route can be used starting from compounds of the general formula (VII) in which R is2bIs F, and R3aAnd R3bAre all H.

As described above, the compound of formula (XVII) can be prepared from the compound of formula (XX) by the compound of formula (XIX).

The compound of formula (XX) may be a compound of formula (XXa), (XXb), (XXc), (XXd) or (XXe):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII).

The compounds of formula (XXa) may be prepared from compounds of formula (XXX):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII);

by a process comprising:

(i) treatment with a strong base, e.g. n-BuLi or Lithium Diisopropylamide (LDA), and a trialkylsilyl halide, e.g. trimethylsilyl chloride, followed by

(ii) For example, having the following structure(1-chloromethyl-4-fluoro-1, 4-diazobicyclo [2.2.2]Octane bis (tetrafluoroborate) electrophilically fluorinates the product of step (i):

when in useThe reaction is suitably carried out at a temperature of from 15 to 25 ℃, typically at room temperature, in a polar organic solvent such as acetonitrile.

Suitable bases include Lithium Diisopropylamide (LDA) and n-butyllithium, which are typically used in combination with trimethylsilyl chloride.

The compound of formula (XXb) can be prepared from the compound of formula (XXa) by further fluorination, typically by a method analogous to that described above for the compound of formula (XXa), i.e. by a method comprising the following steps:

(i) treatment with a strong base such as n-BuLi or LDA and a trialkylsilyl halide, e.g. trimethylsilyl chloride, followed by

(ii) For example by(ii) electrophilic fluorination of the product of step (i).

The compounds of formula (XXc) may be prepared from compounds of formula (XXb) by treatment with a strong base, typically in combination with a trialkylsilyl halide, e.g. trimethylsilylchloride, and then withAnd (3) preparing by fluorination.

The compounds of the formula (XXd) can be prepared from compounds of the formula (XXc) by further fluorination, typically withAnd weak bases such as triethylamine.

Compounds of formula (XXe) may be prepared from compounds of formula (XXa) by reaction with compounds of formula (XXXI)

R51-Si(R13)3 (XXXI)

Wherein R is13Each independently selected from C1-6Alkyl and phenyl, and R51Is halogen, such as chlorine;

in the presence of a strong base such as lithium diisopropylamide, followed by fluorination, typically withAnd reacting to prepare the compound.

Compounds of formula (XXX) may be prepared from compounds of formula (XXXII) by oxidation:

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII); the oxidation is typically carried out using reagents such as (2, 2, 6, 6-tetramethylpiperidin-1-yl) oxidinyl (TEMPO) or (diacetoxyiodo) benzene (BAIB).

Compounds of formula (XXXII) may be prepared by esterification of a carboxylic acid of formula (XXXIII):

wherein Y and R5As defined for the compounds of general formula (I);

by reaction with the formula R47Alcohols of the-OH group are prepared by reaction in the presence of an acid such as p-toluenesulfonic acid or methanesulfonic acid.

Compounds of the general formula (XXXIII) are known. The compound of formula (XXXIII) is obeticholic acid, wherein R5Is ethyl and Y is-CH2CH2- (see WO 02/072598).

Our earlier applications WO 2016/079518, WO 2016/079518, WO 2016/079519, WO 2016/079520, WO 2017/199036 and WO 2017/199033 describe processes for the preparation of compounds of general formula (XXXIII) according to scheme 1, wherein Y is as defined for general formula (I).

Scheme 1

Some compounds of the general formulae (XL) and (XLI) are known and, for example, Uekawa et al (2004) describe the synthesis of ethyl (22E) -3-oxo-4, 22-choledien-24-oate from stigmasterol:

it is then converted into ethyl (22E) -3-oxo-4, 6, 22-choletriene-24-oate:

uekawa et al then proceed to describe the conversion of this compound to ethyl (6 α, 7 α, 22E) -6, 7-epoxy-3-oxo-4, 22-choledien-24-oate.

Other compounds of formula (XL) and (XLI) can be prepared by similar methods from phytosterols similar to stigmasterol.

Stigmasterol and other phytosterols are phytosterols and are readily available or can be prepared by known routes.

The compounds of formula (VIII) can be prepared by reduction of compounds of formula (XLV) wherein R2bIs F, and R2a、R3aAnd R3bAre all H:

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII).

Suitable reducing agents include hydrides, such as sodium borohydride, and the reaction is carried out in an inert atmosphere, such as an argon atmosphere.

The product was obtained as a mixture with the 3 β -hydroxy isomer.

Compounds of formula (XLV) can be prepared by esterifying compounds of formula (XLVI):

wherein Y and R5As defined for the compounds of general formula (I).

Suitably, esterification is by reaction with a halide, such as alkyl or benzyl R47In the presence of a base such as cesium carbonate and in an alcoholic solvent such as methanol.

Compounds of formula (XLVI) may be prepared by epimerization of compounds of formula (XLVII):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII).

The epimerization may be performed by treating a compound of formula (XLVII) with a strong base such as sodium hydroxide, which results in OR47Partially hydrolyzed to OH. The product obtained is a 60: 40 mixture of the compound of formula (XLVI) and its 2 α isomer. This mixture is then used without further isolation in the re-esterification step described above, since the ester of formula (XLV) is more easily separated from its 2 α isomer than the carboxylic acid (XLVI).

The compounds of formula (XIVII) may be prepared by oxidation of a compound of formula (XLVIII):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII). Suitable oxidizing agents include Dess-Martin reagent (Dess-Martin periodinane).

The compound of formula (XLVIII) may be prepared from a compound of formula (XLIX):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII); with hf pyridine (70%) complex in an inert atmosphere, such as argon. Suitably, the reaction is carried out in a halogenated solvent such as dichloromethane at reduced temperature, for example-10 to 10 ℃, typically at a temperature of about 0 ℃.

Compounds of formula (XLIX) can be prepared from compounds of formula (L) by oxidation:

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII) and the dotted line represents one of the bondsA single bond and the other a double bond;

for example, m-perchlorobenzoic acid is used in a halogenated organic solvent such as dichloromethane. Suitably, the reaction is carried out at a temperature of from 15 to 25 ℃, typically at room temperature.

The compounds of formula (L) can be prepared from compounds of formula (LI) by dehydration:

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII); a suitable dehydrating agent, such as trifluoromethanesulfonic anhydride, is used in the presence of a base such as dimethylaminopyridine. Suitably, the reaction is carried out in a halogenated organic solvent such as dichloromethane at a temperature of from 15 to 25 ℃, typically at room temperature.

The compound of formula (LI) may be prepared by reacting a compound of formula (LII):

wherein Y and R5As defined for the compounds of the general formula (I), and R47As defined for formula (VIII); with reducing agents, e.g.(lithium tri-sec-butylborohydride) and then treated with hydrogen peroxide.

Suitably withThe reaction of (a) is carried out under an inert atmosphere, for example under an argon atmosphere, and at reduced temperature, typically at-78 ℃. The reaction with hydrogen peroxide is suitably carried out at a temperature of about 0 ℃.

The compounds of general formula (LII) can be prepared by esterification of a compound of general formula (LIII):

wherein Y and R5As defined for the compounds of general formula (I).

Suitably, esterification may be by reaction with an alcohol R47The reaction of-OH is carried out in the presence of an acid containing a leaving group, such as p-toluenesulfonic acid. The reaction may be carried out at a temperature of about 20-40 c, for example about 30 c.

The compounds of the general formula (LIII) are known and can be prepared by methods known to the person skilled in the art, for example the methods taught in WO 2016/079518, WO 2016/079518, WO 2016/079519, WO 2016/079520, WO 2017/199036 and WO 2017/199033.

The compounds of general formula (I) are FXR agonists and are therefore useful in the treatment or prevention of FXR mediated diseases and conditions.

Accordingly, another aspect of the present invention provides a compound of formula (I) for use in medicine.

The compounds may be used in human or veterinary medicine, and are suitable for the treatment of mammals, particularly humans.

The compounds of formula (I) are particularly useful in the treatment or prevention of metabolic syndrome, including nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; and liver damage due to fibrosis.

The invention also provides the use of a compound of formula (I) in the manufacture of a medicament for the treatment or prevention of: nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis.

Also provided are methods for treating or preventing nonalcoholic steatohepatitis (NASH); primary biliary cirrhosis; primary sclerosing cholangitis; closing the biliary tract; cholestatic liver disease; hepatitis c infection; alcoholic liver disease; fiberizing; or liver damage due to fibrosis, comprising administering to a patient in need of such treatment an effective amount of a compound of formula (I).

Fibrosis includes fibrosis of the liver, kidney and intestine.

Liver fibrosis may be associated with NASH, alcoholic liver disease or non-alcoholic fatty liver. Alternatively, liver fibrosis may be associated with an infection, such as hepatitis, particularly hepatitis b or c or parasitic liver disease. Other causes of liver fibrosis include lesions caused by congenital diseases such as Wilson's disease, gaucher's disease, glycogen storage disease, hemochromatosis, Zellweger's syndrome, and congenital liver fibrosis. Liver fibrosis can also be induced by drugs such as chlorpromazine, tosylbutamide, methotrexate, isoniazid, and methyldopa.

Renal fibrosis may be associated with diseases such as diabetic nephropathy, hypertensive nephrosclerosis, glomerulonephritis, interstitial nephritis, transplantation-related glomerulopathy and polycystic kidney disease.

Intestinal fibrosis may be associated with intestinal diseases.

Bowel diseases include irritable bowel syndrome, crohn's disease and ulcerative colitis.

The compounds of formula (I) are suitably provided in the form of a pharmaceutical composition, and another aspect of the invention provides a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable excipient or carrier.

The particular excipient or carrier employed will depend on the route of administration chosen and must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

The compositions of the present invention may also contain one or more other active ingredients useful for treating one or more of the diseases and conditions described above.

These formulations include those suitable for oral, rectal, nasal, bronchial (inhalation), topical (including eye drops, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and may be prepared by any method well known in the art of pharmacy.

The composition may be prepared by mixing the active agent as defined above with a carrier. In general, the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. The invention extends to a process for the preparation of a pharmaceutical composition which comprises mixing or bringing into association a compound of general formula (I) with a pharmaceutically or veterinarily acceptable excipient or carrier.

The formulations of the invention for oral administration may be prepared as follows: discrete units, such as capsules, sachets or tablets, each containing a predetermined amount of active agent; as a powder or granule; as a solution or suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion; or as boluses, etc.

For compositions for oral administration (e.g., tablets and capsules), the term "acceptable carrier" includes vehicles, e.g., common excipients, such as binding agents, e.g., syrup, acacia, gelatin, tragacanth, polyvinylpyrrolidone (povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sucrose and starch; fillers and carriers, such as corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride, and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metal stearates, glyceryl stearate, stearic acid, silicone fluids, talc, oils and colloidal silica. Flavoring agents such as peppermint, oil of wintergreen, cherry flavoring, and the like may also be used. It may be desirable to add a colorant to make the dosage form easily identifiable. Tablets may also be coated by methods well known in the art.

Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surfactant or dispersing agent. Molded tablets may be prepared by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated to provide slow or controlled release of the active agent.

Other formulations suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.

For topical application to the skin, the compounds of formula (I) or (II) may be formulated as creams, ointments, jellies, solutions or suspensions, and the like. Cream or ointment formulations which may be used in medicine are well known conventional formulations, for example as described in the pharmaceutical standard textbook British Pharmacopoeia (British Pharmacopoeia).

Parenteral formulations are generally sterile.

81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:可用于对皮肤、毛发、指甲和/或粘膜进行处理和/或护理的化合物

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!