WO2013007387A1 - Novel fxr (nr1h4) binding and activity modulating compounds - Google Patents

Novel fxr (nr1h4) binding and activity modulating compounds Download PDF

Info

Publication number
WO2013007387A1
WO2013007387A1 PCT/EP2012/002941 EP2012002941W WO2013007387A1 WO 2013007387 A1 WO2013007387 A1 WO 2013007387A1 EP 2012002941 W EP2012002941 W EP 2012002941W WO 2013007387 A1 WO2013007387 A1 WO 2013007387A1
Authority
WO
WIPO (PCT)
Prior art keywords
liver
alkyl
fxr
compound
group
Prior art date
Application number
PCT/EP2012/002941
Other languages
French (fr)
Inventor
Olaf Kinzel
Christoph Steeneck
Claus Kremoser
Original Assignee
Phenex Pharmaceuticals Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP2014519453A priority Critical patent/JP5986633B2/en
Priority to MEP-2016-81A priority patent/ME02434B/en
Priority to CN201280033148.4A priority patent/CN103702719B/en
Priority to CA2839357A priority patent/CA2839357C/en
Priority to KR1020177021651A priority patent/KR101859533B1/en
Priority to KR1020147002937A priority patent/KR101722410B1/en
Priority to US14/232,118 priority patent/US9139539B2/en
Priority to KR1020177000763A priority patent/KR101766323B1/en
Priority to BR122020008872-9A priority patent/BR122020008872B1/en
Priority to MX2014000534A priority patent/MX368371B/en
Priority to NZ620177A priority patent/NZ620177B2/en
Priority to EP12735100.5A priority patent/EP2731676B1/en
Priority to AU2012283387A priority patent/AU2012283387C1/en
Priority to BR122019026062-1A priority patent/BR122019026062B1/en
Priority to EP17000383.4A priority patent/EP3246070B1/en
Priority to ES12735100.5T priority patent/ES2569718T3/en
Priority to BR112014000260-6A priority patent/BR112014000260B1/en
Application filed by Phenex Pharmaceuticals Ag filed Critical Phenex Pharmaceuticals Ag
Priority to EA201391674A priority patent/EA024843B1/en
Priority to SI201230535A priority patent/SI2731676T1/en
Priority to PL12735100T priority patent/PL2731676T3/en
Priority to DK12735100.5T priority patent/DK2731676T3/en
Priority to KR1020187013304A priority patent/KR101934335B1/en
Priority to RS20160266A priority patent/RS54786B1/en
Publication of WO2013007387A1 publication Critical patent/WO2013007387A1/en
Priority to ZA2013/09521A priority patent/ZA201309521B/en
Priority to IL229944A priority patent/IL229944A/en
Priority to HK14105521.1A priority patent/HK1192181A1/en
Priority to US14/824,971 priority patent/US9539244B2/en
Priority to HRP20160442TT priority patent/HRP20160442T1/en
Priority to SM201600169T priority patent/SMT201600169B/en
Priority to AU2016265993A priority patent/AU2016265993B2/en
Priority to US15/369,521 priority patent/US9820979B2/en
Priority to IL253437A priority patent/IL253437A0/en
Priority to US15/783,530 priority patent/US10220027B2/en
Priority to AU2018203613A priority patent/AU2018203613B2/en
Priority to US16/248,178 priority patent/US10485795B2/en
Priority to IL265973A priority patent/IL265973B/en
Priority to AU2019261667A priority patent/AU2019261667B2/en
Priority to IL277570A priority patent/IL277570A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to compounds which bind to the NR1 H4 receptor (FXR) and act as agonists or modulators of FXR.
  • the invention further relates to the use of the compounds for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds.
  • Multicellular organisms are dependent on advanced mechanisms of information transfer between cells and body compartments.
  • the information that is transmitted can be highly complex and can result in the alteration of genetic programs involved in cellular differentiation, proliferation, or reproduction.
  • the signals, or hormones are often low molecular weight molecules, such as peptides, fatty acid, or cholesterol derivatives.
  • NR nuclear receptors
  • Orphan receptors may be indicative of unknown signalling pathways in the cell or may be nuclear receptors that function without ligand activation. The activation of transcription by some of these orphan receptors may occur in the absence of an exogenous ligand and/or through signal transduction pathways originating from the cell surface (D. J. Mangelsdorf et al., Cell 1995, 83, 835; R. M. Evans, Mol. Endocrinol. 2005, 19, 1429).
  • DBD DNA-binding domain
  • HRE Hormone Responsive Element
  • Coactivators or transcriptional activators are proposed to bridge between sequence specific transcription factors, the basal transcription machinery and in addition to influence the chromatin structure of a target cell.
  • proteins like SRC-1 , ACTR, and Gripl interact with NRs in a ligand enhanced manner (D. M. Heery et al., Nature 1997, 387, 733; T. Heinzel et al., Nature 1997, 387, 43; K. W. Nettles and G. L. Greene, Annu. Rev. Physiol. 2005, 67, 309).
  • Nuclear receptor modulators like steroid hormones affect the growth and function of specific cells by binding to intracellular receptors and forming nuclear receptor-ligand complexes. Nuclear receptor-hormone complexes then interact with a HRE in the control region of specific genes and alter specific gene expression (A. Aranda and A. Pascual, Physiol. Rev. 2001 , 81 , 1269).
  • the Famesoid X Receptor alpha (hereinafter also often referred to as NR1 H4 when referring to the human receptor) is a prototypical type 2 nuclear receptor which activates genes upon binding to promoter region of target genes in a heterodimeric fashion with Retinoid X Receptor (B. M. Forman et al., Cell 1995, 81 , 687).
  • the relevant physiological ligands of NR1 H4 are bile acids (D. J. Parks et al., Science 1999, 284, 1365; M. Makishima et al., Science 1999, 284, 1362).
  • chenodeoxycholic acid which regulates the expression of several genes that participate in bile acid homeostasis.
  • Farnesol and derivatives, together called farnesoids, are originally described to activate the rat orthologue at high concentration but they do not activate the human or mouse receptor.
  • FXR is expressed in the liver, throughout the entire gastrointestinal tract including the esophagus, stomach, duodenum, small intestine, colon, ovary, adrenal gland and kidney. Beyond controlling intracellular gene expression, FXR seems to be also involved in paracrine and endocrine signalling by upregulating the expression of the cytokine Fibroblast Growth Factor 15 (rodents) or 9 (monkeys, humans, J. A. Holt et al., Genes Dev. 2003, 17, 1581 ; T. Inagaki et al., Cell Metab. 2005, 2, 217).
  • Small molecule compounds which act as FXR modulators have been disclosed in the following publications: WO 2000/037077, WO 2003/015771 , WO 2004/048349, WO 2007/076260, WO 2007/092751 , WO 2007/140174, WO 2007/140183, WO 2008/051942, WO 2008/157270, WO 2009/005998, WO 2009/012125, WO 2008/025539 and WO 2008/025540. Further small molecule FXR modulators have been recently reviewed (M. L. Crawley, Expert Opin Ther. Pat. 2010, 20,1047; D. Merk et al., Future Med. Chem. 2012, 4, 1015).
  • the problem underlying the present invention is to generate FXR-agonists with improved physicochemical properties in general, and reduced hydrophobicity, improved aqueous solubility and better membrane permeability, in particular, compared to compounds claimed in WO 2011/020615.
  • R is selected from the group consisting of COORe, CONR7R8, tetrazolyl, SO2NR7R8, C1-6 alkyl, S0 2 -Cve alkyl and H, with R 6 independently selected from the group consisting of H or d-e alkyl, and R 7 and R 8 independently from each other selected from the group consisting of H, alkyl, halo-Ci-6 alkyl, Ci. 6 alkylene-R 9 , S0 2 -C 1 . 6 alkyl, wherein R 9 is selected from the group consisting of COOH, OH and S0 3 H;
  • A is selected from the group consisting of phenyl, pyridyl, pyrimidyl, pyrazolyl, indolyl, thienyl, benzothienyl, indazolyl, benzisoxazolyl, benzofuranyl, benzotriazolyl, furanyl, benzothiazolyl, thiazolyl, oxadiazolyl, each optionally substituted with one or two groups independently selected from the group consisting of OH, alkyl, O-halo-d-e alkyl, d. 6 alkyl, halo-Ci. 6 alkyl, C3.6 cycloalkyl and halogen;
  • Q is selected from the group consisting of phenyl, pyridyl, thiazolyl, thiophenyl, pyrimidyl, each optionally substituted with one or two groups independently selected from the group consisting of d-e alkyl, halo-Ci- 6 alkyl, halogen and CF 3 ;
  • Y is selected from N or CH
  • Ri is selected from the group consisting of hydrogen, d. 3 alkyl, C 3 ⁇ cylcoalkyl, C4-5 alkylcycloalkyl, wherein d. 3 alkyl is optionally substituted with 1 to 3 substituents independently selected from halogen, hydroxy or d ⁇ alkoxy;
  • R 2 and R 3 are independently selected from the group consisting of hydrogen, C 1 -3 alkyl, d. 3 haloalkyl, d- 3 alkoxy, C1.3 haloalkoxy and halogen.
  • R-A in the compound according to Formula (1) is selected from
  • Z in the compound according to Formula (1) is
  • the compound according to Formula (1) is selected from
  • R is selected from the group consisting of C0 2 H, CONHS0 2 Me, and tetrazolyl.
  • the present invention is directed to a compound according to Formula (1) for use as a medicament.
  • the present invention is directed to a compound according to Formula (1) for use in the prophylaxis and/or treatment of diseases mediated by FXR.
  • the present invention is directed to the use of a compound according to Formula (1) for the preparation of a medicament for the prophylaxis and/or treatment of diseases mediated by FXR.
  • the disease is selected from chronic intrahepatic or some forms of extrahepatic cholestatic conditions; liver fibrosis; obstructive or chronic inflammatory disorders of the liver; liver cirrhosis; liver steatosis and associated syndromes, cholestatic or fibrotic effects that are associated with alcohol-induced cirrhosis or with viral-borne forms of hepatitis; liver failure or liver ischemia after major liver resection; chemotherapy associated steatohepatitis (CASH); acute liver failure; and/or Inflammatory Bowel Diseases.
  • CASH chemotherapy associated steatohepatitis
  • the disease is selected from lipid and lipoprotein disorders; Type II Diabetes and clinical complications of Type I and Type II Diabetes, including diabetic nephropathy, diabetic neuropathy, diabetic retinopathy and other observed effects of clinically manifest long term Diabetes; conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways, such as Non- Alcoholic Fatty Liver Disease (NAFLD), or Non- Alcoholic Steatohepatitis (NASH); obesity or metabolic syndrome (combined conditions of dyslipidemia, diabetes or abnormally high body-mass index); and/or cute myocardial infarction, acute stroke or thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis.
  • NAFLD Non- Alcoholic Fatty Liver Disease
  • NASH Non- Alcoholic Steatohepatitis
  • obesity or metabolic syndrome combined conditions of dyslipidemia, diabetes or abnormally high body-mass index
  • the disease is selected from non-malignant hyperproliferative disorders and malignant hyperproliferative disorders, specifically of hepatocellular carcinoma, colon adenoma and polyposis, colon adenocarcinoma, breast cancer, pancreas adenocarcinoma, Barrett's esophagus or other forms of neoplastic diseases of the gastrointestinal tract and the liver.
  • the improved physico-chemical properties have been achieved by the introduction of a polar hydroxyl group on a 1 ,3-cyclobutyltdene or 1 ,3-azetidinylidene group replacing the former 1 ,2- cyclopropylidene ring.
  • the resulting compounds maintained their activity on the FXR receptor but demonstrated improved physico-chemical properties, such as higher aqueous solubility and/or membrane permeability.
  • the compounds of the present invention share a common chemical structure according to Formula (1) in claim 1.
  • the present invention is directed to an enantiomer, diastereomer or pharmaceutically acceptable salt of a compound according to Formula (1).
  • R in Formula (1) is selected from the group consisting of COOR 6 , CONR 7 R 8 , S0 2 NR 7 R 8 , and S0 2 -Ci. 6 alkyl.
  • R 6 in Formula (1) is H.
  • R 7 and R 8 in Formula (1) are independently from each other selected from the group consisting of H and S0 2 -Ci. 6 alkyl.
  • R 7 in Formula (1) is H.
  • R 8 in Formula (1) is SC C ⁇ alkyl.
  • A is selected from the group consisting of phenyl, pyridyl, pyrimidyl, pyrazolyl, indazolyl, and oxadiazolyl.
  • A is substituted with one or two groups independently selected from C 1 .6 alkyl, more preferably C 1 .3 alkyl. In another preferred embodiment in combination with any of the above or below embodiments, A is unsubstituted.
  • Q is phenyl
  • Q is substituted with one or two groups independently selected from halogen, more preferably one group selected from halogen, in particular CI.
  • Z is
  • X CH.
  • Ri is C 3 -6 cylcoalkyl, in particular cyclopropyl.
  • R 2 and R 3 are independently selected from halogen, in particular CI.
  • the compounds of the present invention can be in the form of a prodrug compound.
  • Prodrug compound means a derivative that is converted into a compound according to the present invention by a reaction with an enzyme, gastric acid or the like under a physiological condition in the living body, e.g. by oxidation, reduction, hydrolysis or the like, each of which is carried out enzymatically.
  • prodrug examples include compounds, wherein the amino group in a compound of the present invention is acylated, alkylated or phosphorylated to form, e.g., eicosanoylamino, alanylamino, pivaloyloxymethylamino or wherein the hydroxyl group is acylated, alkylated, phosphorylated or converted into the borate, e.g. acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy or wherein the carboxyl group is esterified or amidated.
  • these compounds can be produced from compounds of the present invention according to well-known methods.
  • prodrug examples are compounds, wherein the carboxylate in a compound of the present invention is, for example, converted into an alkyl-, aryl-, choline-, amino, acyloxymethylester, linolenoylester.
  • Metabolites of compounds of the present invention are also within the scope of the present invention.
  • tautomerism like e.g. keto-enol tautomerism
  • the individual forms like e.g. the keto and enol form, are each within the scope of the invention as well as their mixtures in any ratio. Same applies for stereoisomers, like e.g. enantiomers, cis/trans isomers, conformers and the like.
  • isomers can be separated by methods well known in the art, e.g. by liquid chromatography. Same applies for enantiomers by using e.g. chiral stationary phases. Additionally, enantiomers may be isolated by converting them into diastereomers, i.e. coupling with an enantiomerically pure auxiliary compound, subsequent separation of the resulting diastereomers and cleavage of the auxiliary residue. Alternatively, any enantiomer of a compound of the present invention may be obtained from stereoselective synthesis using optically pure starting materials. Another way to obtain pure enantiomers from racemic mixtures would use enantioselective crystallization with chiral counterions.
  • the compounds of the present invention can be in the form of a pharmaceutically acceptable salt or a solvate.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic bases or acids and organic bases or acids.
  • the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts.
  • the compounds of the present invention which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or ammonium salts.
  • salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanotamine or amino acids.
  • the compounds of the present invention which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids.
  • suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p- toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art.
  • the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions).
  • inner salts or betaines can be obtained by customary methods which are known to the person skilled in the art like, for example, by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts.
  • the present invention also includes all salts of the compounds of the present invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts.
  • solvates such as those which include as solvate water, or pharmaceutically acceptable solvates, such as alcohols, in particular ethanol.
  • the present invention provides pharmaceutical compositions comprising at least one compound of the present invention, or a prodrug compound thereof, or a pharmaceutically acceptable salt or solvate thereof as active ingredient together with a pharmaceutically acceptable carrier.
  • “Pharmaceutical composition” means one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • the pharmaceutical compositions of the present invention encompass any composition made by admixing at least one compound of the present invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition of the present invention may additionally comprise one or more other compounds as active ingredients like a prodrug compound or other nuclear receptor modulators.
  • compositions are suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (nasal or buccal inhalation) or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy.
  • the compounds of the present invention can be prepared by a combination of methods described in Schemes I to III.
  • M metal, e.g. Li
  • Y two isomers can form (A and Q transannular cis or trans to each other). Under optimized conditions the formation of mainly one of the two isomers can be achieved.
  • the two isomers can be separated by appropriate methods known in the art like e.g. silica gel chromatography or preparative RP-HPLC.
  • dehalogenation e.g. with Zn in acetic acid under reflux, the desired 3-substituted cyclobutanones are obtained.
  • the vinyl-intermediates can react with in situ generated unsubtituted ketene to afford in one step the desired cyclobutanone intermediates.
  • 3-methylenecyclobutanecarbonitrile is used as starting material.
  • Substituted heterocycles can be built up from the cyano group in several steps by methods known to those skilled in the art.
  • the desired cyclobutanones can be obtained by oxidative cleavage of the exocyclic double bond using conditions and reagents known to those skilled in the art, e.g. by the use of Os0 4 , ozone or RhCi NalC ⁇ as oxidants.
  • Option c) shows the methods used to prepare the substituted azetidinones.
  • HetAr heteroaromatic ring c) Cu or Pd catalysed
  • Scheme III illustrates some possibilities to perform modifications of the substituents at the A group after the formation of the 4-membered hydroxy-bearing rings.
  • a leaving group X e.g. bromide
  • a cyano group e.g. carboxylic ester, methylsulfonyl or thioether by transition metal catalysed cross coupling reactions.
  • the obtained derivatives can be further transformed into other derivatives by methods known to those skilled in the art.
  • the cyano and the ester group can be hydrolysed under basic conditions to the afford a carboxylic acid which in turn can be transformed into acyl-sulfonamides.
  • a benzyl thioether can be chlorinated to afford the chlorosulfonyl intermediate which reacts with ammonia to the corresponding sulfonamides.
  • the present invention relates to compounds according to the general Formula (1) which bind to FXR and act as agonists or modulators of FXR.
  • the invention further relates to the use of said compounds for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds. Further the present invention relates to the use of said compounds for the preparation of a medicament for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds.
  • the present invention relates to the use of compounds according to Formula (1) in the preparation of a medicament for the prophylaxis and/or treatment of chronic intrahepatic or some forms of extrahepatic cholestatic conditions, of liver fibrosis, of acute intraheptic cholestatic conditions, of obstructive or chronic inflammatory disorders that arise out of improper bile composition, of gastrointestinal conditions with a reduced uptake of dietary fat and fat-soluble dietary vitamins, of inflammatory bowel diseases, of lipid and lipoprotein disorders, of Type II Diabetes and clinical complications of Type I and Type II Diabetes, of conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways, of obesity and metabolic syndrome (combined conditions of dyslipidemia, diabetes and abnormally high body-mass index), of acute myocardial infarction, of acute stroke, of thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis,
  • Medicaments as referred to herein may be prepared by conventional processes, including the combination of a compound according to the present invention and a pharmaceutically acceptable carrier.
  • FXR is proposed to be a nuclear bile acid sensor. As a result, it modulates both, the synthetic output of bile acids in the liver and their recycling in the intestine (by regulating bile acid binding proteins). But beyond bile acid physiology, FXR seems to be involved in the regulation of many diverse physiological processes which are relevant in the etiology and for the treatment of diseases as diverse as cholesterol gallstones, metabolic disorders such as Type II Diabetes, dyslipidemias or obesity, chronic inflammatory diseases such as Inflammatory Bowel Diseases or chronic intrahepatic forms of cholestasis and many others diseases (T. Claudel et al., Arterioscler. Thromb. Vase. Biol. 2005, 25, 2020; Y. D. Wang et al., Cell Res. 2008, 18, 1087.
  • FXR regulates a complex pattern of response genes in the liver and in the gastrointestinal tract.
  • the gene products have impact on diverse physiological processes.
  • the first regulatory network that was analyzed was the regulation of bile acid synthesis. While the LXRs induce the key enzyme of the conversion of cholesterol into bile acids, Cyp7A1 , via the induction of the regulatory nuclear receptor LRH-1 , FXR represses the induction of Cyp7A1 via the upregulation of mRNA encoding SHP, a further nuclear receptor that is dominant repressive over LRH-1.
  • FXR binds the end products of this pathway, primary bile acids such as cholic acid (CA) or CDCA, this can be regarded as an example of feedback inhibition on the gene expression level (B. Goodwin et al., Mol. Cell 2000, 6, 517; T. T. Lu et al., Mol. Cell 2000, 6, 507).
  • CA cholic acid
  • CDCA CDCA
  • ABC ATP-binding cassette
  • MRP-2 (ABCC4)
  • MDR-3 (ABCB4)
  • FXR farnesoid receptor
  • FXR seems to be the major metabolite sensor and regulator for the synthesis, export and re-circulation of bile acids
  • FXR ligands to induce bile flow and change bile acid composition towards more hydrophilic composition.
  • This hepatoprotective effect was further narrowed down to an anti-fibrotic effect that results from the repression of Tissue Inhibitors of Matrix-Metalloproteinases, TIMP-1 and 2, the induction of collagen-deposit resolving Matrix- Metalloproteinase 2 in hepatic stellate cells and the subsequent reduction of alpha-collagen mRNA and Transforming growth factor beta (TGF-beta) mRNA which are both pro-fibrotic factors by FXR agonists (S. Fiorucci et al., Gastroenterology 2004, 127, 1497; S. Fiorucci et al., J. Pharmacol. Exp. Ther. 2005, 314, 584).
  • TGF-beta Transforming growth factor beta
  • FXR binding compounds will demonstrate substantial clinical utility in the therapeutic regimen of chronic cholestatic conditions such as Primary Biliary Cirrhosis (PBC) or Primary Sclerosing Cholangitis (PSC) (reviewed in: G. Rizzo et al., Curr. Drug Targets Immune Endocr. Metabol. Disord. 2005, 5, 289; G. Zollner et al., Mol. Pharm. 2006, 3, 231 ; S. Y. Cai et al., Expert Opin. Ther. Targets 2006, 10, 409).
  • PBC Primary Biliary Cirrhosis
  • PSC Primary Sclerosing Cholangitis
  • FXR activation has on bile acid metabolism and excretion is not only relevant for cholestatic syndromes but even more directly for a therapy against gallstone formation.
  • FXR polymorphisms map as quantitative trait loci as one factor contributing to gallstone disease (H. Wittenburg, Gastroenterology 2003, 125, 868).
  • FXR small molecule agonists that can be used to prevent cholesterol gallstone formation or to prevent re-formation of gallstones after surgical removal or Shockwave lithotripsy (discussed in: S. A. Doggrell, Curr. Opin. Investig. Drugs 2006, 7, 344).
  • the compound according to Formula (1) and pharmaceutical compositions comprising said compound is used for the prophylaxis and/or treatment of obstructive or chronic inflammatory disorders that arise out of improper bile composition such as cholelithiasis also known as cholesterol gallstones.
  • the compounds according to the invention and pharmaceutical compositions comprising said compounds are used in the treatment of liver diseases such as HCC, stimulation of liver regrowth and amelioration of side effects associated with major liver resection, liver cirrhosis independent of the etiology and prevention or treatment of liver ischemia in the course of liver transplantation or major liver surgery.
  • Urizar et al. Science 2002, 296, 1703; G. Lambert et al., J. Biol. Chem. 2003, 278, 2563; M. Watanabe et al., J. Clin. Invest. 2004, 113, 1408; A. Figge et al., J. Biol. Chem. 2004, 279, 2790; S. Bilz et al., Am. J. Physiol. Endocrinol. Metab. 2006, 290, E716).
  • FXR binding compounds are thought to be good candidates for the treatment of Type II Diabetes because of their insulin sensitization, glycogenogenic, and lipid lowering effects.
  • the compounds according to the invention and pharmaceutical compositions comprising said compounds are used in the prophylaxis and/or treatment of Type II Diabetes which can be overcome by FXR-mediated upregulation of systemic insulin sensitivity and intracellular insulin signalling in liver, increased peripheral glucose uptake and metabolisation, increased glycogen storage in liver, decreased output of glucose into serum from liver-borne gluconeogenesis.
  • said compounds and pharmaceutical compositions are used for the prophylaxis and/or treatment of chronic intrahepatic, such as PBC, PSC, progressive familiar cholestasis (PFIC), alcohol-induced cirrhosis and associated cholestasis, and some forms of extrahepatic cholestatic conditions, or liver fibrosis.
  • chronic intrahepatic such as PBC, PSC, progressive familiar cholestasis (PFIC), alcohol-induced cirrhosis and associated cholestasis, and some forms of extrahepatic cholestatic conditions, or liver fibrosis.
  • the invention also relates to a compound of Formula (1) or to a pharmaceutical composition comprising said compound for the prophylaxis and/or treatment of gastrointestinal conditions with a reduced uptake of dietary fat and fat-soluble dietary vitamins which can be overcome by increased intestinal levels of bile acids and phospholipids.
  • said compound or pharmaceutical composition is used for preventing and/or treating a disease selected from the group consisting of lipid and lipoprotein disorders such as hypercholesterolemia, hypertriglyceridemia, and atherosclerosis as a clinically manifest condition which can be ameliorated by FXR ' s beneficial effect on lowering total plasma cholesterol, lowering serum triglycerides, increasing conversion of liver cholesterol into bile acids and increased clearance and metabolic conversion of VLDL and other lipoproteins in the liver.
  • lipid and lipoprotein disorders such as hypercholesterolemia, hypertriglyceridemia, and atherosclerosis
  • said compound and pharmaceutical composition are used for the prophylaxis and/or treatment of diseases where the combined lipid lowering, anti-cholestatic and anti-fibrotic effects of FXR-targeted medicaments can be exploited for the treatment of liver steatosis and associated syndromes such as NASH, or for the treatment of cholestatic and fibrotic effects that are associated with alcohol-induced cirrhosis, or with viral-borne forms of hepatitis.
  • FXR agonists might have clinical utility as anti-atherosclerotic and cardioprotective drugs.
  • the downregulation of Endothelin-1 in Vascular Smooth Muscle Cells might also contribute to such beneficial therapeutic effects (F. He et al., Circ. Res. 2006, 98, 192).
  • the invention also relates to a compound according to Formula (1) or a pharmaceutical composition comprising said compound for preventive and posttraumatic treatment of cardiovascular disorders such as acute myocardial infarction, acute stroke, or thrombosis which occur as an endpoint of chronic obstructive atherosclerosis.
  • cardiovascular disorders such as acute myocardial infarction, acute stroke, or thrombosis which occur as an endpoint of chronic obstructive atherosclerosis.
  • FXR seems to be expressed in breast cancer tissue and cell lines but not in healthy breast tissue and seems to interact with the Estrogen Receptor in ER positive breast cancer cells (K. E. Swales et al., Cancer Res. 2006, 66, 10120 and F. Journe et al., Breast Cancer Res. Treat. 2009, 1 15, 523).
  • FXR also as a potential target for the treatment of proliferative diseases, especially metastasizing cancer forms that express a small molecule responsive form of FXR.
  • said compounds and pharmaceutical compositions are used for the prophylaxis and/or treatment of malignant hyperproliferative disorders such as different forms of cancer, specifically certain forms of breast, liver or colon cancer where interference with an FXR ligand will have a beneficial impact.
  • FXR seems also to be involved in the control of antibacterial defense in the intestine (T. Inagaki et al., PNAS. 2006, 103, 3920) although an exact mechanism is not provided. From these published data, however, one can conclude that treatment with FXR agonists might have a beneficial impact in the therapy of Inflammatory Bowel Disorders (IBD), in particular those forms where the upper (ileal) part of the intestine is affected (e.g. ileal Crohn ' s disease) because this seems to be the site of action of FXR ' s control on bacterial growth. In IBD the desensitization of the adaptive immune response is somehow impaired in the intestinal immune system.
  • IBD Inflammatory Bowel Disorders
  • the invention also relates to a compound according to Formula (1) or a pharmaceutical composition comprising said compound for preventing and/or treating a disease related to Inflammatory Bowel Diseases such as Crohn ' s disease or Colitis ulcerosa.
  • FXR-mediated restoration of intestinal barrier function and reduction in non-commensal bacterial load is believed to be helpful in reducing the exposure of bacterial antigens to the intestinal immune system and can therefore reduce inflammatory responses.
  • the invention further relates to a compound or pharmaceutical composition for the prophylaxis and/or treatment of obesity and associated disorders such as metabolic syndrome (combined conditions of dyslipidemias, diabetes and abnormally high body-mass index) which can be overcome by FXR-mediated lowering of serum triglycerides, blood glucose and increased insulin sensitivity and FXR-mediated weight loss.
  • metabolic syndrome combined conditions of dyslipidemias, diabetes and abnormally high body-mass index
  • the compounds or pharmaceutical composition of the present invention are useful in preventing and/or treating clinical complications of Type I and Type II Diabetes.
  • Such complications include Diabetic Nephropathy, Diabetic Retinopathy, Diabetic Neuropathies, or Peripheral Arterial Occlusive Disease (PAOD).
  • PAOD Peripheral Arterial Occlusive Disease
  • Other clinical complications of Diabetes are also encompassed by the present invention.
  • conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways may also be prevented and/or treated by applying the compounds or pharmaceutical composition of the present invention.
  • Such conditions and diseases encompass NASH and chronic cholestatic conditions in the liver, Glomerulosclerosis and Diabetic Nephropathy in the kidney, Macula Degeneration and Diabetic Retinopathy in the eye and Neurodegenerative diseases such as Alzheimer ' s Disease in the brain, or Diabetic Neuropathies in the peripheral nervous system.
  • the compounds of the present invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed.
  • tablets may be coated by standard aqueous or non-aqueous techniques.
  • Such compositions and preparations should contain at least 0.1 percent of active compound.
  • the percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit.
  • the amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained.
  • the active compounds can also be administered intranasally as, for example, liquid drops or spray.
  • the tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin.
  • a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • tablets may be coated with shellac, sugar or both.
  • a syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
  • the compounds of the present invention may also be used as salts with various countercations to yield an orally available formulation.
  • Such pharmaceutically acceptable cations may be amongst others mono- or bivalent ions such as ammonium, the alkaline metals sodium or potassium or the alkaline earth metals magnesium or calcium, certain pharmaceutically acceptable amines such as tris(hydroxymethyl)aminomethane, ethylendiamine, diethylamine, piperazine or others, or certain cationic amino acids such as lysine or arginine.
  • the compounds of the present invention may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention.
  • oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
  • compounds of the present invention are administered orally.
  • the effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
  • the compounds of the present invention are administered at a daily dosage of from about 0.1 milligram to about 100 milligram per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dosage is from about 1.0 milligrams to about 1000 milligrams, preferably from about 1 milligram to about 50 milligrams.
  • the total daily dose will generally be from about 7 milligrams to about 350 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the compounds of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared.
  • the compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention.
  • the examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds.
  • the instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described above.
  • the amine-free bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogen carbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine-free base into an organic solvent, followed by evaporation.
  • a suitable base such as aqueous sodium hydrogen carbonate, sodium carbonate, sodium hydroxide and potassium hydroxide
  • the amine-free base, isolated in this manner can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallization.
  • the carboxylic free acids corresponding to the isolated salts can be generated by neutralization with a suitable acid, such as aqueous hydrochloric acid, sodium hydrogen sulfate, sodium dihydrogen phosphate, and extraction of the liberated carboxylic-free acid into an organic solvent, followed by evaporation.
  • a suitable acid such as aqueous hydrochloric acid, sodium hydrogen sulfate, sodium dihydrogen phosphate
  • the carboxylic acid, isolated in this manner can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate base and subsequent evaporation, precipitation or crystallization.
  • Step 1 4-((4-Bromo-3-chlorophenoxy)methvn-5-cvclopropyl-3-(2,6-dichlorophenyl)-isoxazole (la)
  • the filtrate was concentrated to give the crude product as a pale yellow oil.
  • Step 3 Methyl 3-(3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)benzoate (1 )
  • A/,AADimethylacetamide (9.0 g, 103 mmol) was dissolved in 1 ,2-dichloroethane (200 mL). The solution was cooled to 0°C before trifluoromethanesulfonic anhydride (63 g, 223 mmol) was added. The reaction was stirred for an additional 60 min at 0°C. Then 1-bromo-3-vinylbenzene (15 g, 81.9 mmol) and 2,4,6-collidine (10.5 g, 86.6 mmol) were added. The reaction was heated to reflux overnight, quenched by addition of water (300 mL) and stirred for 2 hr at rt.
  • Step 2 3-(3-Bromophenyl)-1-(2-chloro-4-((5-cvclopropyl-3- 2,6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)cvclobutanol (2b)
  • Step 3 3-(3-Cvanophenyl)-1 -(2-chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4- vOmethoxy)phenvOcvclobutanol (2c)
  • Step 4 3-((1s.3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)benzoic acid (2)
  • Step 1 3-(3-(Benzylthio)phenyl)-1 -(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)cvclobutanol (5a)
  • Step 2 3-(3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4-yl)methoxy)phenyl)-3- hvdroxycvclobutyl)benzene-1 -sulfonyl chloride (5b)
  • compound 5a 34 mg, 0.05 mmol
  • CH 3 CN/HOAc/H 2 0 1 mL/37 ⁇ _/25 ⁇
  • 2,4-dichloro-5,5-dimethylhydantion (20 mg, 0.1 mmol). The mixture was stirred at 0- 5°C for 2 h.
  • Step 3 3-(3-(2-Chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)methoxy)phenyl)-3- hydroxycvclobutvObenzenesulfonamide (5)
  • Step 3 Methyl 5-f(1s.3s)-3-(2-chloro-4-((5-cvclODropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)- 1 -isopropyl- 1 H-pyrazole-3-carboxylate (7)
  • Example 8 5-((1s,3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1 -isopropyl- 1 H-pyrazole-3-carboxylic acid (8)
  • Ester 7 (98.3 mg, 0.156 mmol) was dissolved in a mixture of THF (7.5 mL), MeOH (2.5 mL) and water (2.5 mL) and LiOH H 2 0 (65 mg, 1.56 mmol) was added at rt. The mixture was stirred at rt for 18 h. The mixture was partitioned between diluted aq. NH 4 CI solution and EA and the organic layer was washed once with water. The combined aq. layer was extracted twice with EA. The combined organic layer was dried (Na 2 S0 4 ), filtered and concentrated to give 103 mg of an almost white solid.
  • Step 5 4-((4-Bromo-3-chlorophenoxy)methyl)-5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazole (8e)
  • Step 6 Ethyl 5-((1s.3s)-3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1-isopropyl-1 H-pyrazole-3-carboxylate (8f)
  • Step 7 5-((1s.3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutylH-isopropyl-1 H-pyrazole-3-carboxylic acid (8)
  • Example 8A 5-((1 r.3r)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenylVisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutylH -isopropyl-1 H-pyrazole-3-carboxylate (8A)
  • Example 8A can be prepared by subjecting the crude product 8f to the ester hydrolysis as described for 8 and isolation from the crude product 8 as a minor isomer by preparative RP- HPLC.
  • ⁇ -NMR (CDCI 3 ), ⁇ (ppm): 7.42-7.30 (m, 2H), 7.11 (d, J 8.0 Hz, 1 H), 6.75-6.65 (m, 1 H), 6.57 (s.
  • Step 3 Methyl 6-(3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1-methyl-1 H-indazole-3-carboxylate (9)
  • Determination of a ligand mediated cofactor peptide interaction to quantify ligand binding to the nuclear receptor FXR was performed as follows: Preparation of human FXR alpha ligand binding domain: The human FXRalpha LBD was expressed in E coli strain BL21(DE3) as an N- terminally GST tagged fusion protein. The DNA encoding the FXR ligand binding domain was cloned into vector pDEST15 (Invitrogen). Expression was under control of an IPTG inducible T7 promoter. The amino acid boundaries of the ligand binding domain were amino acids 187-472 of Database entry NM_005123 (RefSeq).
  • Glutathione 4B sepharose beads were pelleted by centrifugation (2000 x g, 15 sec, 4°C) and washed twice in wash buffer (25 mM Tris, 50 mM KCI, 4 mM MgCI 2 and 1 M NaCI). The pellet was resuspended in 3 mL elution buffer per liter of original culture (elution buffer: 20 mM Tris, 60 mM KCI, 5 mM MgCI 2 and 80 mM glutathione added immediately prior to use as powder). The suspension was left rotating for 15 min at 4°C, the beads pelleted and eluted again with half the volume of elution buffer than the first time.
  • the eluates were pooled and dialysed overnight in 20 mM Hepes buffer (pH 7.5) containing 60 mM KCI, 5 mM MgCI 2 as well as 1 mM dithiothreitol and 10% (v/v) glycerol.
  • the protein was analysed by SDS- Page.
  • the method measures the ability of putative ligands to modulate the interaction between the purified bacterial expressed FXR ligand binding domain (LBD) and a synthetic biotinylated peptide based on residues 676-700 of SRC-1 (LCD2, 676-700).
  • the sequence of the peptide used was B-CPSSHSSLTERHKILHRLLQEGSPS-COOH where the N-terminus was biotinylated (B).
  • the ligand binding domain (LBD) of FXR was expressed as fusion protein with GST in BL- 21 cells using the vector pDEST15. Cells were lysed by sonication, and the fusion proteins purified over glutathione sepharose (Pharmacia) according to the manufacturers instructions.
  • the Perkin Elmer LANCE technology was applied. This method relies on the binding dependent energy transfer from a donor to an acceptor fluorophor attached to the binding partner of interest.
  • This method relies on the binding dependent energy transfer from a donor to an acceptor fluorophor attached to the binding partner of interest.
  • For ease of handling and reduction of background from compound fluorescence LANCE technology makes use of generic fluorophore labels and time resolved detection Assays were done in a final volume of 25 pL in a 384 well plate, in a Tris-based buffer (20 mM Tris-HCI pH 7.5; 60 mM KCI, 5 mM MgCI 2 ; 35 ng/pL BSA), containing 20-60 ng well recombinantly expressed FXR-LBD fused to GST, 200-600 nM N-terminally biotinylated peptide, representing SRC1 aminoacids 676-700, 200 ng/well Streptavidin-xlAPC conjugate(Prozyme)
  • DMSO content of the samples was kept at 1%.
  • the assay was equilibrated for 1 h in the dark at rt in FIA-plates black 384 well (Greiner).
  • the LANCE signal was detected by a Perkin Elmer VICTOR2VTM Multilabel Counter. The results were visualized by plotting the ratio between the emitted light at 665 and 615 nm. A basal level of FXR-peptide formation is observed in the absence of added ligand. Ligands that promote the complex formation induce a concentration-dependent increase in time-resolved fluorescent signal.
  • Mammalian one hybrid (M1H) assay Determination of a ligand mediated Gal4 promoter driven transactivation to quantify ligand binding mediated activation of FXR was performed as follows: The cDNA part encoding the FXR ligand binding domain was cloned into vector pCMV-BD (Stratagene) as a fusion to the yeast GAL4 DNA binding domain under the control of the CMV promoter. The amino acid boundaries of the ligand binding domain were amino acids 187-472 of Database entry NM_005123 (RefSeq).
  • the plasmid pFR-Luc (Stratagene) was used as the reporter plasmid, containing a synthetic promoter with five tandem repeats of the yeast GAL4 binding sites, driving the expression of the Photinus pyralis (American firefly) luciferase gene as the reporter gene.
  • the plasmid pRL-CMV (Promega) was cotransfected.
  • pRL-CMV contains the constitutive CMV promoter, controlling the expression of the Renilla reniformis luciferase.
  • Gal4 reporter gene assays were done in HEK293 cells (obtained from DSMZ, Braunschweig, Germany) grown in MEM with L-Glutamine and Earle's BSS supplemented with 10% fetal bovine serum, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate, and 100 units Penicilin/Streptavidin per ml_ at 37°C in 5% C0 2 . Medium and supplements were obtained from Invitrogen.
  • 5 x 10 5 cells were plated per well in 96well plates in 100 ⁇ _ per well MEM without Phenol Red and L-Glutamine and with Earle's BSS supplemented with 10% charcoal/dextran treated FBS (HyClone, South Logan, Utah), 0.1 mM nonessential amino acids, 2 mM glutamine, 1 mM sodium pyruvate, and 100 units Penicilin/ Streptavidin per mL, incubated at 37 °C in 5% C0 2 . The following day the cells were >90% confluence.
  • OptiMEM polyethylene-imine-based transfection-reagent
  • MEM with the same composition as used for plating cells was added 2-4 h after addition of transfection mixture. Then compound stocks, prediluted in MEM were added (final vehicle concentration not exceeding 0.1%). Cells were incubated for additional 16 h before firefly and renilla luciferase activities were measured sequentially in the same cell extract using a Dual- Light-Luciferase- Assay system (Dyer et al., Anal. Biochem. 2000, 282, 158-161). All experiments were done in triplicates.
  • the aq. solubility in PBS, pH 7.4 was determined as follows. A 10 mM compound stock solution in DMSO was added to PBS (pH 7.4) to reach a theoretical final concentration of 200 ⁇ . The resulting solution/suspension was shaken at 1250 rpm for 1 h and then stored in the dark at rt for 23 h. At this time any precipitate is separated from the solution by centrifugation at 3900 rpm for 30 min. The aq. solubility was determined by comparing the peak area of the principle peak in a calibration standard (200 ⁇ ) in an organic solvent (methanol/water 60:40, v/v) with the peak area of the corresponding peak in the buffer sample. As detection method was used HPLC-UV VIS at 230 nm.
  • Permeation experiments were carried out in a Multiscreen 96 well tray (donor) covered by a 96-well Multiscreen Immobilon (acceptor).
  • the hydrophobic filter material of the Immobilon plate was pre-wetted with 70% ethanol and treated with a solution of lipids (lecithin dissolved in dodecane).
  • the donor plate was filled with test compounds and reference compounds and both plates were inserted into each other and placed onto an orbital shaker for 15 min at 100 rpm.
  • the transport study was started by applying 150 ⁇ _ PBS-buffer containing the test and reference compounds to the donor plate.
  • the contents of the acceptor and donor plate were collected and quantified using LC/MS-detection (test items) or by UV spectroscopy using a Spectramax Plus 384 (Molecular Devices) (reference items).
  • the absorption maxima for the reference items ceftriaxone, guanabenz and carbamazepine were 240 nm, 270 nm and 286 nm, respectively.
  • Recovery samples were prepared as described for the permeation assay samples and were incubated in representative vials during the permeation period under the same conditions.
  • test item samples from the lipid layer were extracted by flushing each well two times with 150 pL EA. The solutions were collected in 1.5 mL reaction tubes and the solvent was evaporated. The dried residues were resuspended in a PBS/DMSO/ACN mixture reflecting the composition of the acceptor and donor samples (i.e. 100 pL buffer supplemented with 5% DMSO, 200 pL ACN+ISTD). The final solvent content of each sample was 66% ACN.
  • Samples from donor and acceptor compartments and calibration standards were precipitated by addition of 200 pL ACN/ISTD or 400 pL ACN/ISTD, respectively. After vigorous shaking (10 seconds) and centrifugation (5 min at 4800 x g, rt), the particle free supernatants were subjected to LC-MS/MS. Membrane compartments were extracted as described above. After reconstitution, samples were vigorously shaken (10 seconds) and spun down (5 min at 4800 x g, rt). The particle free supernatants were subjected to LC-MS/MS.
  • the HPLC system consisted of an Accela U-HPLC pump and an Accela auto sampler (Thermo Fisher Scientific, USA). Mass spectrometry was performed on an Exactive mass spectrometer (orbitrap technology with accurate mass) equipped with an heated electrospray (H-ESI2) interface (Thermo Fisher Scientific, USA) connected to a PC running the standard software Xcalibur 2.1.
  • H-ESI2 heated electrospray
  • the LC was performed in the gradient mode (Table 3) using ACN/0.1% formic acid as organic phase (A) and 10 mM ammonium formate/0.1% formic acid as aq. phase (B); and the pump flow rate was set to 500 pL min. Separation was performed on a Gemini C6-Phenyl, 3 pm, 50x2.0 mm (Phenomenex, Germany) analytical column with a pre-column (Gemini C6-Phenyl, 3 pm, 4x2.0 mm).
  • Analyte was acquired by scanning ⁇ 1 Thomson around the expected mass of the monoisotopic [M+H] + or [M-H] ⁇ ion.
  • the mass resolution of the Orbitrap was set to 50,000.
  • the accurate mass of each analyte was used for peak integration. Further instruments settings were as follows: HCD-Gas off, AGC high dynamic range, max. trap injection time 100 ms, sheath gas 30, aux gas 8, sweep gas 2, spray voltage 4 kV, capillary temperature 250°C, ESI 2 heater temperature 250°C.
  • the objective of the present invention was to generate FXR-agonists with improved physico- chemical properties compared to compounds claimed in WO 201 1/020615. This was achieved by the introduction of a polar hydroxyl group on a 1 ,3-cyclobutylidene or 1 ,3-azetidinylidene group replacing the former 1 ,2-cyclopropylidene ring.
  • aqueous solubility or the PAMPA membrane permeability or both are significantly improved by the introduction of the hydroxy-cyclobutyl or hydroxy-azetidyl moiety.
  • FXR agonists are generally very lipophilic (M. L Crawley, Expert Opin. Ther. Patents 2010, 20, 1047). Therefore, better aqeous solubility and membrane permeability are supposed to result in a higher oral bioavailability and in general in a better suitability for clinical development of those compounds as drugs (L. Huang, J. Dong, S. Karki in Evaluation of drug candidates for preclinical development (Eds. C. Han, C. B. Davis, B. Wang), Wiley & Sons, Hoboken 2010, 187-217).

Abstract

The present invention relates to compounds which bind to the NR1H4 receptor (FXR) and act as agonists of FXR. The invention further relates to the use of the compounds for the preparation of a medicament for the treatment of diseases and/or conditions through binding of said nuclear receptor by said compounds and to a process for the synthesis of said compounds (1). Z is selected from (a), (b), (c) or (d).

Description

Novel FXR (NR1 H4) binding and activity modulating compounds
The present invention relates to compounds which bind to the NR1 H4 receptor (FXR) and act as agonists or modulators of FXR. The invention further relates to the use of the compounds for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds.
Multicellular organisms are dependent on advanced mechanisms of information transfer between cells and body compartments. The information that is transmitted can be highly complex and can result in the alteration of genetic programs involved in cellular differentiation, proliferation, or reproduction. The signals, or hormones, are often low molecular weight molecules, such as peptides, fatty acid, or cholesterol derivatives.
Many of these signals produce their effects by ultimately changing the transcription of specific genes. One well-studied group of proteins that mediate a cell's response to a variety of signals is the family of transcription factors known as nuclear receptors, hereinafter referred to often as "NR". Members of this group include receptors for steroid hormones, vitamin D, ecdysone, cis and trans retinoic acid, thyroid hormone, bile acids, cholesterol-derivatives, fatty acids (and other peroxisomal proliferators), as well as so-called orphan receptors, proteins that are structurally similar to other members of this group, but for which no ligands are known. Orphan receptors may be indicative of unknown signalling pathways in the cell or may be nuclear receptors that function without ligand activation. The activation of transcription by some of these orphan receptors may occur in the absence of an exogenous ligand and/or through signal transduction pathways originating from the cell surface (D. J. Mangelsdorf et al., Cell 1995, 83, 835; R. M. Evans, Mol. Endocrinol. 2005, 19, 1429).
In general, three functional domains have been defined in NRs. An amino terminal domain is believed to have some regulatory function. It is followed by a DNA-binding domain hereinafter referred to as "DBD" which usually comprises two zinc finger elements and recognizes a specific Hormone Responsive Element hereinafter referred to as "HRE" within the promoters of responsive genes. Specific amino acid residues in the "DBD" have been shown to confer DNA sequence binding specificity (M. Schena and K. R. Yamamoto, Science 1988, 241 , 965). A ligand-binding-domain hereinafter referred to as "LBD" is at the carboxy-terminal region of known NRs.
In the absence of hormone, the LBD appears to interfere with the interaction of the DBD with its HRE. Hormone binding seems to result in a conformational change in the NR and thus opens this interference (A. M. Brzozowski et al., Nature 1997, 389, 753). A NR without the LBD constitutively activates transcription but at a low level.
Coactivators or transcriptional activators are proposed to bridge between sequence specific transcription factors, the basal transcription machinery and in addition to influence the chromatin structure of a target cell. Several proteins like SRC-1 , ACTR, and Gripl interact with NRs in a ligand enhanced manner (D. M. Heery et al., Nature 1997, 387, 733; T. Heinzel et al., Nature 1997, 387, 43; K. W. Nettles and G. L. Greene, Annu. Rev. Physiol. 2005, 67, 309).
Nuclear receptor modulators like steroid hormones affect the growth and function of specific cells by binding to intracellular receptors and forming nuclear receptor-ligand complexes. Nuclear receptor-hormone complexes then interact with a HRE in the control region of specific genes and alter specific gene expression (A. Aranda and A. Pascual, Physiol. Rev. 2001 , 81 , 1269).
The Famesoid X Receptor alpha (hereinafter also often referred to as NR1 H4 when referring to the human receptor) is a prototypical type 2 nuclear receptor which activates genes upon binding to promoter region of target genes in a heterodimeric fashion with Retinoid X Receptor (B. M. Forman et al., Cell 1995, 81 , 687). The relevant physiological ligands of NR1 H4 are bile acids (D. J. Parks et al., Science 1999, 284, 1365; M. Makishima et al., Science 1999, 284, 1362). The most potent one is chenodeoxycholic acid (CDCA), which regulates the expression of several genes that participate in bile acid homeostasis. Farnesol and derivatives, together called farnesoids, are originally described to activate the rat orthologue at high concentration but they do not activate the human or mouse receptor. FXR is expressed in the liver, throughout the entire gastrointestinal tract including the esophagus, stomach, duodenum, small intestine, colon, ovary, adrenal gland and kidney. Beyond controlling intracellular gene expression, FXR seems to be also involved in paracrine and endocrine signalling by upregulating the expression of the cytokine Fibroblast Growth Factor 15 (rodents) or 9 (monkeys, humans, J. A. Holt et al., Genes Dev. 2003, 17, 1581 ; T. Inagaki et al., Cell Metab. 2005, 2, 217).
Small molecule compounds which act as FXR modulators have been disclosed in the following publications: WO 2000/037077, WO 2003/015771 , WO 2004/048349, WO 2007/076260, WO 2007/092751 , WO 2007/140174, WO 2007/140183, WO 2008/051942, WO 2008/157270, WO 2009/005998, WO 2009/012125, WO 2008/025539 and WO 2008/025540. Further small molecule FXR modulators have been recently reviewed (M. L. Crawley, Expert Opin Ther. Pat. 2010, 20,1047; D. Merk et al., Future Med. Chem. 2012, 4, 1015).
In WO 2011/020615 we disclosed chiral cyclopropylidene compounds of the following general formula
Figure imgf000003_0001
wherein the variables are defined similar as in this application.
The problem underlying the present invention is to generate FXR-agonists with improved physicochemical properties in general, and reduced hydrophobicity, improved aqueous solubility and better membrane permeability, in particular, compared to compounds claimed in WO 2011/020615.
Said problem has been solved by a compound according to the following Formula (1), an enantiomer, diastereomer, tautomer, solvate, prodrug or pharmaceutical acceptable salt thereof
Figure imgf000004_0001
wherein
R is selected from the group consisting of COORe, CONR7R8, tetrazolyl, SO2NR7R8, C1-6 alkyl, S02-Cve alkyl and H, with R6 independently selected from the group consisting of H or d-e alkyl, and R7 and R8 independently from each other selected from the group consisting of H, alkyl, halo-Ci-6 alkyl, Ci.6 alkylene-R9, S02-C1.6 alkyl, wherein R9 is selected from the group consisting of COOH, OH and S03H;
A is selected from the group consisting of phenyl, pyridyl, pyrimidyl, pyrazolyl, indolyl, thienyl, benzothienyl, indazolyl, benzisoxazolyl, benzofuranyl, benzotriazolyl, furanyl, benzothiazolyl, thiazolyl, oxadiazolyl, each optionally substituted with one or two groups independently selected from the group consisting of OH,
Figure imgf000004_0002
alkyl, O-halo-d-e alkyl, d.6 alkyl, halo-Ci.6 alkyl, C3.6 cycloalkyl and halogen;
Q is selected from the group consisting of phenyl, pyridyl, thiazolyl, thiophenyl, pyrimidyl, each optionally substituted with one or two groups independently selected from the group consisting of d-e alkyl, halo-Ci-6 alkyl, halogen and CF3;
Y is selected from N or CH;
Z is selected from
Figure imgf000004_0003
wherein
X = CH, N, NO;
Ri is selected from the group consisting of hydrogen, d.3 alkyl, C3^ cylcoalkyl, C4-5 alkylcycloalkyl, wherein d.3 alkyl is optionally substituted with 1 to 3 substituents independently selected from halogen, hydroxy or d^ alkoxy;
R2 and R3 are independently selected from the group consisting of hydrogen, C1 -3 alkyl, d.3 haloalkyl, d-3 alkoxy, C1.3 haloalkoxy and halogen. In another embodiment in combination with any of the above or below embodiments, R-A in the compound according to Formula (1) is selected from
Figure imgf000005_0001
In another embodiment in combination with any of the above or below embodiments, Q in the compound according to Formula (1) is
Figure imgf000005_0002
In another embodiment in combination with any of the above or below embodiments, Z in the compound according to Formula (1) is
Figure imgf000005_0003
In another embodiment in combination with any of the above or below embodiments, the compound according to Formula (1) is selected from
Figure imgf000005_0004
-5-
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000007_0002
Figure imgf000007_0003
wherein R is selected from the group consisting of C02H, CONHS02Me, and tetrazolyl. In another embodiment, the present invention is directed to a compound according to Formula (1) for use as a medicament.
In another embodiment, the present invention is directed to a compound according to Formula (1) for use in the prophylaxis and/or treatment of diseases mediated by FXR.
In another embodiment, the present invention is directed to the use of a compound according to Formula (1) for the preparation of a medicament for the prophylaxis and/or treatment of diseases mediated by FXR.
In another embodiment in combination with any of the above or below embodiments, the disease is selected from chronic intrahepatic or some forms of extrahepatic cholestatic conditions; liver fibrosis; obstructive or chronic inflammatory disorders of the liver; liver cirrhosis; liver steatosis and associated syndromes, cholestatic or fibrotic effects that are associated with alcohol-induced cirrhosis or with viral-borne forms of hepatitis; liver failure or liver ischemia after major liver resection; chemotherapy associated steatohepatitis (CASH); acute liver failure; and/or Inflammatory Bowel Diseases.
In another embodiment in combination with any of the above or below embodiments, the disease is selected from lipid and lipoprotein disorders; Type II Diabetes and clinical complications of Type I and Type II Diabetes, including diabetic nephropathy, diabetic neuropathy, diabetic retinopathy and other observed effects of clinically manifest long term Diabetes; conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways, such as Non- Alcoholic Fatty Liver Disease (NAFLD), or Non- Alcoholic Steatohepatitis (NASH); obesity or metabolic syndrome (combined conditions of dyslipidemia, diabetes or abnormally high body-mass index); and/or cute myocardial infarction, acute stroke or thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis. In another embodiment in combination with any of the above or below embodiments, the disease is selected from non-malignant hyperproliferative disorders and malignant hyperproliferative disorders, specifically of hepatocellular carcinoma, colon adenoma and polyposis, colon adenocarcinoma, breast cancer, pancreas adenocarcinoma, Barrett's esophagus or other forms of neoplastic diseases of the gastrointestinal tract and the liver.
The improved physico-chemical properties have been achieved by the introduction of a polar hydroxyl group on a 1 ,3-cyclobutyltdene or 1 ,3-azetidinylidene group replacing the former 1 ,2- cyclopropylidene ring.
Figure imgf000008_0001
Surprisingly, the resulting compounds maintained their activity on the FXR receptor but demonstrated improved physico-chemical properties, such as higher aqueous solubility and/or membrane permeability.
The compounds of the present invention share a common chemical structure according to Formula (1) in claim 1.
In a preferred embodiment in combination with any of the above or below embodiments, the present invention is directed to an enantiomer, diastereomer or pharmaceutically acceptable salt of a compound according to Formula (1).
In a preferred embodiment in combination with any of the above or below embodiments, R in Formula (1) is selected from the group consisting of COOR6, CONR7R8, S02NR7R8, and S02-Ci. 6 alkyl.
In a preferred embodiment in combination with any of the above or below embodiments, R6 in Formula (1) is H.
In a preferred embodiment in combination with any of the above or below embodiments, R7 and R8 in Formula (1) are independently from each other selected from the group consisting of H and S02-Ci.6alkyl.
In a preferred embodiment in combination with any of the above or below embodiments, R7 in Formula (1) is H.
In a preferred embodiment in combination with any of the above or below embodiments, R8 in Formula (1) is SC C^ alkyl.
In a preferred embodiment in combination with any of the above or below embodiments, A is selected from the group consisting of phenyl, pyridyl, pyrimidyl, pyrazolyl, indazolyl, and oxadiazolyl.
In a preferred embodiment in combination with any of the above or below embodiments, A is substituted with one or two groups independently selected from C1.6 alkyl, more preferably C1.3 alkyl. In another preferred embodiment in combination with any of the above or below embodiments, A is unsubstituted.
In a preferred embodiment in combination with any of the above or below embodiments, Q is phenyl.
In a preferred embodiment in combination with any of the above or below embodiments, Q is substituted with one or two groups independently selected from halogen, more preferably one group selected from halogen, in particular CI.
In a preferred embodiment in combination with any of the above or below embodiments, Z is
Figure imgf000010_0001
In a preferred embodiment in combination with any of the above or below embodiments, X = CH.
In a preferred embodiment in combination with any of the above or below embodiments, Ri is C3-6 cylcoalkyl, in particular cyclopropyl.
In a preferred embodiment in combination with any of the above or below embodiments, R2 and R3 are independently selected from halogen, in particular CI.
The compounds of the present invention can be in the form of a prodrug compound. "Prodrug compound" means a derivative that is converted into a compound according to the present invention by a reaction with an enzyme, gastric acid or the like under a physiological condition in the living body, e.g. by oxidation, reduction, hydrolysis or the like, each of which is carried out enzymatically. Examples of the prodrug are compounds, wherein the amino group in a compound of the present invention is acylated, alkylated or phosphorylated to form, e.g., eicosanoylamino, alanylamino, pivaloyloxymethylamino or wherein the hydroxyl group is acylated, alkylated, phosphorylated or converted into the borate, e.g. acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy or wherein the carboxyl group is esterified or amidated. These compounds can be produced from compounds of the present invention according to well-known methods. Other examples of the prodrug are compounds, wherein the carboxylate in a compound of the present invention is, for example, converted into an alkyl-, aryl-, choline-, amino, acyloxymethylester, linolenoylester.
Metabolites of compounds of the present invention are also within the scope of the present invention.
Where tautomerism, like e.g. keto-enol tautomerism, of compounds of the present invention or their prodrugs may occur, the individual forms, like e.g. the keto and enol form, are each within the scope of the invention as well as their mixtures in any ratio. Same applies for stereoisomers, like e.g. enantiomers, cis/trans isomers, conformers and the like.
If desired, isomers can be separated by methods well known in the art, e.g. by liquid chromatography. Same applies for enantiomers by using e.g. chiral stationary phases. Additionally, enantiomers may be isolated by converting them into diastereomers, i.e. coupling with an enantiomerically pure auxiliary compound, subsequent separation of the resulting diastereomers and cleavage of the auxiliary residue. Alternatively, any enantiomer of a compound of the present invention may be obtained from stereoselective synthesis using optically pure starting materials. Another way to obtain pure enantiomers from racemic mixtures would use enantioselective crystallization with chiral counterions.
The compounds of the present invention can be in the form of a pharmaceutically acceptable salt or a solvate. The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic bases or acids and organic bases or acids. In case the compounds of the present invention contain one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts. Thus, the compounds of the present invention which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanotamine or amino acids. The compounds of the present invention which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids. .Examples of suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p- toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the compounds of the present invention simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions). The respective salts can be obtained by customary methods which are known to the person skilled in the art like, for example, by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts. The present invention also includes all salts of the compounds of the present invention which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts.
Further the compounds of the present invention may be present in the form of solvates, such as those which include as solvate water, or pharmaceutically acceptable solvates, such as alcohols, in particular ethanol.
Furthermore, the present invention provides pharmaceutical compositions comprising at least one compound of the present invention, or a prodrug compound thereof, or a pharmaceutically acceptable salt or solvate thereof as active ingredient together with a pharmaceutically acceptable carrier. "Pharmaceutical composition" means one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing at least one compound of the present invention and a pharmaceutically acceptable carrier.
The pharmaceutical composition of the present invention may additionally comprise one or more other compounds as active ingredients like a prodrug compound or other nuclear receptor modulators.
The compositions are suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (nasal or buccal inhalation) or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy.
The compounds of the present invention can be prepared by a combination of methods described in Schemes I to III. As depicted in Scheme I a 4-membered cyclic ketone, substituted with substituent A in the 3-position can react with a metalated aromatic or heteroaromatic ring M-Q-0-CH2Z (M = metal, e.g. Li) in aprotic solvents and preferably at low temperatures to afford a hydroxyl substituted 4-membered ring bearing the substituents A and Q. In the case where Y is CH two isomers can form (A and Q transannular cis or trans to each other). Under optimized conditions the formation of mainly one of the two isomers can be achieved. The two isomers can be separated by appropriate methods known in the art like e.g. silica gel chromatography or preparative RP-HPLC.
Scheme I
Figure imgf000012_0001
In Scheme II the methods are summarized which are used to prepare the 4-membered cyclic ketones needed for the synthesis of the compounds of this invention. In option a) a vinyl bearing intermediate, e.g. prepared by vinylation of a corresponding halogen-containing starting material R-A-X (X = halogen) can react with in situ formed α,α-dichloro ketene to form a 2,2- dichlorocyclobutanone. After dehalogenation, e.g. with Zn in acetic acid under reflux, the desired 3-substituted cyclobutanones are obtained. Alternatively, the vinyl-intermediates can react with in situ generated unsubtituted ketene to afford in one step the desired cyclobutanone intermediates. In option b) 3-methylenecyclobutanecarbonitrile is used as starting material. Substituted heterocycles can be built up from the cyano group in several steps by methods known to those skilled in the art. The desired cyclobutanones can be obtained by oxidative cleavage of the exocyclic double bond using conditions and reagents known to those skilled in the art, e.g. by the use of Os04, ozone or RhCi NalC^ as oxidants. Option c) shows the methods used to prepare the substituted azetidinones. Cu- or Pd-catalysed C-N cross coupling between 3-hydroxy-azetidine and halo-aromatic or halo-heteroaromatic rings afford the corresponding A/-substituted 3-hydroxy-azetidines which can be transformed into the desired azetidinones by oxidation.
Scheme II
b)
Figure imgf000013_0001
HetAr = heteroaromatic ring c) Cu or Pd catalysed
C-N coupling
Figure imgf000013_0002
Scheme III illustrates some possibilities to perform modifications of the substituents at the A group after the formation of the 4-membered hydroxy-bearing rings. For example, a leaving group X (e.g. bromide) can be substituted by a cyano group, a carboxylic ester, methylsulfonyl or thioether by transition metal catalysed cross coupling reactions. The obtained derivatives can be further transformed into other derivatives by methods known to those skilled in the art. For example, the cyano and the ester group can be hydrolysed under basic conditions to the afford a carboxylic acid which in turn can be transformed into acyl-sulfonamides. A benzyl thioether can be chlorinated to afford the chlorosulfonyl intermediate which reacts with ammonia to the corresponding sulfonamides.
Scheme HI
Figure imgf000014_0001
As a result, the present invention relates to compounds according to the general Formula (1) which bind to FXR and act as agonists or modulators of FXR.
The invention further relates to the use of said compounds for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds. Further the present invention relates to the use of said compounds for the preparation of a medicament for the treatment and/or prophylaxis of diseases and/or conditions through binding of said nuclear receptor by said compounds. Specifically, the present invention relates to the use of compounds according to Formula (1) in the preparation of a medicament for the prophylaxis and/or treatment of chronic intrahepatic or some forms of extrahepatic cholestatic conditions, of liver fibrosis, of acute intraheptic cholestatic conditions, of obstructive or chronic inflammatory disorders that arise out of improper bile composition, of gastrointestinal conditions with a reduced uptake of dietary fat and fat-soluble dietary vitamins, of inflammatory bowel diseases, of lipid and lipoprotein disorders, of Type II Diabetes and clinical complications of Type I and Type II Diabetes, of conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways, of obesity and metabolic syndrome (combined conditions of dyslipidemia, diabetes and abnormally high body-mass index), of acute myocardial infarction, of acute stroke, of thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis, of persistant infections by intracellular bacteria or parasitic protozoae, of non-malignant hyperproliferative disorders, of malignant hyperproliferative disorders, of colon adenocarcinoma and hepatocellular carcinoma in particular, of liver steatosis and associated syndromes, of liver failure or liver malfunction as an outcome of chronic liver diseases or of surgical liver resection, of Hepatitis B infection, of Hepatitis C infection and/or of cholestatic and fibrotic effects that are associated with alcohol-induced cirrhosis or with viral- borne forms of hepatitis.
Medicaments as referred to herein may be prepared by conventional processes, including the combination of a compound according to the present invention and a pharmaceutically acceptable carrier. FXR is proposed to be a nuclear bile acid sensor. As a result, it modulates both, the synthetic output of bile acids in the liver and their recycling in the intestine (by regulating bile acid binding proteins). But beyond bile acid physiology, FXR seems to be involved in the regulation of many diverse physiological processes which are relevant in the etiology and for the treatment of diseases as diverse as cholesterol gallstones, metabolic disorders such as Type II Diabetes, dyslipidemias or obesity, chronic inflammatory diseases such as Inflammatory Bowel Diseases or chronic intrahepatic forms of cholestasis and many others diseases (T. Claudel et al., Arterioscler. Thromb. Vase. Biol. 2005, 25, 2020; Y. D. Wang et al., Cell Res. 2008, 18, 1087.
FXR regulates a complex pattern of response genes in the liver and in the gastrointestinal tract. The gene products have impact on diverse physiological processes. In the course of functional analysis of FXR, the first regulatory network that was analyzed was the regulation of bile acid synthesis. While the LXRs induce the key enzyme of the conversion of cholesterol into bile acids, Cyp7A1 , via the induction of the regulatory nuclear receptor LRH-1 , FXR represses the induction of Cyp7A1 via the upregulation of mRNA encoding SHP, a further nuclear receptor that is dominant repressive over LRH-1. Since FXR binds the end products of this pathway, primary bile acids such as cholic acid (CA) or CDCA, this can be regarded as an example of feedback inhibition on the gene expression level (B. Goodwin et al., Mol. Cell 2000, 6, 517; T. T. Lu et al., Mol. Cell 2000, 6, 507). Parallel to the repression of bile acid synthesis via SHP, FXR induces a range of so-called ABC (for ATP-binding cassette) transporters that are responsible for the export of toxic bile acids from the hepatocyte cytosol into the canaliculi, the small bile duct ramifications where the bile originates. This hepatoprotective function of FXR became first apparent with the analysis of FXR knockout mice (C. J. Sinai et al., Cell 2000, 102, 731). where under- or overexpression of several ABC-transporters in the liver was shown. Further detailed analysis revealed that the major bile salt excretory pump BSEP or ABCB1 1 (M. Ananthanarayanan et al., J. Biol. Chem. 2001 , 276, 28857; J. R. Plass et al., Hepatology 2002, 35, 589) as well as the key enzyme which mediates lipid transfer from lipoproteins to phospholipids, PLTP (N. L. Urizar et al., J. Biol. Chem. 2000, 275, 39313), and the two key canalicular membrane transporters for phospholipids, MRP-2 (ABCC4) (H. R. Kast et al., J. Biol. Chem. 2002, 277, 2908) and MDR-3 (ABCB4); L. Huang et al., J. Biol. Chem. 2003, 278, 51085) are direct targets for ligand-directed transcriptional activation by FXR (summarized in: M. Miyata, J. Pharmacol. Exp. Ther. 2005, 312, 759; G. Rizzo et al., Curr. Drug Targets Immune Endocr. Metabol. Disord. 2005, 5, 289).
The fact that FXR seems to be the major metabolite sensor and regulator for the synthesis, export and re-circulation of bile acids suggested the use of FXR ligands to induce bile flow and change bile acid composition towards more hydrophilic composition. With the development of the first synthetic FXR ligand GW4064 (P. R. Maloney et al., J. Med. Chem. 2000, 43, 2971 ; T. M. Willson et al., Med. Res. Rev. 2001 , 21 , 513) as a tool compound and of the semi-synthetic artificial bile acid ligand 6-alpha-ethyl-CDCA, the effects of superstimulation of FXR by potent agonists could be analyzed. It was shown that both ligands induce bile flow in bile duct ligated animals. Moreover, in addition to choleretic effects, also hepatoprotective effects could be demonstrated (R. Pellicciari et al., J. Med. Chem. 2002, 45, 3569; Y. Liu et al., J. Clin. Invest. 2003, 1 12, 1678). This hepatoprotective effect was further narrowed down to an anti-fibrotic effect that results from the repression of Tissue Inhibitors of Matrix-Metalloproteinases, TIMP-1 and 2, the induction of collagen-deposit resolving Matrix- Metalloproteinase 2 in hepatic stellate cells and the subsequent reduction of alpha-collagen mRNA and Transforming growth factor beta (TGF-beta) mRNA which are both pro-fibrotic factors by FXR agonists (S. Fiorucci et al., Gastroenterology 2004, 127, 1497; S. Fiorucci et al., J. Pharmacol. Exp. Ther. 2005, 314, 584). Furthermore, anti-cholestatic activity was demonstrated in bile-duct ligated animal models as well as in animal models of estrogen-induced cholestasis (S. Fiorucci et al., J. Pharmacol. Exp. Ther. 2005, 313, 604).
Genetic studies demonstrate that in hereditary forms of cholestasis (Progressive Familiar Intrahepatic Cholestasis = PFIC, Type I - IV) either nuclear localization of FXR itself is reduced as a consequence of a mutation in the FIC1 gene (in PFIC Type I, also called Byler's Disease) (F. Chen et al., Gastroenterology 2004, 126, 756; L. Alvarez et al., Hum. Mol. Genet. 2004, 13, 2451 ) or levels of the FXR target gene encoding MDR-3 phospholipid export pump are reduced (in PFIC Type III). Taken together there is a growing body of evidence that FXR binding compounds will demonstrate substantial clinical utility in the therapeutic regimen of chronic cholestatic conditions such as Primary Biliary Cirrhosis (PBC) or Primary Sclerosing Cholangitis (PSC) (reviewed in: G. Rizzo et al., Curr. Drug Targets Immune Endocr. Metabol. Disord. 2005, 5, 289; G. Zollner et al., Mol. Pharm. 2006, 3, 231 ; S. Y. Cai et al., Expert Opin. Ther. Targets 2006, 10, 409).
The deep impact that FXR activation has on bile acid metabolism and excretion is not only relevant for cholestatic syndromes but even more directly for a therapy against gallstone formation. Cholesterol gallstones form due to low solubility of cholesterol that is actively pumped out of the liver cell into the lumen of the canaliculi. It is the relative percentage of content of the three major components, bile acids, phospholipids and free cholesterol that determines the formation of mixed micelles and hence apparent solubility of free cholesterol in the bile. FXR polymorphisms map as quantitative trait loci as one factor contributing to gallstone disease (H. Wittenburg, Gastroenterology 2003, 125, 868). Using the synthetic FXR tool compound GW4064 it could be demonstrated that activation of FXR leads to an improvement of the Cholesterol Saturation Index (CSI) and directly to an abolishment of gallstone formation in C57L gallstone susceptible mice whereas drug treatment in FXR knockout mice shows no effect on gallstone formation (A. Moschetta et al., Nature Medicine 2004, 10, 1352).
These results qualify FXR as a good target for the development of small molecule agonists that can be used to prevent cholesterol gallstone formation or to prevent re-formation of gallstones after surgical removal or Shockwave lithotripsy (discussed in: S. A. Doggrell, Curr. Opin. Investig. Drugs 2006, 7, 344).
Thus, in one embodiment of the invention, the compound according to Formula (1) and pharmaceutical compositions comprising said compound is used for the prophylaxis and/or treatment of obstructive or chronic inflammatory disorders that arise out of improper bile composition such as cholelithiasis also known as cholesterol gallstones.
Beyond its strong hepatoprotective and choleretic as well as anti-fibrotic effects that FXR shows upon small molecule stimulated activation in the liver, FXR seems to have a role in protecting the intestine from neoplastic transformation and from the development of polyps and their transition into adenocarcinoma in the gut (S. Modica et al., Cancer Res. 2008, 68, 9589 and R. R. aran et al., J. Pharmacol. Exp. Ther. 2009, 328, 469). Similar to the situation in the intestine absence of FXR leads to a high increase in the formation of Hepatocellular Cacrcinoma (HCC), the most prominent form of liver cancer (I. Kim et al., Carcinogenesis 2007, 28, 940 and F. Yang et al., Cancer Res. 2007, 67, 863). Whereas a functional FXR prevents the formation of colon adenocarcinoma and hepatocellular carcinoma, FXR activation induces liver regeneration after hepatectomy (W. Huang et al., Science 2006, 312, 233).
The combined hepatoprotective, anti-neoplastic and liver regenerative effects associated with FXR activation can be therapeutically exploited for the use of FXR agonists in the treatment of sever liver diseases. In one embodiment, the compounds according to the invention and pharmaceutical compositions comprising said compounds are used in the treatment of liver diseases such as HCC, stimulation of liver regrowth and amelioration of side effects associated with major liver resection, liver cirrhosis independent of the etiology and prevention or treatment of liver ischemia in the course of liver transplantation or major liver surgery.
Since the discovery of the first synthetic FXR agonist and its administration to rodents it became evident that FXR is a key regulator of serum triglycerides (P. Maloney et al., J. Med. Chem. 2000, 43, 2971 ; T. Willson et al., Med. Res. Rev. 2001 , 21 , 513). Over the past six years accumulating evidence has been published that activation of FXR by synthetic agonists leads to significant reduction of serum triglycerides, mainly in the form of reduced VLDL, but also to reduced total serum cholesterol (H. R. Kast et al., Mol. Endocrinol. 2001 , 15, 1720; N. L. Urizar et al., Science 2002, 296, 1703; G. Lambert et al., J. Biol. Chem. 2003, 278, 2563; M. Watanabe et al., J. Clin. Invest. 2004, 113, 1408; A. Figge et al., J. Biol. Chem. 2004, 279, 2790; S. Bilz et al., Am. J. Physiol. Endocrinol. Metab. 2006, 290, E716).
But the lowering of serum triglycerides is not a stand alone effect. Treatment of db/db or ob/ob mice with synthetic FXR agonist GW4064 resulted in marked and combined reduction of serum triglycerides, total cholesterol, free fatty acids, ketone bodies such as 3-OH Butyrate. Moreover, FXR activation engages with the intracellular insulin signaling pathway in hepatocytes, resulting in reduced output of glucose from liver gluconeogenesis but concomitant increase in liver glycogen. Insulin sensitivity as well as glucose tolerance were positively impacted by FXR treatment (K. R. Stayrook et ah, Endocrinology 2005, 146, 984; Y. Zhang et al., PNAS 2006, 103, 1006; B. Cariou et al., J. Biol. Chem. 2006, 281 , 11039; K. Ma et al., J. Clin. Invest. 2006, 116, 1102; D. Duran-Sandoval et al., Biochimie 2005, 87, 93). An effect on reduction of body weight was also recently observed in mice overfed with a high lipid diet (C. Lihong et al., American Diabetes Association (ADA) 66th annual scientific sessions, June 2006, Abstract Number 856-P). This weight loss effect might results from FXR's induction of FGF-19, a fibroblast growth factor that is known to lead to weight loss and athletic phenotype (J. Holt et al., Genes Dev. 2003, 17, 1581 ; E. Tomlinson et al., Endocrinology 2002, 143, 1741). In recent patent applications, the effect of FXR agonist on reduction of body weight was demonstrated (WO 2004/087076; WO 2003/080803).
Taken together, these pharmacological effects of FXR agonists can be exploited in different therapeutic ways: FXR binding compounds are thought to be good candidates for the treatment of Type II Diabetes because of their insulin sensitization, glycogenogenic, and lipid lowering effects.
In one embodiment, the compounds according to the invention and pharmaceutical compositions comprising said compounds are used in the prophylaxis and/or treatment of Type II Diabetes which can be overcome by FXR-mediated upregulation of systemic insulin sensitivity and intracellular insulin signalling in liver, increased peripheral glucose uptake and metabolisation, increased glycogen storage in liver, decreased output of glucose into serum from liver-borne gluconeogenesis.
In a further embodiment, said compounds and pharmaceutical compositions are used for the prophylaxis and/or treatment of chronic intrahepatic, such as PBC, PSC, progressive familiar cholestasis (PFIC), alcohol-induced cirrhosis and associated cholestasis, and some forms of extrahepatic cholestatic conditions, or liver fibrosis.
The invention also relates to a compound of Formula (1) or to a pharmaceutical composition comprising said compound for the prophylaxis and/or treatment of gastrointestinal conditions with a reduced uptake of dietary fat and fat-soluble dietary vitamins which can be overcome by increased intestinal levels of bile acids and phospholipids.
In a further embodiment, said compound or pharmaceutical composition is used for preventing and/or treating a disease selected from the group consisting of lipid and lipoprotein disorders such as hypercholesterolemia, hypertriglyceridemia, and atherosclerosis as a clinically manifest condition which can be ameliorated by FXR's beneficial effect on lowering total plasma cholesterol, lowering serum triglycerides, increasing conversion of liver cholesterol into bile acids and increased clearance and metabolic conversion of VLDL and other lipoproteins in the liver. In one further embodiment, said compound and pharmaceutical composition are used for the prophylaxis and/or treatment of diseases where the combined lipid lowering, anti-cholestatic and anti-fibrotic effects of FXR-targeted medicaments can be exploited for the treatment of liver steatosis and associated syndromes such as NASH, or for the treatment of cholestatic and fibrotic effects that are associated with alcohol-induced cirrhosis, or with viral-borne forms of hepatitis.
In conjunction with the hypolipidemic effects it was also shown that loss of functional FXR leads to increased atherosclerosis in ApoE knockout mice (E. A. Hanniman et al., J. Lipid Res. 2005, 46, 2595). Therefore, FXR agonists might have clinical utility as anti-atherosclerotic and cardioprotective drugs. The downregulation of Endothelin-1 in Vascular Smooth Muscle Cells might also contribute to such beneficial therapeutic effects (F. He et al., Circ. Res. 2006, 98, 192).
The invention also relates to a compound according to Formula (1) or a pharmaceutical composition comprising said compound for preventive and posttraumatic treatment of cardiovascular disorders such as acute myocardial infarction, acute stroke, or thrombosis which occur as an endpoint of chronic obstructive atherosclerosis.
Beyond controlling intestinal and colonic polyp formation, FXR seems to be expressed in breast cancer tissue and cell lines but not in healthy breast tissue and seems to interact with the Estrogen Receptor in ER positive breast cancer cells (K. E. Swales et al., Cancer Res. 2006, 66, 10120 and F. Journe et al., Breast Cancer Res. Treat. 2009, 1 15, 523).
This would allow to regard FXR also as a potential target for the treatment of proliferative diseases, especially metastasizing cancer forms that express a small molecule responsive form of FXR.
In a further embodiment, said compounds and pharmaceutical compositions are used for the prophylaxis and/or treatment of malignant hyperproliferative disorders such as different forms of cancer, specifically certain forms of breast, liver or colon cancer where interference with an FXR ligand will have a beneficial impact.
Finally, FXR seems also to be involved in the control of antibacterial defense in the intestine (T. Inagaki et al., PNAS. 2006, 103, 3920) although an exact mechanism is not provided. From these published data, however, one can conclude that treatment with FXR agonists might have a beneficial impact in the therapy of Inflammatory Bowel Disorders (IBD), in particular those forms where the upper (ileal) part of the intestine is affected (e.g. ileal Crohn 's disease) because this seems to be the site of action of FXR 's control on bacterial growth. In IBD the desensitization of the adaptive immune response is somehow impaired in the intestinal immune system. Bacterial overgrowth might then be the causative trigger towards establishment of a chronic inflammatory response. Hence, dampening of bacterial growth by FXR-borne mechanisms might be a key mechanism to prevent acute inflammatory episodes. Thus, the invention also relates to a compound according to Formula (1) or a pharmaceutical composition comprising said compound for preventing and/or treating a disease related to Inflammatory Bowel Diseases such as Crohn's disease or Colitis ulcerosa. FXR-mediated restoration of intestinal barrier function and reduction in non-commensal bacterial load is believed to be helpful in reducing the exposure of bacterial antigens to the intestinal immune system and can therefore reduce inflammatory responses.
The invention further relates to a compound or pharmaceutical composition for the prophylaxis and/or treatment of obesity and associated disorders such as metabolic syndrome (combined conditions of dyslipidemias, diabetes and abnormally high body-mass index) which can be overcome by FXR-mediated lowering of serum triglycerides, blood glucose and increased insulin sensitivity and FXR-mediated weight loss.
In a further embodiment, the compounds or pharmaceutical composition of the present invention are useful in preventing and/or treating clinical complications of Type I and Type II Diabetes. Examples of such complications include Diabetic Nephropathy, Diabetic Retinopathy, Diabetic Neuropathies, or Peripheral Arterial Occlusive Disease (PAOD). Other clinical complications of Diabetes are also encompassed by the present invention.
Furthermore, conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways may also be prevented and/or treated by applying the compounds or pharmaceutical composition of the present invention. Such conditions and diseases encompass NASH and chronic cholestatic conditions in the liver, Glomerulosclerosis and Diabetic Nephropathy in the kidney, Macula Degeneration and Diabetic Retinopathy in the eye and Neurodegenerative diseases such as Alzheimer's Disease in the brain, or Diabetic Neuropathies in the peripheral nervous system.
In practical use, the compounds of the present invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or non-aqueous techniques. Such compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained. The active compounds can also be administered intranasally as, for example, liquid drops or spray.
The tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin. When a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
Since the compounds of the present invention mostly represent carboxylic acids or similar anionic isosters thereof, and since it is well known that salt forms of ionic drug compounds can substantially affect the bioavailability of drug compounds, the compounds of the present invention may also be used as salts with various countercations to yield an orally available formulation. Such pharmaceutically acceptable cations may be amongst others mono- or bivalent ions such as ammonium, the alkaline metals sodium or potassium or the alkaline earth metals magnesium or calcium, certain pharmaceutically acceptable amines such as tris(hydroxymethyl)aminomethane, ethylendiamine, diethylamine, piperazine or others, or certain cationic amino acids such as lysine or arginine.
The compounds of the present invention may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. Preferably compounds of the present invention are administered orally.
The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
When treating or preventing FXR mediated conditions for which compounds of the present invention are indicated, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.1 milligram to about 100 milligram per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. For most large mammals, the total daily dosage is from about 1.0 milligrams to about 1000 milligrams, preferably from about 1 milligram to about 50 milligrams. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 milligrams to about 350 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
The compounds of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. The examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. The instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described above.
The amine-free bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogen carbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine-free base into an organic solvent, followed by evaporation. The amine-free base, isolated in this manner, can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallization. The carboxylic free acids corresponding to the isolated salts can be generated by neutralization with a suitable acid, such as aqueous hydrochloric acid, sodium hydrogen sulfate, sodium dihydrogen phosphate, and extraction of the liberated carboxylic-free acid into an organic solvent, followed by evaporation. The carboxylic acid, isolated in this manner, can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent, followed by addition of the appropriate base and subsequent evaporation, precipitation or crystallization.
An illustration of the preparation of compounds of the present invention is shown below. Unless otherwise indicated in the schemes, the variables have the same meaning as described above. The examples presented below are intended to illustrate particular embodiments of the invention. Suitable starting materials, building blocks and reagents employed in the synthesis as described below are commercially available from Sigma-Aldrich or Acros Organics, for example, or can be routinely prepared by procedures described in the literature, for example in "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 5th Edition; John Wiley & Sons or T. Eicher, S. Hauptmann "The Chemistry of Heterocycles; Structures, Reactions, Synthesis and Application", 2nd edition, Wiley- VCH 2003; Fieser et al. "Fiesers' Reagents for organic Synthesis" John Wiley & Sons 2000.
Examples
Example 1: Methyl 3-((1s,3s)-3-(2-chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenylVisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)benzoate (1 )
Figure imgf000023_0001
Step 1 : 4-((4-Bromo-3-chlorophenoxy)methvn-5-cvclopropyl-3-(2,6-dichlorophenyl)-isoxazole (la)
To a solution of (5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)methanol (13 g, 45.8 mmol) in CH2CI2 (DCM) (200 mL) was added dropwise SOCI2 (40 mL, 336 mmol). The resulting mixture was stirred at rt for 2 h and the solvents were removed under reduced pressure. The residue was dissolved in Λ ,/V-dimethylformamide (DMF) (200 ml) and 4-bromo-3-chlorophenol (9.7 g, 47 mmol), K2C03 (40 g, 290 mmol) and Nal (12 g, 80 mmol) were added to this solution. The mixture was stirred at 60°C overnight, then cooled to rt, diluted with water (1000 mL) and extracted with ethyl acetate (EA) (500 mL x 3). The combined organic phases were washed with brine (500 mL x 3), dried over Na2S04 and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (CC) to give the title compound 1a (19 g, 88%) as a white solid.
Step 1 : Methyl 3-(2.2-dichloro-3-oxocvclobutyl)benzoate (1 b)
To a 3-necked round bottomed flask, under a nitrogen atmosphere, fitted with a condenser, an overhead stirrer and pressure equalised dropping funnel was dissolved methyl 3-vinylbenzoate (5 g, 31 mmol) in dry Et20 (150 mL). To this flask was added zinc dust (6 g, 3 eq) and the reaction was sonicated for 30 min. After this time a solution of trichloroacetylchloride (8.7 mL, 2.5 eq) in dry Et20 (50mL) was added dropwise whilst continuing the sonication over the next 30 min. During the process the reaction mixture was heated to 35°C. The sonication was continued for 2.5 h at reflux and the reaction appeared to be complete by NMR analysis. The reaction was allowed to cool to rt and quenched with water (-50 mL). This was done in a dropwise manner interspersed sveral times by a few minutes since a delayed exothermic reaction occurred. After 20 min stirring in water the reaction mixture was filtered through a pad of celite and rinsed through with Et20. The organic layer was washed with portions of water (2 x 250 mL), saturated sodium bicarbonate (2 x 250 mL) and brine (1 x 250 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to afford the crude product 1b as a dark yellow thick oil (crude 8.7 g).
Step 2: Methyl 3-(3-oxocvclobutyl)benzoate (1c)
Crude compound 1b (8.7 g) was dissolved in glacial acetic acid (55 mL) in a round bottomed flask under a nitrogen atmosphere. To this flask was added zinc dust (4.6 g, 2.2 eq) and the reaction was stirred and heated to 120°C for 3 h. After cooling to rt the mixture was filtered though a pad of celite, this was washed with portions of EA. The combined solution was concentrated under reduced pressure before being dissolved in EA (500 mL), washed with brine (150 mL x 2) and then dried over sodium sulfate, filtered and concentrated again. The crude mixture was stirred for 5 min in chloroform (250 mL) and filtered through a sintered funnel. The filtrate was concentrated to give the crude product as a pale yellow oil. The crude product was purified by CC in (PE/EA = 9:1 , PE = petroleum ether) to give the desired product 1c (2.5 g, 38% for 2 steps) as a pale yellow oil.
Step 3: Methyl 3-(3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)benzoate (1 )
To a stirring solution of compound 1a (1.67 g, 3.5 mmol) in dry THF (30 mL) was added n-BuLi (2.5 M in hexane, 1.2 eq, 1.69 mL) dropwise over 10 min at -78°C under a nitrogen atmosphere. This was stirred for 1 h at this temperature before adding a solution of compound 1c (0.72 g, 1 eq) in dry THF (10 mL) dropwise and stirred for 1 h at this temperature. The reaction mixture was allowed to warm to rt slowly and left stirring overnight. The reaction was quenched with a solution of saturated ammonium chloride solution (50 mL) and EA (250 mL). The organic layer was separated and the aq. layer was washed with EA (2 x 100 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated to give the crude product as a brown oil. The product was isolated following CC with PE/EA (19:1 to 3:1). The reaction and purification was repeated twice on the same scale and the combined product (3.13 g) was repurified under the same conditions to afford the final product 1 (1.7 g, 19%). 1H NMR (CDCI3): 7.93 (m, 1H), 7.90-7.85 (m, 1 H), 7.50-7.30 (m, 5H), 6.88 (s, 1H), 6.75-6.72 (m, 1H), 4.80 (s, 2H), 3.88 (s, 3H), 3.20-3.10 (m, 1H), 3.00-2.91 (m, 2H), 2.60-2.49 (m, 2H), 2.15- 2.08 (m, 1 H), 1.30-1.25 (m, 2H), 1.15-1.10 (m, 2H).
Example 2: 3-((1 s.3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4- yl)methoxytohenv0-3-hvdroxycvclobutyl}benzoic acid (2)
Figure imgf000025_0001
Compound 1 (1.7 g, 2.84 mmol) was dissolved in THF (100 mL) at rt. A solution of LiOH (285 mg, 4.2 eq) in water (20 mL) was added and the solution was stirred and warmed to 35°C for three days. After this time the THF was removed under reduced pressure. The remaining aq. solution was diluted with water (25 mL) and washed with Et20 (2 x 50 mL). The aq. layer was then transferred to a round bottomed flask and acidified to pH 6 using 1 N HCI. The formed white precipitated was filtered off and dried under reduced pressure at 50°C to give title compound 2 (1.3 g, 78%, single isomer by 1H-NMR and LC-MS) as white solid. Ή NMR (400 MHz, CD3OD) δ: 7.98 (s, 1 H), 7.86 (d, J = 7.6 Hz, H), 7.58-7.46 (m, 5H), 7.41 (t, J = 7.6 Hz, 1 H), 6.91 (d, J = 2.4 Hz, 1H), 6.80 (dd, J = 8.8, 2.4 Hz, 1H), 4.95 (s, 2H), 3.29-3.25 (m, 2H), 2.96 (m, 1 H), 2.55- 2.49 (m, 2H), 2.37 (m, 1H), 1.24-1.22 (m, 4H). MS (ESI") m/z: 584 (582) [M-1]'.
Relevant intensive NOEs (obtained from the ROESY spectra; arrows below) indicate that the two aromatic moieties are 1 ,3-trans oriented in Example 2.
Figure imgf000026_0001
Alternative route to Example 2
Step 1 : 3-(3-Bromophenyl)cvclobutanone (2a)
A/,AADimethylacetamide (9.0 g, 103 mmol) was dissolved in 1 ,2-dichloroethane (200 mL). The solution was cooled to 0°C before trifluoromethanesulfonic anhydride (63 g, 223 mmol) was added. The reaction was stirred for an additional 60 min at 0°C. Then 1-bromo-3-vinylbenzene (15 g, 81.9 mmol) and 2,4,6-collidine (10.5 g, 86.6 mmol) were added. The reaction was heated to reflux overnight, quenched by addition of water (300 mL) and stirred for 2 hr at rt. The mixture was extracted with DCM (300 mL x 3). The combined organic layers were dried over Na2S04 and concentrated in vacuo. Purification by CC (EA PE = 1 :20) gave the title compound 2a (5.0 g, 27%) as a pale yellow solid.
Step 2: 3-(3-Bromophenyl)-1-(2-chloro-4-((5-cvclopropyl-3- 2,6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)cvclobutanol (2b)
To a solution of compound 1a (14 g, 29.6 mmol) in dry THF (500 ml) at -78°C was added dropwise n-BuLi (18.5 mL, 1.6 M in hexane, 29.6 mmol). The mixture was stirred for an additional 1 h at -78°C and a solution of compound 2a (6.5 g, 28.9 mmol) in dry THF (50 mL) was added dropwise. The resulting mixture was stirred at -78°C for 1 h and then warmed to rt and quenched with saturated aq. NH4CI (500 mL). The mixture was extracted with EA (500 mL x 2), the combined organic layers were washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by CC (EA/PE = 1 :5) to give the title compound 2b (6.5 g, 37%) as a white solid.
Step 3: 3-(3-Cvanophenyl)-1 -(2-chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4- vOmethoxy)phenvOcvclobutanol (2c)
To a solution of compound 2b (3.1 g, 5 mmol) in D F (50 mL) were added under argon atmosphere Zn(CN)2 (500 mg, 4.3 mmol), Pd2(dba)3 (300 mg, 0. 33 mmol) and Xantphos (150 mg, 0.31 mmol). The mixture was stirred for 10 h at 115°C under microwave irradiation. After cooling to rt the reaction mixture was diluted with water (250 mL) and extracted with EA (250 mL x 2). The combined organic layers were washed with brine (100 mL x 3) and dried over Na2S04. The residue was purified by CC (EA/PE) to give the title compound 2c (1.2 g, 42%) as a pale yellow solid.
Step 4: 3-((1s.3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)benzoic acid (2)
To a solution of compound 2c (15 g, 24.2 mmol) in EtOH (750 ml_) was added aq. NaOH (40 g in 100 mL of water). The resulting mixture was heated to reflux overnight and then cooled to rt. The reaction was concentrated in vacuo to remove the volatile solvent, diluted with water (1000 mL) and the pH was adjusted to 2 with diluted aq. HCI (1 N). The formed precipitate was collected by filtration to give the crude product as a yellow solid (13.8 g). Purification by preparative preversed phase HPLC (RP-HPLC) afforded the title compound 2 (8.0 g, 56%, single isomer by 1H-NMR) as a white solid.
Preparative Example 3
Figure imgf000027_0001
Step 1 : Methyl 3-(3-hvdroxyazetidin-1-yl)benzoate (3a)
To a solution of methyl 3-iodobenzoate (4.5 g, 17.2 mmol) in DMSO (30 mL) was added 3- azetidin-3-ol hydrogen chloride salt (1.3 g, 11.8 mmol), Cs2C03 (9.5 g, 29.2 mmol), Cul (446 mg, 2.3 mmol) and L-proline (540 mg, 4.7 mmol) and then the mixture was heated at 90°C for 18 h under argon atmosphere. The solution was diluted with EA and water and the organic layer was washed with brine three times, concentrated under reduced pressure and purified by CC (PE/EA = 2:1) to give compound 3a (1.6 g, 66%) as a yellow solid.
Step 2: Methyl 3-(3-oxoazetidin-1-yl)benzoate (3)
To a solution of compound 3a (1.60 g, 7.7 mmol) in dry DCM (30 mL) was added Dess-Martin periodinane (6.5 g, 15.4 mmol) at 0°C and the mixture was stirred at rt for 2 h under N2 atmosphere. The mixture was quenched with saturated sodium bicarbonate solution and diluted with EA. The organic portion was washed with brine, dried over Na2S04, filtered, concentrated under reduced pressure and purified by CC (PE EA = 4:1) to give compound 3 (1.2 g, 75%) as a white solid.
Example 4: 3-((1s,3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutvn-N-(methylsulfonyl)benzamide (4¾
Figure imgf000028_0001
To the solution of compound 2 (100 mg, 0.17 mmol) in DCM (5 mL) were added EDCI HCI (100 mg, 0.52 mmol), DMAP (100 mg, 0.81 mmol) and MeS02NH2 (40 mg, 0.42 mmol). The mixture was stirred at 30°C overnight and then diluted with EA and washed by H20, brine and dried over Na2S04. Concentration in vacuo and purification by prep-TLC gave crude target compound as a light yellow solid. RP-HPLC purification afforded the title compound 4 (38 mg, 33%) as a white solid. 1H NMR (400 MHz, CD3OD) δ: 7.87 (s, 1 H), 7.74 (d, J = 7.6 Hz, 1 H), 7.61-7.53 (m, 4H), 7.50-7.46 (m, 2H), 6.91 (d, J = 2.4 Hz, 1 H), 6.80 (dd, J = 8.8, 2.4 Hz, 1H), 4.95 (s, 2H), 3.38(s, 3H), 3.30-3.26 (m, 2H), 3.01 (m, 1 H), 2.57-2.51 (m, 2H), 2.37 (m, 1 H)( 1.25-1.23 (m, 4H). MS (ESP) m/z: 659 [Μ-1Γ·
Example 5: 3-(3-(2-Chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobuM)benzenesulfonamide (5)
Figure imgf000028_0002
Step 1 : 3-(3-(Benzylthio)phenyl)-1 -(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)cvclobutanol (5a)
To a solution of compound 2b (619 mg, 1 mmol) in toluene (20 mL) under argon atmosphere were added K2C03 (276 mg, 2 mmol), phenylmethanethiol (125 mg, 1 mmol), Pd2(dba)3 (200 mg, 0. 22 mmol) and Xantphos (75 mg, 0.16 mmol). Then the mixture was stirred at 115°C for 4 h. After being cooled to rt, the reaction was diluted with water (100 mL) and extracted with EA (100 mL x 2). The combined organic layers were washed with brine (100 mL x 2), dried over Na2S0 and concentrated to dryness. Purification by CC gave compound the compound 5a (200 mg; 30%) as a pale yellow solid. 1H NMR (400 MHz, CDCI3) δ: 7.36-7.32 (m, 3H), 7.28- 7.07 (m, 9H), 7.01 (d, J = 7.2 Hz, 1 H), 6.82 (d, J = 2.0 Hz, 1H), 6.66 (dd, J = 8.8, 2.0 Hz, 1 H), 4.75 (s, 2H), 4.04 (s, 2H), 3.06-3.00 (m, 2H), 2.84-2.78 (m, 2H), 2.44-2.38 (m, 3H), 2.09 (m, 1 H), 1.24-1.18 (m, 2H), 1.11-1.08 (m, 2H). MS (ESI+) m/z: 662 [M+1]+.
Step 2: 3-(3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4-yl)methoxy)phenyl)-3- hvdroxycvclobutyl)benzene-1 -sulfonyl chloride (5b) To a solution of compound 5a (34 mg, 0.05 mmol) in CH3CN/HOAc/H20 (1 mL/37 μΙ_/25 μί) was added 2,4-dichloro-5,5-dimethylhydantion (20 mg, 0.1 mmol). The mixture was stirred at 0- 5°C for 2 h. The reaction was diluted with water and extracted with CH2CI2. The combined organic layers were washed with a 5% NaHC03 solution, brine and dried over Na2S04. Concentration to dryness afforded the crude product 5b (30 mg) as a colorless oil, which was used directly in the next step.
Step 3: 3-(3-(2-Chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)methoxy)phenyl)-3- hydroxycvclobutvObenzenesulfonamide (5)
To the solution of compound 5b (30 mg) in CH3CN (2 mL) was added NH4OH (0.3 mL). The mixture was stirred at rt for 1 h. Concentration to dryness and purification by prep. RP-HPLC gave the title compound 5 (3.5 mg, 10% for two steps) as a white solid. 1H NMR (400 MHz, CDCI3) δ: 7.85 (s, 1 H), 7.77 (d, J = 7.6 Hz, 1 H), 7.54-7.41 (m, 5H), 7.35 (d, J = 8.4 Hz, 1 H), 6.90 (s, 1H), 6.75 (d, J = 8.4 Hz, 1 H), 4.83 (s, 2H), 4.77 (s, broad, 2H), 3.20 (t, J = 10.4 Hz, 2H), 3.04 (m, 1 H), 2.58 (t, J = 10.6 Hz, 2H), 2.17 (m, 1H), 1.31-1.30 (m, 2H), 1.20-1.16 (m, 2H). MS (ESP) m/z: 617 [M-1 ]-.
Example 6: 1 -(2-Chloro-4-((5-cvclopropyl-3-i2.6-dichlorophenyl)isoxazol-4-yl)methoxy)phenyl)- 3-(3-(methylsulfonyl)phenyl)cvclobutanol (6)
Figure imgf000029_0001
To the solution of compound 2b (200 mg, 0.32 mmol) in DMSO, sodium methanesulfinate (50 mg, 0.46 mmol), Cul (20 mg, 0.1 mmol), L-proline (37 mg, 0.32 mmol) and diisopropylethylamine (DIEA) (41 mg, 0.32 mmol) was added. The mixture was stirred at 95°C overnight and then diluted with water and extracted with EA. The combined organic layers were washed with water and dried over Na2S04. Concentration to dryness under reduced pressure and purification by prep. RP-HPLC gave the title compound 6 as a white solid (35 mg, 21%, single isomer by 'H NMR and LC-MS). 1H NMR (400 MHz, CDCI3) δ: 7.84 (s, 1H), 7.79 (d, J = 7.6 Hz, 1 H), 7.60 (d, J = 7.6 Hz, 1 H), 7.53 (t, J = 7.6 Hz, 1 H), 7.44-7.41 (m, 3H), 7.34 (t, J = 7.2 Hz, 1 H), 6.90 (d, J = 2.8 Hz, 1 H), 6.75 (dd, J = 8.4, 2.0 Hz, 1 H), 4.83 (s, 2H), 3.24-3.19 (m, 2H), 3.08-3.04 (m, 4H), 2.62-2.56 (m, 2H), 2.17 (m, 1H), 1.31-1.29 (m, 2H), 1.20-1.16 (m, 2H). MS (ESI+) m/z: 618 (620) [M+1]+, 600 (602) [M-H20+1]+. Example 7: Methyl 5-((1s.3sV3-(2-chloro-4-((5-cvcloproDyl-3-(2,6-clichlorophenvnisoxazol-4- yl)methoxy)phenvn-3-hvdroxycvclobutyl)-1 -isopropyl-1 H-Dyrazole-3-carboxylate (7)
Figure imgf000030_0001
Step 1 : Methyl 1-isopropyl-5-vinyl-1 H-pyrazole-3-carboxylate (7a)
A suspension of methyltriphenylphosphonium bromide (2.69 g, 7.52 mmol) in dry THF (40 mL) was cooled to -78°C and n-butyllithium (1.6 M solution in hexane, 3.7 mL, 5.91 mmol) was added dropwise. The yellow-orange suspension was stirred at -78°C for 50 min and then a solution of methyl 5-formyl-1-isopropyl-1 H-pyrazole-3-carboxylate (prepared as described in WO 2011/020615, 1.05 g, 5.37 mmol) in dry THF (10 mL) was added dropwise. The mixture was stirred at -78°C for 1.75 h, the cooling bath was removed and the mixture (off-white suspension) was stirred at rt for 1 h. The mixture was then partitioned between diluted aq. NaHC03 solution (150 mL) and EA (150 mL). The aq. layer was extracted twice with EA (50 mL each) and the combined organic layer was washed twice with water (50 mL each) and concentrated without drying to give 2.74 g of a yellow oil which slowly crystallized. The crude product was purified by CC (preadsorption with CH2Cl2> hexane/EA 4:1) to give alkene 7a (590 mg, 57%) as a colorless oil. 1H NMR (DMSO-d6) δ: 7.02 (s, 1 H), 6.87 (dd, J = 17.3, 1 1.2 Hz, 1 H), 5.94 (dd, J = 17.3, 1.3 Hz, 1 H), 5.45 (dd, J = 11.2, 1.3 Hz, 1H), 4.80 (sept, J = 6.6 Hz, 1 H), 3.79 (s, 3H), 1.38 (d, J = 6.6 Hz, 6H). C10H14N2O2 (194.23). LC-MS (ESI): 195 [M+H]+.
Step 2: Methyl 1-isopropyl-5-(3-oxocvclobutvn-1 H-pyrazole-3-carboxylate (7b)
The reaction was performed in two dry sealed tubes (two batches of equal quantity). The batches were combined for workup and purification. Single batch procedure: To a solution of A ,A/-dimethylacetamide (0.22 mL, 2.34 mmol) in 1 ,2-dichloroethane (12 mL) under nitrogen at - 15 to -20°C was added dropwise trifluoromethanesulfonic anhydride (0.43 mL, 2.57 mmol), forming an opaque suspension. The mixture was stirred at -15°C for 10 min, and a solution of alkene 7a (151 mg, 0.78 mmol) and sym.-collidine (0.42 mL, 3.12 mmol) in 1 ,2-dichloroethane (3 mL) was added dropwise (yellow solution formed). Upon completion of the addition the cooling was bath removed, the mixture was allowed to warm to rt (orange turbid solution) and the tube was sealed. The mixture was then stirred at 90°C for 15 h (brown mixtures). Water (5 mL) was added at rt and the mixtures were stirred at 100°C for 2 h (turbid two-phase solutions). After cooling to rt, the mixtures were combined and partitioned between diluted aq. NaHC03 solution and CH2CI2 and the aq. layer was extracted three times with CH2CI2 (30 mL each). The combined organic layer was dried (Na2S04), filtered and concentrated to give a brown oil (2.2 g). Purification by CC (6x13 cm, preadsorption with CH2CI2, toluene/EA 3:1) gave cyclobutanone 7b (1 15.5 mg, 31%) as a yellow oil. Ή NMR (DMSO-d6) δ: 6.81 (s, 1 H), 4.58 (sept, J = 6.5 Hz, 1H), 3.78 (s, 3H), 3.85-3.73 (m, 1 H), 3.59-3.45 (m, 2H), 3.37-3.24 (m, 2H, partially overlapped by water signal), 1.39 (d, J = 6.6 Hz, 6H). C,2H16N203 (236.27). LC-MS (ESI): 237 [M+H]+.
Step 3: Methyl 5-f(1s.3s)-3-(2-chloro-4-((5-cvclODropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)- 1 -isopropyl- 1 H-pyrazole-3-carboxylate (7)
A solution of bromide 1a (368 mg, 0.78 mmol) in dry THF (6 mL) was cooled to -78°C and a 1.6M n-butyllithium solution in hexanes (0.48 mL, 0.76 mmol) was added dropwise. The mixture was stirred at -78°C for 20 min and a solution of cyclobutanone 7b (164 mg, 0.69 mmol) in dry THF (4 mL) was added dropwise. The mixture was stirred at -78°C for 2.5 h and saturated aq. NH4CI solution (1 mL) was added dropwise at this temperature. The cooling bath was removed and the mixture was allowed to warm to rt and stirred at rt for 0.5 h. The mixture was then added to diluted aq. NH4CI solution and extracted three times with EA. The combined organic layer was dried (Na2S04), filtered and concentrated to give 516 mg of an almost colorless oil. Purification by CC (4.5x23 cm, preadsorption with CH2CI2, eluent hexane/acetone = 2:1) afforded recovered cyclobutanone 7b (31.3 mg, 19%, slightly yellow oil) and impure product (333 mg). Repurification by CC (4x22 cm, hexane/EA = 1:1) or prep-TLC gave pure product 7 (210 mg, 48%) as white foam. 1H NMR (DMSO-d6) δ: 7.65 (d, J = 2.1 Hz, 1 H), 7.62 (s, 1 H), 7.59-7.48 (m, 2H), 6.92 (d, J = 2.4 Hz, 1 H), 6.76 (dd, J = 8.6, 2.6 Hz, 1 H), 6.66 (s, 1H), 5.49 (s, 1H), 4.92 (s, 2H), 4.42 (quint-like m, J = 6.5 Hz, 1 H), 3.78 (s, 3H), 3.24-3.11 (m, 2H, partially overlapped by water signal), 3.04-2.90 (m, 1 H), 2.54-2.33 (m, 3H, partially overlapped by DMSO signal), 1.32 (d, J = 6.5 Hz, 6H), 1.26-1.08 (m, 4H). C31H3oCl3 305 (630.95). LC-MS (ESI): 630, 632 [M+H]+.
Example 8: 5-((1s,3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1 -isopropyl- 1 H-pyrazole-3-carboxylic acid (8)
Figure imgf000031_0001
Ester 7 (98.3 mg, 0.156 mmol) was dissolved in a mixture of THF (7.5 mL), MeOH (2.5 mL) and water (2.5 mL) and LiOH H20 (65 mg, 1.56 mmol) was added at rt. The mixture was stirred at rt for 18 h. The mixture was partitioned between diluted aq. NH4CI solution and EA and the organic layer was washed once with water. The combined aq. layer was extracted twice with EA. The combined organic layer was dried (Na2S04), filtered and concentrated to give 103 mg of an almost white solid. The product was purified by CC (3x3.5 cm, EA/EtOH = 10:1 to 1 :4) to afford 8 (94.8 mg, 99%) as a white solid. 1H NMR (DMSO-de) δ: 7.66-7.60 (m, 1 H), 7.62 (s, 1 H), 7.59-7.49 (m, 2H), 6.91 (d, J = 2.5 Hz, 1H), 6.76 (dd, J = 8.6, 2.4 Hz, 1H), 6.38 (s, 1H), 5.51 (s, 1H, exchangeable with D20), 4.92 (s, 2H), 4.31 (quint-like m, J = 6.5 Hz, 1 H), 3.25-3.08 (m, 2H, partially overlapped by water signal), 2.93-2.77 (m, 1H), 2.57-2.43 (m, 1 H, hidden by DMSO signal), 2.43-2.29 (m, 2H, partially overlapped by DMSO signal), 1.29 (d, J = 6.5 Hz, 6H), 1.26- 1.08 (m, 4H). The C02H signal does not appear in the spectrum. C3oH28Cl3 305 (616.92). LC- MS (ESI): 616, 618 [M+H]+.
Alternative route to Example 8
Step 1 : 1-(3-Methylenecvclobutyl)ethanone (8a)
Methylene cyclobutane carbonitrile (5.0 g, 53.7 mmol) was dissolved in dry diethyiether (25 mL), cooled in an ice bath and MeMgBr (26.8 mL, 80.5 mmol, 3 M in ether) was added dropwise. The mixture was left stirring overnight at rt, cooled to 0°C, quenched carefully with 5% NaHS04 aq. sol. (100 mL). The mixture was stirred at rt for 30 min. and the layers were separated. The aq. phase was extracted with pentane (50 mL) and diethyiether (50 mL). The combined organic layers were washed with brine and dried over Na2S04. The solvents were removed under vacuum at rt and the crude product was obtained as a yellowish liquid.
Step 2: Ethyl 4-(3-methylenecyclobutyl)-2,4-dioxobutanoate (8b)
Sodium (1.15 g, 49.9 mmol) was dissolved in dry EtOH (30 mL, denaturated with 5% diethyiether). Compound 8a (5.5 g, 49.9 mmol, crude) was dissolved in dry EtOH (45 mL) and the above prepared sodium ethoxide solution was added. This mixture was stirred at rt for 15 min and then diethyl oxalate (6.8 mL, 49.9 mmol) was added dropwise. The reaction mixture was placed in a pre-heated (to 67 °C) oil bath and stirred at this temperature for 4.5 h. The mixture was left at rt overnight. The solvent was removed, EA (100 mL) and 1 M HCI (70 mL) were added and organic phase was separated. The aq. phase was re-extracted with EA (50 mL). The combined organic phases were washed with water, brine and dried over anh. Na2S04. The solvent was removed under reduced pressure and the residue was purified on silica using hexanes/MTBE 9:1 as eluent giving pure product 8b. Yield: 6.29 g, 56% over two steps. 1H- NMR (CDCI3), δ (ppm): 6.36 (s, 1 H), 4.85-4.80 (m, 2H), 4.34 (q, J = 8.0 Hz, 2H), 3.35-3.25 (m, 1 H), 3.05-2.85 (m, 4H), 1.36 (t, J = 8.0 Hz, 3H).
Step 3: Ethyl 1-isopropyl-5-(3-methylenecyclobutyl)-1 H-pyrazole-3-carboxylate (8c)
Compound 8b (6.29 g, 29.9 mmol) was dissolved in dry EtOH (65 mL, denaturated with 5% of MeOH) and isopropyl hydrazine hydrochloride (3.97 g, 35.9 mmol) was added. The reaction mixture was stirred for 3 h at rt. The solvent was removed and to the oily residue were added EA (100 mL), water (50 mL) and sat. NaHC03 (50 mL) sequentially. The layers were separated and the aq. phase was re-extracted with EA (50 mL). The combined organic phases were washed with brine (70 mL) and dried over anh. Na2S04. The solvent was removed under vacuum and the residue was dried under reduced pressure. Yield: 7.23 g (contains 3.4% of EtOAc by NMR, recalculated pure yield: 6.98 g, 94%). Crude product 8c is 98% pure by HPLC and NMR. 1H-NMR (CDCI3), δ (ppm): 6.62 (s, 1 H), 4.88-4.82 (m, 2H), 4.42-4.32 (m, 3H), 3.56- 3.45 (m, 1 H), 3.17-3.07 (m, 2H), 2.88-2.79 (m, 2H), 1.49 (d, J = 8.0 Hz, 6H), 1.37 (t, J = 8.0 Hz, 3H).
Step 4: Ethyl 1-isopropyl-5-(3-oxocyclobutyl)-1 H-pyrazole-3-carboxylate (8d)
Compound 8c (6.45 g, 26.0 mmol) was dissolved in a mixture of MeCN (77 mL) and water (13 mL) and cooled in an ice-bath. To this solution RuCI3xH20 (0.19 g, 0.86 mmol) was added, followed by portion-wise addition of Nal04 (19.35 g, 90.9 mmol). An exotherm was observed during this addition. The obtained thick slurry was stirred at rt for 45 min. The reaction mixture was diluted with Na2S203 aq. sol. (10%, 260 mL), water (50 mL) and DCM (100 mL). The phases were separated and the aq. phase was extracted with DCM (2x70 mL). The combined organic phases were washed with Na2S203 aq. sol. (10%, 50 mL), water (100 mL), brine (100 mL) and dried over anh. Na2S04.The crude product (6.5 g) was purified on silica, eluting with hexanes/MTBE to give pure product as an oil that solidified upon storage at -20°C. Yield: 5.8 g (78% over two steps). Ή-NMR (DMSO-d6), δ (ppm): 6.78 (s, 1H), 4.57 (h, J = 8.0 Hz, 1H), 4.26 (q, J = 8.0 Hz, 2H), 3.85-3.75 (m, 1 H), 3.58-3.45 (m, 2H), 3.35-3.25 (m, 2H), 1.39 (d, J = 8.0 Hz, 6H), 1.28 (t, J = 8.0 Hz, 3H).
Step 5: 4-((4-Bromo-3-chlorophenoxy)methyl)-5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazole (8e)
3-Chloro-4-bromophenol (3.8 g, 18.3 mmol) was mixed with (5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methanol (3.47 g, 12.2 mmol) and triphenylphosphine (6.41 g, 24.4 mmol) in toluene (150 mL). The mixture was cooled in an ice-bath and DIAD (4.8 mL, 24.4 mmol) as a solution in toluene (10 mL) was added drop-wise. The reaction was stirred at rt for 21 h and the solvents were removed on a rotavap leaving a yellow oily residue. This was dissolved in DCM (200 mL), silica (-20 g) was added and the mixture was evaporated to dryness. This material was loaded on the top of a silica column and purified eluting with hexanes/MTBE 9:1. The product containing fractions were pooled and the solvent removed under reduced pressure, leaving pure product 8e as a colourless oil that crystallized upon drying under vacuum overnight. Yield: 5.07 g (88%).'H-NMR (CDCI3), δ (ppm): 7.45-7.30 (m, 4H), 6.90 (s, 1H), 6.60-6.55 (m, 1 H), 2.15-2.07 (m, 1 H), 1.32-1.25 (m, 2H), 1.20-1.11 (m, 2H).
Step 6: Ethyl 5-((1s.3s)-3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenvnisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1-isopropyl-1 H-pyrazole-3-carboxylate (8f)
LiCI (0.684 g, 16.15 mmol) was dissolved in THF (20 mL) at rt and /PrMgCI (2.0 M in THF, 8.1 mL, 16.15 mmol) was added. The mixture was stirred for 10 min at rt, cooled in an ice-bath and a solution of compound 8e (2.55 g, 5.38 mmol) in THF (20 mL) was added over 5 min. The cooling bath was removed and the mixture was stirred at rt for 4 h. The mixture was cooled to - 10°C and a solution of compound 8d (1.48 g, 5.92 mmol) in THF (16 mL) was added rapidly. The mixture was stirred at rt for 90 min. and then 0.5 M NaHS0 aq. (35 mL) and EA (50 mL) were added. The resulting mixture was stirred for 10 min., the layers were separated and the aq. layer was extracted with EA (30 mL). The combined organic phases were washed with NaHC03 aq. (50 mL), brine (50 mL) and dried over anh. Na2S04. The crude product (3.79 g) was obtained after removal of the solvent as a white foam. 3.6 g of this crude was purified on silica column, eluting with hexanes/EA 3:2 to give pure product 8f as a solid foam. Yield: 1.62 g (49%). 1H-NMR (DMSO-d6), δ (ppm): 7.65-7.47 (m, 4H), 6.93-6.91 (m, 1 H), 6.79-6.72 (m, 1 H), 6.65 (s, 1 H), 5.48 (s, 1 H), 4.92 (s, 2H), 4.42 (h, J = 8.0 Hz, 1 H), 4.26 (q, J = 8.0 Hz, 2H), 3.32 (s, 2H), 3.22-3.14 (m, 2H), 3.05-2.90 (m, 1 H), 2.45-2.35 (m, 2H), 1.35-1.10 (m, 14H).
Step 7: 5-((1s.3s)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutylH-isopropyl-1 H-pyrazole-3-carboxylic acid (8)
Compound 8f (1.60 g, 2.48 mmol) was dissolved in THF (100 mL), then MeOH (50 mL), water (50 mL) and LiOHxH20 (1.04 g, 24.8 mmol) were added sequentially. The mixture was stirred for 4.5 h at rt and then concentrated under reduced pressure to remove MeOH and THF. The remaining aq. solution was acidified by addition of 1 M HCI aq. (24 mL) to reach pH of 4.05 (pH electrode control). Already at approx. pH 7 a precipitate started to form. The formed solid was filtered off, washed on the filter with water and dried under vacuum at rt to give product 8 as a white powder. Yield: 1.40 g (92%). 1H-NMR (CDCI3), δ (ppm): 7.44-7.32 (m, 4H), 6.91 (d, J = 4.0 Hz, 1 H), 6.78 (s, 1 H), 6.75 (dd, J = 4.0 Hz, J = 8.0 Hz, 1 H), 4.83 (s, 2H), 4.35-4.20 (m, 1 H), 3.25-3.14 (m, 2H), 3.04-2.90 (m, 1 H), 2.62-2.54 (m, 2H), 2.21-2.11 (m, 1 H), 1.46 (d, J = 8.0 Hz, 6H), 1.34-1.28 (m, 2H), 1.20-1.14 (m, 2H). 13C-NMR (CDCI3), δ (ppm): 172.7, 164.8, 159.2, 158.4, 147.2, 141.3, 135.8, 134.1 , 132.8, 131.3, 128.1 , 127.6, 127.3, 117.7, 113.3, 1 10.0, 106.3, 73.1 , 59.8, 51.1 , 41.7, 22.6, 22.0, 8.5, 7.8. MS (ESI+) m/z: 616.4 [M+1]+.
Example 8A: 5-((1 r.3r)-3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenylVisoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutylH -isopropyl-1 H-pyrazole-3-carboxylate (8A)
Figure imgf000034_0001
Example 8A can be prepared by subjecting the crude product 8f to the ester hydrolysis as described for 8 and isolation from the crude product 8 as a minor isomer by preparative RP- HPLC. Ή-NMR (CDCI3), δ (ppm): 7.42-7.30 (m, 2H), 7.11 (d, J = 8.0 Hz, 1 H), 6.75-6.65 (m, 1 H), 6.57 (s. 1 H), 4.79 (s, 2H), 4.50-4.41 (m, 1 H), 3.96-3.85 (m, 1 H), 2.98-2.90 (m, 2H), 2.67-2.57 (m, 2H), 2.20-2.09 (m, 1 H), 1.51 (d, J = 8.0 Hz, 6H), 1.32-1.14 (m, 4H). 13C-NMR (CDCI3), δ (ppm): 172.6, 166.2, 159.2, 158.4, 147.4, 141.2, 135.7, 134.6, 132.8, 131.3, 128.1 , 127.7, 127.5, 116.8, 1 13.5, 1 10.0, 105.8, 75.1 , 59.8, 51.2, 41.8, 25.4, 22.6, 8.5, 7.8. MS (ESI+) m/z: 616.3 [M+1 ]+.
The transannular configuration of the major isomer (compound 8) and the minor isomer (compound 8A) was confirmed by NOE experiments. The detected indicative NOEs between protons are indicated in the following pictures by double arrows:
Figure imgf000035_0001
NOEs detected for example 8 with 1,3-trans transannular configuration of the aromatic moieties
Figure imgf000035_0002
NOEs detected for example 8A with 1,3-cis transannular configuration of the aromatic moieties
Example 9: Methyl 6-(3-(2-chloro-4-((5-cvclopropyl-3-(2,6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutv0-1 -methyl- 1 Wndazole-3-carpoxylate (9)
Figure imgf000036_0001
Step 1 : Methyl 1-methyl-6-vinyl-1 H-indazole-3-carboxylate (9a)
To the solution of methyl 6-bromo- 1 -methyl- 1 H-indazole-3-carboxylate (60 mg, 0.22 mmol) in DMF (10 mL), tributyl(vinyl)tin (99 pL, 0.34 mmol), Pd(Ph3)4 (11 mg, 9 pmol) was added. After the addition was completed, the mixture was stirred at 90°C for 4 h under Ar. Then the solvent was removed under reduced pressure. Purification by CC afforded compound 9a (52 mg, 88%).
Step 2: Methyl 1-methyl-6-(3-oxocvclobutyl)-1 H-indazole-3-carboxylate (9b)
Following the procedure as described in Example 7/Step 2, compound 9b was obtained from 9a in 57% yield. 1H NMR (400 MHz, CDCI3) δ: 8.14 (d, J = 8.4 Hz, 1 H), 7.31 (s, 1H), 7.23 (d, J = 8.8 Hz, 1H), 4.13 (s, 3H), 3.99 (s, 3H), 3.87-3.79 (m, 1 H), 3.58-3.51 (m, 2H), 3.33-3.26 (m, 2H). m/z: 259 [M+1]+.
Step 3: Methyl 6-(3-(2-chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-1-methyl-1 H-indazole-3-carboxylate (9)
Following the procedure as described in Example 7/Step 3, compound 9 was obtained from 9b in 40% yield.
Example 0: 6-(3-(2-Chloro-4-((5-cvclopropyl-3-(2.6-dichlorophenyl)isoxazol-4- yl)methoxy)phenyl)-3-hvdroxycvclobutyl)-l-methyl-1 H-indazole-3-carboxylic acid (10)
Figure imgf000036_0002
Following the procedure as described in Example 8, compound 10 was obtained from compound 9 in 45% yield as a white solid.1H NMR (400 MHz, CDCI3) δ: 8.14 (d, J = 8.0 Hz, 1 H), 7.48 (d, J = 8.8 Hz, 1 H), 7.43-7.32 (m, 4H), 7.29 (m, 1 H), 6.92 (d, J = 2.4 Hz, 1 H), 6.76 (dd, J = 7.2 Hz, 2.4 Hz, 1 H), 4.84 (s, 2H), 4.18 (s, 3H), 3.45-3.40 (m, 1H), 3.28-3.23 (m, 2H), 3.19-3.10 (m, 1 H), 2.68-2.63 (m, 2H), 2.21-2.14 (m, 1 H), 1.33-1.29 (m, 2H), 1.20-1.15 (m, 2H). m/z: 638 [M+1]+. Preparative Example 11
Figure imgf000037_0001
Stepl : Methyl 5-(3-hvdroxyazetidin-1-yl)nicotinate (11a)
A mixture of methyl 5-bromonicotinate (2.00 g, 9.26 mmol), azetidin-3-ol (1.01 g, 9.26 mmol), Cs2CO3 (9.06 g, 27.8 mmol), BINAP (1.15 g, 1.85 mmol) and Pd(OAc)2 (0.44 g, 1.85 mmol) in dry dioxane (1 15 mL) was heated overnight at 85°C under N2 atmosphere. The resulting mixture was filtrated, concentrated under reduced pressure and purified by prep-HPLC to give compound 11a (250 mg, 13%) of as a yellow solid.
Step 2: Methyl 5-(3-oxoazetidin-1-yl)nicotinate (11)
To a solution of compound 11a (250 mg, 1.20 mmol) in dry DCM (15 mL) was added Dess- Martin periodinane (1.014 g, 2.40 mmol) at 0°C under N2 atmosphere and the solution was stirred at rt for 2 h. The resulting solution was quenched with saturated sodium bicarbonate solution and diluted with ΕΞΑ. The organic portion was washed with brine, dried over Na2S04, filtered, concentrated under reduced pressure and purified by CC (DCM/MeOH = 150:1) to give compound 11 (140 mg, 57%) of as a yellow solid.
Preparative Example 12
Using a similar procedure as that described in Preparative Example 11 the following compound has been prepared:
Figure imgf000037_0002
12
Example 13/1 to 13/9
The following table lists further examples prepared according the above mentioned preparative examples and examples. All listed compounds were prepared as single isomers.
# Structure Analytical data Analytical data
Hz, DMSO-d6) δ: 1.15-1.25 (m, 4H), H, partially under solvent signal), H), 3.1 1-3.20 (m, 2H), 4.93 (s, 2H), H), 6, 93 (s, 1H), 7.40-7.51 (m, 2H), H), 7.62-7.66 (m, 2H), 7.85-7.90 (m, .: 583; MS Found: 584 [M+H]+. Hz, CD3OD) δ: 1.05-1.12 (m, 4H), H), 2.62-2.71 (m, 1H), 2.99-3.09 (m, H), 6.60-6.64 (m, 1H), 6.72-6.78 (m, H), 7.30-7.42 (m, 4H). MS Calcd.: d: 612 [M+Hf. Hz, CD3OD) δ: 1.10-1.23 (m, 4H), H), 3.00-3.12 (m, 1 H), 3.15-3.25 (m, H), 4.95 (s, 2H), 6.72-6.80 (m, 1 H), .90-7.00 (m, 1H), 7.42-7.60 (m, 4H), 1 H), 7.98 (s, 1H). MS Calcd.: 613; MS -H]". Hz, CD3OD) δ: 1.20-1.30 (m, 4H), .30-2.55 (m, 3H), 2.98-3.10 (m, 1 H), 2H, partially under solvent signal), .76-6.84 (m, 1 H), 6.91 (s, 1 H), 7.20- .43-7.63 (m, 4H), 7.75-7.82 (m, 1 H), S Calcd.: 597; MS Found: 596 [M-H]" MHz, CD3OD) δ: 1.20-1.25 (m, 4H), 4H), 2.46-2.56 (m, 2H), 2.88-2.97 (m, (m, 2H), 4.94 (s, 2H), 6.78-6.82 (m, H), 7.37 (s, 1 H), 7.43-7.60 (m, 4H), .78 (s, 1 H). MS Calcd.: 597; MS -H]". MHz, CD3OD) δ: 1.17-1.23 (m, 4H), 4H), 2.42-2.50 (m, 2H), 2.83-2.92 (m, (m, 2H), 4.92 (s, 2H), 6.74-6.80 (m, H), 7.19-7.22 (m, 1 H), 7.43-7.57 (m, H). MS Calcd.: 597; MS Found: 598
MHz, CD3OD) δ: 1.09-1.1 1 (m, 4H), 1H), 4.08-4.10 (m, 2H), 4.19-4.21 (m, H), 6.64-6.67 (m, 2H), 6.75 (s, 1 H), 2H), 7.28-7.39 (m, 6H); MS Calcd.: d: 585 (M+1).
Figure imgf000038_0001
Structure Analytical data
13/8
13/9
Figure imgf000039_0001
1H); M Calcd.: 585; MS oun : 586 (M+1).
Example 14/1 and 14/2
Using a similar procedure as described in the Examples 1 to 13 and Schemes above, the following compounds were obtained by using the appropriate building blocks.
Structure Analytical data
1H NMR (400 MHz, DMSO-d6): δ 1.13-1.23 (m, 4H),
1.33 (d, J = 6.4 Hz, 6H), 2.37-2.47 (m, 3H), 2.90- 2.95 (m, 1 H), 3.14-3.19 (t, J = 8.8 Hz, 2H), 3.57 (d, J = 4.0 Hz, 2H), 4.38 (m, 1H), 4.92 (s, 2H), 5.51 (s, 1 H ), 6.51 (s, 1 H ), 6.76 (dd, J = 2.4 Hz, J = 8.4 Hz, 1 H), 6.91 (d, J = 2.4 Hz, 1 H), 7.51-7.58 (m, 2H),
Figure imgf000039_0002
7.62-7.65 (m, 2H), 7.69 (s, 1 H); MS Calcd.: 672; MS
Found: 673 [M+Hf.
,
Figure imgf000039_0003
1H NMR (400 MHz, DMSO-d6): δ 1.10-1.25 (m, 4H),
1.34 (d, J = 6.4 Hz, 6H), 3.03-2.95 (m, 1 H), 2.50- 2.30 (m, 3H), 3.20-3.10 (m, 2H), 4.50-4.35 (m, 1 H), 4.92 (s, 2H), 5.5 (s, 1 H), 6.92 (s. 1 H), 6.78-6.70 (m, 2H), 7.70-7.49 (m, 4H), 11.44 (s, 1H); MS Calcd.: 694; MS Found: 695 [M+H]\
Figure imgf000039_0004
The following compound can be prepared in the same manner by using similar procedures as described above:
Figure imgf000040_0001
Assays
FRET activity assay
Determination of a ligand mediated cofactor peptide interaction to quantify ligand binding to the nuclear receptor FXR was performed as follows: Preparation of human FXR alpha ligand binding domain: The human FXRalpha LBD was expressed in E coli strain BL21(DE3) as an N- terminally GST tagged fusion protein. The DNA encoding the FXR ligand binding domain was cloned into vector pDEST15 (Invitrogen). Expression was under control of an IPTG inducible T7 promoter. The amino acid boundaries of the ligand binding domain were amino acids 187-472 of Database entry NM_005123 (RefSeq). Expression and purification of the FXR-LBD: An overnight preculture of a transformed E.coli strain was diluted 1 :20 in LB-Ampicillin medium and grown at 30°C to an optical density of OD60o=0.4-0.6. Gene expression was then induced by addition of 0.5 mM IPTG. Cells were incubated an additional 6 h at 30°C, 180 rpm. Cells were collected by centrifugation (7000 x g, 7 min, rt). Per liter of original cell culture, cells were resuspended in 10 mL lysis buffer (50 mM Glucose, 50 mM Tris pH 7.9, 1 mM EDTA and 4 mg/mL lysozyme) and left on ice for 30 min. Cells were then subjected to sonication and cell debris removed via centrifugation (22000 x g, 30 min, 4°C). Per 10 mL of supernatant 0.5 mL prewashed Glutathione 4B sepharose slurry (Qiagen) was added and the suspension kept slowly rotating for 1 h at 4°C. Glutathione 4B sepharose beads were pelleted by centrifugation (2000 x g, 15 sec, 4°C) and washed twice in wash buffer (25 mM Tris, 50 mM KCI, 4 mM MgCI2 and 1 M NaCI). The pellet was resuspended in 3 mL elution buffer per liter of original culture (elution buffer: 20 mM Tris, 60 mM KCI, 5 mM MgCI2 and 80 mM glutathione added immediately prior to use as powder). The suspension was left rotating for 15 min at 4°C, the beads pelleted and eluted again with half the volume of elution buffer than the first time. The eluates were pooled and dialysed overnight in 20 mM Hepes buffer (pH 7.5) containing 60 mM KCI, 5 mM MgCI2 as well as 1 mM dithiothreitol and 10% (v/v) glycerol. The protein was analysed by SDS- Page.
The method measures the ability of putative ligands to modulate the interaction between the purified bacterial expressed FXR ligand binding domain (LBD) and a synthetic biotinylated peptide based on residues 676-700 of SRC-1 (LCD2, 676-700). The sequence of the peptide used was B-CPSSHSSLTERHKILHRLLQEGSPS-COOH where the N-terminus was biotinylated (B). The ligand binding domain (LBD) of FXR was expressed as fusion protein with GST in BL- 21 cells using the vector pDEST15. Cells were lysed by sonication, and the fusion proteins purified over glutathione sepharose (Pharmacia) according to the manufacturers instructions. For screening of compounds for their influence on the FXR-peptide interaction, the Perkin Elmer LANCE technology was applied. This method relies on the binding dependent energy transfer from a donor to an acceptor fluorophor attached to the binding partner of interest. For ease of handling and reduction of background from compound fluorescence LANCE technology makes use of generic fluorophore labels and time resolved detection Assays were done in a final volume of 25 pL in a 384 well plate, in a Tris-based buffer (20 mM Tris-HCI pH 7.5; 60 mM KCI, 5 mM MgCI2; 35 ng/pL BSA), containing 20-60 ng well recombinantly expressed FXR-LBD fused to GST, 200-600 nM N-terminally biotinylated peptide, representing SRC1 aminoacids 676-700, 200 ng/well Streptavidin-xlAPC conjugate(Prozyme) and 6-10 ng/well Eu W1024 - antiGST (Perkin Elmer). DMSO content of the samples was kept at 1%. After generation of the assay mix and diluting the potentially FXR modulating ligands, the assay was equilibrated for 1 h in the dark at rt in FIA-plates black 384 well (Greiner). The LANCE signal was detected by a Perkin Elmer VICTOR2VTM Multilabel Counter. The results were visualized by plotting the ratio between the emitted light at 665 and 615 nm. A basal level of FXR-peptide formation is observed in the absence of added ligand. Ligands that promote the complex formation induce a concentration-dependent increase in time-resolved fluorescent signal. Compounds which bind equally well to both monomeric FXR and to the FXR-peptide complex would be expected to give no change in signal, whereas ligands which bind preferentially to the monomeric receptor would be expected to induce a concentration-dependent decrease in the observed signal.
To assess the inhibitory potential of the compounds, EC50-values were determined for example compounds as listed below in Table 1 (A = EC50 < 25 nM; B = 25≤ EC50 < 100 nM; C = EC50≥ 100 nM).
Table 1
Figure imgf000041_0001
Mammalian one hybrid (M1H) assay Determination of a ligand mediated Gal4 promoter driven transactivation to quantify ligand binding mediated activation of FXR was performed as follows: The cDNA part encoding the FXR ligand binding domain was cloned into vector pCMV-BD (Stratagene) as a fusion to the yeast GAL4 DNA binding domain under the control of the CMV promoter. The amino acid boundaries of the ligand binding domain were amino acids 187-472 of Database entry NM_005123 (RefSeq). The plasmid pFR-Luc (Stratagene) was used as the reporter plasmid, containing a synthetic promoter with five tandem repeats of the yeast GAL4 binding sites, driving the expression of the Photinus pyralis (American firefly) luciferase gene as the reporter gene. In order to improve experimental accuracy the plasmid pRL-CMV (Promega) was cotransfected. pRL-CMV contains the constitutive CMV promoter, controlling the expression of the Renilla reniformis luciferase. All Gal4 reporter gene assays were done in HEK293 cells (obtained from DSMZ, Braunschweig, Germany) grown in MEM with L-Glutamine and Earle's BSS supplemented with 10% fetal bovine serum, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate, and 100 units Penicilin/Streptavidin per ml_ at 37°C in 5% C02. Medium and supplements were obtained from Invitrogen. For the assay, 5 x 105 cells were plated per well in 96well plates in 100 μΐ_ per well MEM without Phenol Red and L-Glutamine and with Earle's BSS supplemented with 10% charcoal/dextran treated FBS (HyClone, South Logan, Utah), 0.1 mM nonessential amino acids, 2 mM glutamine, 1 mM sodium pyruvate, and 100 units Penicilin/ Streptavidin per mL, incubated at 37 °C in 5% C02. The following day the cells were >90% confluence. Medium was removed and cells were transiently transfected using 20 μΙ_ per well of a OptiMEM - polyethylene-imine-based transfection-reagent (OptiMEM, Invitrogen; Polyethyleneimine, Aldrich Cat No. 40,827-7) including the three plasmids described above. MEM with the same composition as used for plating cells was added 2-4 h after addition of transfection mixture. Then compound stocks, prediluted in MEM were added (final vehicle concentration not exceeding 0.1%). Cells were incubated for additional 16 h before firefly and renilla luciferase activities were measured sequentially in the same cell extract using a Dual- Light-Luciferase- Assay system (Dyer et al., Anal. Biochem. 2000, 282, 158-161). All experiments were done in triplicates.
To assess the FXR agonistic potency of the example compounds, potency ranges were determined in the M H assay as listed below in Table 2 (A = EC50 < 25 nM; B = 25≤ EC50 < 00 nM; C = EC50≥ 100 nM).
Table 2
Group Example #
A 13/4, 13/5, 13/6
B 2, 8, 8A, 10, 13/1 , 13/3, 13/7
Figure imgf000043_0001
Aqueous solubility assay
The aq. solubility in PBS, pH 7.4 was determined as follows. A 10 mM compound stock solution in DMSO was added to PBS (pH 7.4) to reach a theoretical final concentration of 200 μΜ. The resulting solution/suspension was shaken at 1250 rpm for 1 h and then stored in the dark at rt for 23 h. At this time any precipitate is separated from the solution by centrifugation at 3900 rpm for 30 min. The aq. solubility was determined by comparing the peak area of the principle peak in a calibration standard (200 μΜ) in an organic solvent (methanol/water 60:40, v/v) with the peak area of the corresponding peak in the buffer sample. As detection method was used HPLC-UV VIS at 230 nm.
Parallel Artificial Membrane Permeation Assay (PAMPA)
For the PAMPA, 5 mM stock solutions of test items were prepared in DMSO. 5 mM stock solutions of reference items were prepared in EtOH (carbamazepine, guanabenz) or in EtOH:H20 1 :1 (v/v) (ceftriaxone), respectively. Compounds were diluted in PBS (pH 7.4) to obtain the starting solutions containing 5% of the respective organic solvent and 250 μΜ reference compounds or 10 μΜ test items, respectively. For the assay, a modified procedure of the PAMPA as described by Kansy et al. Kansy et al. (J. Med. Chem. 1998, 41 , 1007) was used. The reference compounds for low (ceftriaxone), medium (guanabenz) and high permeation (carbamazepine) were included as internal controls.
Permeation experiments were carried out in a Multiscreen 96 well tray (donor) covered by a 96-well Multiscreen Immobilon (acceptor). The hydrophobic filter material of the Immobilon plate was pre-wetted with 70% ethanol and treated with a solution of lipids (lecithin dissolved in dodecane). The donor plate was filled with test compounds and reference compounds and both plates were inserted into each other and placed onto an orbital shaker for 15 min at 100 rpm. The transport study was started by applying 150 μΐ_ PBS-buffer containing the test and reference compounds to the donor plate. After 15 - 16 h of diffusion at rt, the contents of the acceptor and donor plate were collected and quantified using LC/MS-detection (test items) or by UV spectroscopy using a Spectramax Plus384 (Molecular Devices) (reference items). The absorption maxima for the reference items ceftriaxone, guanabenz and carbamazepine were 240 nm, 270 nm and 286 nm, respectively. Recovery samples were prepared as described for the permeation assay samples and were incubated in representative vials during the permeation period under the same conditions.
For LC/MS analysis of the test items, 100 pL incubate were removed from acceptor and donor compartment and processed for acetonitrile (ACN) precipitation as described below. Additionally, test item samples from the lipid layer were extracted by flushing each well two times with 150 pL EA. The solutions were collected in 1.5 mL reaction tubes and the solvent was evaporated. The dried residues were resuspended in a PBS/DMSO/ACN mixture reflecting the composition of the acceptor and donor samples (i.e. 100 pL buffer supplemented with 5% DMSO, 200 pL ACN+ISTD). The final solvent content of each sample was 66% ACN.
Samples from donor and acceptor compartments and calibration standards were precipitated by addition of 200 pL ACN/ISTD or 400 pL ACN/ISTD, respectively. After vigorous shaking (10 seconds) and centrifugation (5 min at 4800 x g, rt), the particle free supernatants were subjected to LC-MS/MS. Membrane compartments were extracted as described above. After reconstitution, samples were vigorously shaken (10 seconds) and spun down (5 min at 4800 x g, rt). The particle free supernatants were subjected to LC-MS/MS.
For analysis of compounds under the present invention, the HPLC system consisted of an Accela U-HPLC pump and an Accela auto sampler (Thermo Fisher Scientific, USA). Mass spectrometry was performed on an Exactive mass spectrometer (orbitrap technology with accurate mass) equipped with an heated electrospray (H-ESI2) interface (Thermo Fisher Scientific, USA) connected to a PC running the standard software Xcalibur 2.1.
The LC was performed in the gradient mode (Table 3) using ACN/0.1% formic acid as organic phase (A) and 10 mM ammonium formate/0.1% formic acid as aq. phase (B); and the pump flow rate was set to 500 pL min. Separation was performed on a Gemini C6-Phenyl, 3 pm, 50x2.0 mm (Phenomenex, Germany) analytical column with a pre-column (Gemini C6-Phenyl, 3 pm, 4x2.0 mm).
Table 3: HPLC gradients
Figure imgf000044_0001
As MS tune file a generic tune file was used for all analytes applying the positive or negative ion mode. As lock mass for internal mass calibration the [M+H]+ ion of diisooctyl phthalate(m/z 391.28429), which is ubiquitously present in the solvent system, was used.
Analyte was acquired by scanning ±1 Thomson around the expected mass of the monoisotopic [M+H]+ or [M-H]~ ion. The mass resolution of the Orbitrap was set to 50,000. The accurate mass of each analyte was used for peak integration. Further instruments settings were as follows: HCD-Gas off, AGC high dynamic range, max. trap injection time 100 ms, sheath gas 30, aux gas 8, sweep gas 2, spray voltage 4 kV, capillary temperature 250°C, ESI 2 heater temperature 250°C. The objective of the present invention was to generate FXR-agonists with improved physico- chemical properties compared to compounds claimed in WO 201 1/020615. This was achieved by the introduction of a polar hydroxyl group on a 1 ,3-cyclobutylidene or 1 ,3-azetidinylidene group replacing the former 1 ,2-cyclopropylidene ring.
Figure imgf000045_0001
Surprisingly, the resulting compounds maintained their activity on the FXR receptor but demonstrated improved physico-chemical properties, such as higher aq. solubility and/or membrane permeability. A direct comparison of the corresponding compounds of the two series is given in Table 4.
Table 4
Aqueous PAMPA, clogD
Structure solubility Membrane permeability (ChemAxon re)
Figure imgf000045_0002
Aqueous PAMPA, c!ogD
Structure solubility Membrane permeability (ChemAxon re)
Figure imgf000046_0001
* Flux (%) = (c acceptor well) / sum (c donor well + c acceptor well) x 100 x 2
** n.d. = not determined
In each case either the aqueous solubility or the PAMPA membrane permeability or both are significantly improved by the introduction of the hydroxy-cyclobutyl or hydroxy-azetidyl moiety. As most nuclear receptor active molecules, FXR agonists are generally very lipophilic (M. L Crawley, Expert Opin. Ther. Patents 2010, 20, 1047). Therefore, better aqeous solubility and membrane permeability are supposed to result in a higher oral bioavailability and in general in a better suitability for clinical development of those compounds as drugs (L. Huang, J. Dong, S. Karki in Evaluation of drug candidates for preclinical development (Eds. C. Han, C. B. Davis, B. Wang), Wiley & Sons, Hoboken 2010, 187-217).

Claims

Claims
1. A compound according to the following Formula (1), an enantiomer, diastereomer, tautomer, solvate, prodrug or pharmaceutical acceptable salt thereof
Figure imgf000047_0001
wherein
R is selected from the group consisting of COOR6> CONR7Ra, tetrazolyl, S02NR7R8, C^.6 alkyl, S02-C -6 alkyl and H, with R6 independently selected from the group consisting of H or Ci.6 alkyl, and R7 and R8 independently from each other selected from the group consisting of H, C^e alkyl, halo-Ci-6 alkyl, Ci.6 alkylene-R9, S02-Ci.6 alkyl, wherein R9 is selected from the group consisting of COOH, OH and S03H;
A is selected from the group consisting of phenyl, pyridyi, pyrimidyl, pyrazolyl, indolyl, thienyl, benzothienyl, indazolyl, benzisoxazolyl, benzofuranyl, benzotriazolyl, furanyl, benzothiazolyl, thiazolyl, oxadiazolyl, each optionally substituted with one or two groups independently selected from the group consisting of OH, 0-Ci.6 alkyl, 0-halo-Ci-6 alkyl, Ci.6 alkyl, halo-C^ alkyl, C3-6 cycloalkyl and halogen;
Q is selected from the group consisting of phenyl, pyridyi, thiazolyl, thiophenyl, pyrimidyl, each optionally substituted with one or two groups independently selected from the group consisting of C1-6alkyl, halo-C1-6 alkyl, halogen and CF3;
Y is selected from N or CH;
Z is selected from
Figure imgf000047_0002
wherein
X = CH, N, NO;
Ri is selected from the group consisting of hydrogen, C1-3 alkyl, C3.6 cylcoalkyl, C4.5 alkylcycloalkyl, wherein Ci_3 alkyl is optionally substituted with 1 to 3 substituents independently selected from halogen, hydroxy or
Figure imgf000047_0003
R2 and R3 are independently selected from the group consisting of hydrogen, d.3 alkyl, C .3 haloalkyl, Ci-3 alkoxy, Ci-3 haloalkoxy and halogen.
2. The compound according to claim 1 wherein R-A is selected from
Figure imgf000048_0001
3. The compound according to claim 1 or 2 wherein Q is
Figure imgf000048_0002
4. The compound according to any of claims 1 to 3 wherein Z is
Figure imgf000048_0003
Figure imgf000048_0004
-48-
Figure imgf000049_0001
Figure imgf000050_0001
6. A compound according to any of claims 1 to 5 for use as a medicament.
7. A compound according to any of claims 1 to 5 for use in the prophylaxis and/or treatment of diseases mediated by FXR.
8. The compound for use according to claim 7 wherein the disease is selected from
chronic intrahepatic or some forms of extrahepatic cholestatic conditions;
liver fibrosis;
obstructive or chronic inflammatory disorders of the liver;
liver cirrhosis;
liver steatosis and associated syndromes, cholestatic or fibrotic effects that are associated with alcohol-induced cirrhosis or with viral-borne forms of hepatitis;
liver failure or liver ischemia after major liver resection;
chemotherapy associated steatohepatitis (CASH);
acute liver failure; and/or
Inflammatory Bowel Diseases.
9. The compound for use according to claim 7 wherein the disease is selected from
lipid and lipoprotein disorders;
Type II Diabetes and clinical complications of Type I and Type II Diabetes, including diabetic nephropathy, diabetic neuropathy, diabetic retinopathy and other observed effects of clinically manifest long term Diabetes;
conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and
specifically triglyceride accumulation and subsequent activation of profibrotic pathways, such as Non-Alcoholic Fatty Liver Disease (NAFLD), or Non-Alcoholic Steatohepatitis (NASH);
obesity or metabolic syndrome (combined conditions of dyslipidemia, diabetes or abnormally high body-mass index); and/or
acute myocardial infarction, acute stroke or thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis.
10. The compound for use according to claim 7 wherein the disease is selected from non-malignant hyperproliferative disorders and malignant hyperproliferative disorders, specifically of hepatocellular carcinoma, colon adenoma and polyposis, colon adenocarcinoma, breast cancer, pancreas adenocarcinoma, Barrett's esophagus or other forms of neoplastic diseases of the gastrointestinal tract and the liver.
11. Use of a compound according to any of claims 1 to 5 for the preparation of a medicament for the prophylaxis and/or treatment of diseases mediated by FXR.
12. The use according to claim 1 1 the disease is selected from
chronic intrahepatic or some forms of extrahepatic cholestatic conditions;
liver fibrosis;
obstructive or chronic inflammatory disorders of the liver;
liver cirrhosis;
liver steatosis and associated syndromes, cholestatic or fibrotic effects that are associated with alcohol-induced cirrhosis or with viral-borne forms of hepatitis;
liver failure or liver ischemia after major liver resection;
chemotherapy associated steatohepatitis (CASH);
acute liver failure; and/or
Inflammatory Bowel Diseases.
13. The use according to claim 1 1 wherein the disease is selected from
lipid and lipoprotein disorders;
Type II Diabetes and clinical complications of Type I and Type II Diabetes, including diabetic nephropathy, diabetic neuropathy, diabetic retinopathy and other observed effects of clinically manifest long term Diabetes;
conditions and diseases which result from chronic fatty and fibrotic degeneration of organs due to enforced lipid and specifically triglyceride accumulation and subsequent activation of profibrotic pathways; such as Non-Alcoholic Fatty Liver Disease (NAFLD) or Non-Alcoholic Steatohepatitis (NASH);
obesity or metabolic syndrome (combined conditions of dyslipidemia, diabetes or abnormally high body-mass index); and/or acute myocardial infarction, acute stroke or thrombosis which occurs as an endpoint of chronic obstructive atherosclerosis.
14. The use according to claim 1 1 wherein the disease is selected from
non-malignant hyperproliferative disorders and malignant hyperproliferative disorders, specifically of hepatocellular carcinoma, colon adenoma and polyposis, colon adenocarcinoma, breast cancer, pancreas adenocarcinoma, Barrett's esophagus or other forms of neoplastic diseases of the gastrointestinal tract and the liver.
PCT/EP2012/002941 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds WO2013007387A1 (en)

Priority Applications (38)

Application Number Priority Date Filing Date Title
SI201230535A SI2731676T1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) modulating compounds
CN201280033148.4A CN103702719B (en) 2011-07-13 2012-07-12 New FXR(NR1H4)With reference to and activity modulating compounds
CA2839357A CA2839357C (en) 2011-07-13 2012-07-12 Farnesoid x receptor (fxr) binding and activity modulating compounds
KR1020177021651A KR101859533B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
KR1020147002937A KR101722410B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
EA201391674A EA024843B1 (en) 2011-07-13 2012-07-12 Farnesoid x receptor (nr1h4) binding and activity modulating compounds
KR1020177000763A KR101766323B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
BR122020008872-9A BR122020008872B1 (en) 2011-07-13 2012-07-12 Use of a compound in the treatment of diseases and/or conditions by binding said nuclear receptor
MX2014000534A MX368371B (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds.
NZ620177A NZ620177B2 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
EP12735100.5A EP2731676B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) modulating compounds
AU2012283387A AU2012283387C1 (en) 2011-07-13 2012-07-12 Novel FXR (NR1H4) binding and activity modulating compounds
BR122019026062-1A BR122019026062B1 (en) 2011-07-13 2012-07-12 COMPOUNDS 3- (4 - ((3- (PIRIDIN-4-IL) ISOXAZOL-4-IL) METOXI) PHENYL) -3-HYDROXY-AZETIDIN-1-IL, 3- (4 - ((1- (PIRIDIN- ) 4-IL) -1H-1,2,3-TRIAZOL-5-IL) METOXI) PHENYL) -3-HYDROXY-AZETIDIN-1-IL, 3- (4 - ((4- (PIRIDIN-4-IL ) -1H-1,2,3-TRIAZOL-5-IL) METOXI) PHENYL) -3-HYDROXY-AZETIDIN-1-IL, E 3- (4 - ((1- (PIRIDIN-4-IL) -1H -PIRAZOL-5-IL) METOXI) PHENYL) -3-HYDROXY-AZETIDIN- 1-IL REPLACED AND OXIDES OF THE SAME
EP17000383.4A EP3246070B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating azoles
MEP-2016-81A ME02434B (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
BR112014000260-6A BR112014000260B1 (en) 2011-07-13 2012-07-12 FXR AGONIST COMPOUNDS, THEIR USE AND PHARMACEUTICAL COMPOSITION
PL12735100T PL2731676T3 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) modulating compounds
US14/232,118 US9139539B2 (en) 2011-07-13 2012-07-12 FXR (NR1H4) binding and activity modulating compounds
ES12735100.5T ES2569718T3 (en) 2011-07-13 2012-07-12 New compounds that modulate the activity of FXR (NR1H4)
JP2014519453A JP5986633B2 (en) 2011-07-13 2012-07-12 Novel FXR (NR1H4) binding and activity modulating compounds
DK12735100.5T DK2731676T3 (en) 2011-07-13 2012-07-12 New FXR (NR1H4) modulating compounds
KR1020187013304A KR101934335B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
RS20160266A RS54786B1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) modulating compounds
ZA2013/09521A ZA201309521B (en) 2011-07-13 2013-12-17 Novel fxr (nr1h4)binding and activity modulating compounds
IL229944A IL229944A (en) 2011-07-13 2013-12-17 Fxr (nr1h4) binding and activity modulating compounds
HK14105521.1A HK1192181A1 (en) 2011-07-13 2014-06-11 Novel fxr (nr1h4) modulating compounds fxr(nr1h4)
US14/824,971 US9539244B2 (en) 2011-07-13 2015-08-12 FXR (NR1H4) binding and activity modulating compounds
HRP20160442TT HRP20160442T1 (en) 2011-07-13 2016-04-25 Novel fxr (nr1h4) modulating compounds
SM201600169T SMT201600169B (en) 2011-07-13 2016-06-10 NEW COMPOUNDS FOR FXR MODULATION (NR1 H4)
AU2016265993A AU2016265993B2 (en) 2011-07-13 2016-11-29 Novel FXR (NR1H4) binding and activity modulating compounds
US15/369,521 US9820979B2 (en) 2011-07-13 2016-12-05 FXR (NR1H4) binding and activity modulating compounds
IL253437A IL253437A0 (en) 2011-07-13 2017-07-12 Novel fxr (nr1h4) binding and activity modulating compounds
US15/783,530 US10220027B2 (en) 2011-07-13 2017-10-13 FXR (NR1H4) binding and activity modulating compounds
AU2018203613A AU2018203613B2 (en) 2011-07-13 2018-05-22 Novel FXR (NR1H4) binding and activity modulating compounds
US16/248,178 US10485795B2 (en) 2011-07-13 2019-01-15 FXR (NR1H4) binding and activity modulating compounds
IL265973A IL265973B (en) 2011-07-13 2019-04-11 Novel fxr (nr1h4) binding and activity modulating compounds
AU2019261667A AU2019261667B2 (en) 2011-07-13 2019-11-04 Novel FXR (NR1H4) binding and activity modulating compounds
IL277570A IL277570A (en) 2011-07-13 2020-09-24 Novel fxr (nr1h4) binding and activity modulating compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161507153P 2011-07-13 2011-07-13
EP11005722.1 2011-07-13
US61/507,153 2011-07-13
EP11005722A EP2545964A1 (en) 2011-07-13 2011-07-13 Novel FXR (NR1H4) binding and activity modulating compounds

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/232,118 A-371-Of-International US9139539B2 (en) 2011-07-13 2012-07-12 FXR (NR1H4) binding and activity modulating compounds
US14/824,971 Continuation US9539244B2 (en) 2011-07-13 2015-08-12 FXR (NR1H4) binding and activity modulating compounds

Publications (1)

Publication Number Publication Date
WO2013007387A1 true WO2013007387A1 (en) 2013-01-17

Family

ID=44513243

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/002941 WO2013007387A1 (en) 2011-07-13 2012-07-12 Novel fxr (nr1h4) binding and activity modulating compounds

Country Status (31)

Country Link
US (5) US9139539B2 (en)
EP (4) EP2545964A1 (en)
JP (2) JP5986633B2 (en)
KR (4) KR101766323B1 (en)
CN (2) CN107252424B (en)
AR (1) AR087127A1 (en)
AU (4) AU2012283387C1 (en)
BR (3) BR122020008872B1 (en)
CA (1) CA2839357C (en)
CY (1) CY1117453T1 (en)
DK (2) DK2731676T3 (en)
EA (1) EA024843B1 (en)
ES (2) ES2632492T3 (en)
FI (1) FI3246070T3 (en)
HK (2) HK1192181A1 (en)
HR (1) HRP20160442T1 (en)
HU (1) HUE027931T2 (en)
IL (1) IL229944A (en)
LT (1) LT3246070T (en)
ME (1) ME02434B (en)
MX (2) MX368371B (en)
MY (1) MY161158A (en)
PL (2) PL2731676T3 (en)
PT (1) PT2987532T (en)
RS (1) RS54786B1 (en)
SI (2) SI2731676T1 (en)
SM (1) SMT201600169B (en)
TW (1) TWI439455B (en)
UY (1) UY34196A (en)
WO (1) WO2013007387A1 (en)
ZA (1) ZA201309521B (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3006939A1 (en) 2014-10-06 2016-04-13 Gilead Sciences, Inc. Histidine-rich Glycoprotein as a marker for hepatic Farnesoid X receptor activation
WO2016086218A1 (en) 2014-11-26 2016-06-02 Enanta Pharmaceuticals, Inc. Bile acid derivatives as fxr/tgr5 agonists and methods of use thereof
EP3034499A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Novel FXR (NR1H4) modulating compounds
EP3034501A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Hydroxy containing FXR (NR1H4) modulating compounds
WO2016127924A1 (en) 2015-02-13 2016-08-18 Sunshine Lake Pharma Co., Ltd. Tricyclic compounds and uses thereof in medicine
CN106588804A (en) * 2016-12-09 2017-04-26 都创(上海)医药科技有限公司 Preparation method of compound serving as farnesoid X receptor (FXR)
WO2017201150A1 (en) * 2016-05-18 2017-11-23 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
WO2017218337A1 (en) 2016-06-13 2017-12-21 Gilead Sciences, Inc. Fxr (nr1h4) modulating compounds
WO2018153933A1 (en) 2017-02-21 2018-08-30 Genfit Combination of a ppar agonist with a fxr agonist
US10080743B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10080742B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10080741B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2018178260A1 (en) 2017-03-30 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for reducing persistence and expression of episomal viruses
US20180333401A1 (en) * 2017-04-12 2018-11-22 Gilead Sciences, Inc. Methods of treating liver disease
US10138228B2 (en) 2016-05-18 2018-11-27 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use therof
US10149835B2 (en) 2016-05-18 2018-12-11 Elmore Patent Law Group, P.C. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2018231851A1 (en) 2017-06-13 2018-12-20 Gilead Sciences, Inc. Methods of treating liver fibrosis
KR20190017028A (en) * 2016-06-13 2019-02-19 길리애드 사이언시즈, 인코포레이티드 FXR (NR1H4) adjusting compound
US10220027B2 (en) 2011-07-13 2019-03-05 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
WO2019094777A1 (en) 2017-11-13 2019-05-16 Gilead Sciences, Inc. Compositions and methods for identifying and treating liver diseases and monitoring treatment outcomes
WO2019149158A1 (en) 2018-02-02 2019-08-08 Sunshine Lake Pharma Co., Ltd. Nitrogenous tricyclic compounds and uses thereof in medicine
US10450306B2 (en) 2016-10-04 2019-10-22 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
CN110461328A (en) * 2017-03-28 2019-11-15 吉利德科学公司 The therapeutic combination for treating liver disease
JP2019535811A (en) * 2016-10-22 2019-12-12 へパジーン セラピューティクス インコーポレイテッド Heterocyclic FXR modulator
US10597391B2 (en) 2016-10-26 2020-03-24 Enanta Pharmaceuticals, Inc. Urea-containing isoxazole derivatives as FXR agonists and methods of use thereof
WO2020065597A1 (en) 2018-09-28 2020-04-02 Richter Gedeon Nyrt. Bicyclic derivatives as gabaa α5 receptor modulators
US10676500B2 (en) 2017-04-07 2020-06-09 Enanta Pharmaceuticals, Inc. Process for preparation of sulfonyl carbamate bile acid derivatives
US10689391B2 (en) 2017-12-12 2020-06-23 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
EP3711762A1 (en) 2013-09-11 2020-09-23 INSERM (Institut National de la Santé et de la Recherche Médicale) A farnesoid x receptor agonsits foruse and pharmaceutical compositions for the treatment of chronic hepatitis b virus infection
US10829486B2 (en) 2018-02-14 2020-11-10 Enanta Pharmacueticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2021009332A1 (en) 2019-07-18 2021-01-21 Enyo Pharma Method for decreasing adverse-effects of interferon
US10988449B2 (en) 2017-04-12 2021-04-27 Il Dong Pharmaceutical Co., Ltd. Isoxazole derivatives as nuclear receptor agonists and uses thereof
US11040998B2 (en) 2015-03-31 2021-06-22 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
WO2021144330A1 (en) 2020-01-15 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of fxr agonists for treating an infection by hepatitis d virus
WO2021191837A1 (en) 2020-03-26 2021-09-30 Richter Gedeon Nyrt. 1,3-dihydro-2h-pyrrolo[3,4-c]pyridine derivatives as gabaa α5 receptor modulators
CN113573700A (en) * 2019-03-11 2021-10-29 吉利德科学公司 Formulations of compounds and uses thereof
US11168079B2 (en) 2017-11-01 2021-11-09 Bristol-Myers Squibb Company Alkene compounds as farnesoid x receptor modulators
US11225473B2 (en) 2019-01-15 2022-01-18 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11286252B2 (en) 2017-11-01 2022-03-29 Bristol-Myers Squibb Company Alkene spirocyclic compounds as farnesoid X receptor modulators
US11370785B2 (en) 2017-11-01 2022-06-28 Bristol-Myers Squibb Company Multicyclic compounds as farnesoid X receptor modulators
WO2022134146A1 (en) * 2020-12-22 2022-06-30 江苏天士力帝益药业有限公司 Novel fxr agonist having pyrazine structure, and preparation method and use
WO2022152770A1 (en) 2021-01-14 2022-07-21 Enyo Pharma Synergistic effect of a fxr agonist and ifn for the treatment of hbv infection
US11478533B2 (en) 2020-04-27 2022-10-25 Novo Nordisk A/S Semaglutide for use in medicine
WO2022229302A1 (en) 2021-04-28 2022-11-03 Enyo Pharma Strong potentiation of tlr3 agonists effects using fxr agonists as a combined treatment
US11524005B2 (en) 2019-02-19 2022-12-13 Gilead Sciences, Inc. Solid forms of FXR agonists
WO2022266444A1 (en) 2021-06-18 2022-12-22 Gilead Sciences, Inc. Il-31 modulators for treating fxr-induced pruritis
US11555032B2 (en) 2019-05-13 2023-01-17 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US11919879B2 (en) 2021-06-16 2024-03-05 Celgene Corporation Carboxylic acid containing azetidinyl compounds for the treatment of neurodegenerative diseases
WO2024089582A1 (en) 2022-10-25 2024-05-02 Assia Chemical Industries Ltd. Solid state forms of cilofexor salts

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI598347B (en) 2009-07-13 2017-09-11 基利科學股份有限公司 Apoptosis signal-regulating kinase inhibitors
IL253437A0 (en) * 2011-07-13 2017-09-28 Gilead Sciences Inc Novel fxr (nr1h4) binding and activity modulating compounds
NZ729678A (en) 2014-09-24 2018-07-27 Gilead Sciences Inc Methods of treating non-alcoholic steatohepatitis using ask1 inhibitor in combination with a fxr agonist
WO2016097933A1 (en) * 2014-12-18 2016-06-23 Novartis Ag Azabicyclooctane derivatives as fxr agonists for use in the treatment of liver and gastrointestinal diseases
MA41252A (en) 2014-12-23 2017-10-31 Gilead Sciences Inc SOLID FORMS OF AN ASK 1 INHIBITOR
CA3202893A1 (en) 2014-12-23 2016-06-30 Gilead Science, Inc. Processes for preparing ask1 inhibitors
IL274568B (en) 2015-07-06 2022-07-01 Gilead Sciences Inc Cot modulators and methods of use thereof
CN106995416A (en) * 2016-01-26 2017-08-01 上海翰森生物医药科技有限公司 FXR activators and its preparation method and application
WO2017128896A1 (en) * 2016-01-26 2017-08-03 江苏豪森药业集团有限公司 Fxr agonist and preparation method and use thereof
TW201741307A (en) * 2016-02-22 2017-12-01 艾洛斯生物製藥公司 FXR modulators and methods of their use
JP2019510752A (en) 2016-03-04 2019-04-18 ギリアード サイエンシーズ, インコーポレイテッド Compositions and combinations of autotaxin inhibitors
WO2017177179A1 (en) 2016-04-08 2017-10-12 Gilead Sciences, Inc. Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
AR108711A1 (en) * 2016-06-13 2018-09-19 Gilead Sciences Inc FXR MODULATING COMPOUNDS (NR1H4)
TW201808283A (en) 2016-08-05 2018-03-16 廣東東陽光藥業有限公司 Nitrogen-containing tricyclic compounds and uses thereof in medicine
CN106237332A (en) * 2016-08-11 2016-12-21 河南大学 Nuclear receptor FXR application in liver-cancer stem cell targeted therapy
AU2017356160A1 (en) * 2016-11-11 2019-05-23 Gilead Sciences, Inc. Methods of treating liver disease
CN108218852A (en) * 2016-12-15 2018-06-29 宁波百纳西药业有限公司 A kind of spiro-compound, preparation method, composition and purposes
WO2018133730A1 (en) * 2017-01-20 2018-07-26 四川科伦博泰生物医药股份有限公司 Heterocyclic compound, preparation method and use therefor
JOP20180017A1 (en) 2017-03-14 2019-01-30 Gilead Sciences Inc Apoptosis signal-regulating kinase inhibitor
JP7029583B2 (en) 2017-05-31 2022-03-04 パナソニックIpマネジメント株式会社 washing machine
US10980810B2 (en) 2017-10-06 2021-04-20 Gilead Sciences, Inc. Combination therapy comprising an ACC inhibitor
WO2019089667A1 (en) 2017-11-01 2019-05-09 Bristol-Myers Squibb Company Bridged bicyclic compounds as farnesoid x receptor modulators
EA202091101A1 (en) 2017-11-01 2020-08-06 Бристол-Маерс Сквибб Компани SPIROCYCLIC COMPOUNDS AS MODULATORS OF THE PHARNESOID X-RECEPTOR
ES2907187T3 (en) * 2018-08-08 2022-04-22 Inorbit Therapeutics Ab Compounds useful in modulating the farnesoid X receptor and methods of making and using the same
WO2020029908A1 (en) * 2018-08-08 2020-02-13 广州市恒诺康医药科技有限公司 Spiro bridged ring compound, pharmaceutical composition thereof and use thereof
MA53665A (en) 2018-09-18 2021-07-28 Metacrine Inc FARNESOID X RECEPTOR AGONISTS AND THEIR USES
AR118050A1 (en) 2019-02-15 2021-09-15 Bristol Myers Squibb Co BICYCLIC COMPOUNDS REPLACED AS MODULATORS OF THE FARNESOID X RECEIVER
TW202235416A (en) 2019-06-14 2022-09-16 美商基利科學股份有限公司 Cot modulators and methods of use thereof
US20230105984A1 (en) 2019-12-23 2023-04-06 Svetlana Marukian Compositions and methods for the treatment of liver diseases and disorders
EP4121048A1 (en) * 2020-03-18 2023-01-25 Metacrine, Inc. Farnesoid x receptor agonists for the treatment of disease
JP2023520650A (en) 2020-03-30 2023-05-18 ギリアード サイエンシーズ, インコーポレイテッド (S)-6-(((1-(bicyclo[1.1.1]pentan-1-yl)-1H-1,2,3-triazol-4-yl)2-methyl-1-oxo-1 ,2-dihydroisoquinolin-5-yl)methyl)))amino)8-chloro-(neopentylamino)quinoline-3-carbonitrile Cot inhibitor compounds solid forms
AU2021245924B2 (en) 2020-04-02 2024-02-29 Gilead Sciences, Inc. Process for preparing a Cot inhibitor compound
WO2022192428A1 (en) 2021-03-11 2022-09-15 Gilead Sciences, Inc. Glp-1r modulating compounds
US20230079863A1 (en) 2021-03-29 2023-03-16 Gilead Sciences, Inc. Khk inhibitors
TW202304435A (en) 2021-06-04 2023-02-01 美商基利科學股份有限公司 Methods of treating nash
WO2023125904A1 (en) * 2021-12-30 2023-07-06 苏州晶云药物科技股份有限公司 Crystal form of azacyclobutyl nicotinic acid compound and preparation method therefor

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000037077A1 (en) 1998-12-23 2000-06-29 Glaxo Group Limited Assays for ligands for nuclear receptors
WO2003015771A1 (en) 2001-08-13 2003-02-27 Lion Bioscience Ag Fxr nr1h4 nuclear receptor binding compounds
WO2003080803A2 (en) 2002-03-21 2003-10-02 Smithkline Beecham Corporation Methods of using farnesoid x receptor (fxr) agonists
WO2004048349A1 (en) 2002-11-22 2004-06-10 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2004087076A2 (en) 2003-03-31 2004-10-14 The Rockefeller University Methods for inhibiting adipogenesis and for treating type 2 diabetes
WO2007076260A2 (en) 2005-12-19 2007-07-05 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2007092751A2 (en) 2006-02-03 2007-08-16 Eli Lilly And Company Compounds and methods for modulating fx-receptors
WO2007140183A1 (en) 2006-05-24 2007-12-06 Eli Lilly And Company Fxr agonists
WO2007140174A2 (en) 2006-05-24 2007-12-06 Eli Lilly And Company Compounds and methods for modulating fxr
EP1894924A1 (en) * 2006-08-29 2008-03-05 Phenex Pharmaceuticals AG Heterocyclic FXR binding compounds
WO2008025539A1 (en) 2006-08-29 2008-03-06 Phenex Pharmaceuticals Ag Heterocyclic fxr binding compounds
WO2008051942A2 (en) 2006-10-24 2008-05-02 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2008157270A1 (en) 2007-06-13 2008-12-24 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2009005998A1 (en) 2007-07-02 2009-01-08 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2009012125A1 (en) 2007-07-16 2009-01-22 Eli Lilly And Company Compounds and methods for modulating fxr
WO2011020615A1 (en) 2009-08-19 2011-02-24 Phenex Pharmaceuticals Ag Novel fxr (nr1h4 ) binding and activity modulating compounds

Family Cites Families (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ193011A (en) 1979-03-19 1983-03-15 Ici Australia Ltd Diarylamine derivatives intermediates herbicidal compositions
EP0287959B1 (en) 1987-04-21 1993-04-28 BASF Aktiengesellschaft P-phenoxy--phenoxymethyl substituted five membered heteroaromatics
JP3121061B2 (en) 1991-10-04 2000-12-25 塩野義製薬株式会社 Method for producing intermediate for producing alkoxyiminoacetamides and intermediate used therein
DE4137940A1 (en) 1991-11-18 1993-05-19 Basf Ag 3-ISOXAZOLYLPHENYL COMPOUNDS, THEIR PRODUCTION AND THEIR USE
US5258551A (en) 1991-12-18 1993-11-02 Shionogi & Co., Ltd. Process for producing α-ketoamide derivative
AU5891494A (en) 1993-01-29 1994-08-15 Nippon Soda Co., Ltd. Heterocyclic derivative
WO1994024095A1 (en) 1993-04-16 1994-10-27 Abbott Laboratories Immunosuppressive agents
IL112721A0 (en) 1994-03-10 1995-05-26 Zeneca Ltd Azole derivatives
EP0804434A1 (en) 1994-08-02 1997-11-05 MERCK SHARP &amp; DOHME LTD. Azetidine, pyrrolidine and piperidine derivatives
GB9501865D0 (en) 1995-01-31 1995-03-22 Merck Sharp & Dohme Therapeutic agents
US5633272A (en) 1995-02-13 1997-05-27 Talley; John J. Substituted isoxazoles for the treatment of inflammation
DE19536811A1 (en) 1995-10-02 1997-04-03 Basf Ag Intermediates and processes for the production of substituted salicylic acid derivatives as crop protection agents
ES2183140T3 (en) 1996-02-13 2003-03-16 Searle & Co COMBINATIONS, WHICH HAVE IMMUNOSUPPRESSOR EFFECTS, CONTAINING A CYCLOXYGENASA-2 INHIBITOR AND A A4 HYDROLASSES LEUCOTRENE INHIBITOR.
WO2000077011A1 (en) 1999-06-11 2000-12-21 Allergan Sales, Inc. Organosilyl compounds having nuclear hormone receptor modulating activity
US7022725B2 (en) 2000-11-17 2006-04-04 Takeda Pharmaceutical Company Limited Isoxazole derivatives
US20040105885A1 (en) 2001-04-17 2004-06-03 Ping Gao Gelatin capsule exhibiting reduced cross-linking
US20040105883A1 (en) 2001-04-17 2004-06-03 Ping Gao Pharmaceutical dosage form capable of maintaining stable dissolution profile upon storage
US20040105884A1 (en) 2001-04-17 2004-06-03 Ping Gao Pharmaceutical dosage form comprising a sulfite compound
US20040131670A1 (en) 2001-04-17 2004-07-08 Ping Gao Pellicle-resistant gelatin capsule
EP1405636A4 (en) 2001-06-26 2009-04-15 Takeda Pharmaceutical Function regulator for retinoid relative receptor
US20070010562A1 (en) 2001-08-13 2007-01-11 Ulrike Bauer Nr1h4 nuclear receptor binding compounds
EP1423113A4 (en) 2001-08-13 2007-04-18 Phenex Pharmaceuticals Ag Nr1h4 nuclear receptor binding compounds
US7595311B2 (en) 2002-05-24 2009-09-29 Exelixis, Inc. Azepinoindole derivatives as pharmaceutical agents
EP1581525A2 (en) 2002-08-09 2005-10-05 AstraZeneca AB Compounds having an activity at metabotropic glutamate receptors
WO2004014881A2 (en) 2002-08-09 2004-02-19 Astra Zeneca Ab '1,2,4'oxadiazoles as modulators of metabotropic glutamate receptor-5
EP1536790A2 (en) 2002-08-09 2005-06-08 AstraZeneca AB Oxadiazoles as modulators of metabotropic glutamate receptor-5
EP1407774A1 (en) 2002-09-10 2004-04-14 LION Bioscience AG 2-Amino-4-quinazolinones as LXR nuclear receptor binding compounds
WO2004046162A2 (en) 2002-11-14 2004-06-03 The Scripps Research Institute Non-steroidal fxr agonists
US20050143449A1 (en) 2002-11-15 2005-06-30 The Salk Institute For Biological Studies Non-steroidal farnesoid X receptor modulators and methods for the use thereof
WO2005077345A1 (en) 2004-02-03 2005-08-25 Astrazeneca Ab Compounds for the treatment of gastro-esophageal reflux disease
WO2005077373A2 (en) 2004-02-03 2005-08-25 Astrazeneca Ab Treatment of gastro-esophageal reflux disease (gerd)
US7585881B2 (en) 2004-02-18 2009-09-08 Astrazeneca Ab Additional heteropolycyclic compounds and their use as metabotropic glutamate receptor antagonists
AU2005245411B2 (en) 2004-05-14 2009-04-23 Irm Llc Compounds and compositions as PPAR modulators
MY144903A (en) 2004-06-17 2011-11-30 Novartis Ag Pyrrolopyridine derivatives and their use as crth2 antagonists
EP1812071A2 (en) 2004-10-13 2007-08-01 PTC Therapeutics, Inc. Compounds for nonsense suppression, use of these compounds for the manufacture of a medicament for treating somatic mutation-related diseases
JP2008137894A (en) 2005-03-22 2008-06-19 Nippon Kayaku Co Ltd New acetylene derivative
WO2006132197A1 (en) 2005-06-07 2006-12-14 Shionogi & Co., Ltd. HETEROCYCLIC COMPOUND HAVING TYPE I 11β HYDROXYSTEROID DEHYDROGENASE INHIBITORY ACTIVITY
WO2007070796A1 (en) 2005-12-15 2007-06-21 Exelixis, Inc. Azepinoindole derivatives as pharmaceutical agents
US7560551B2 (en) 2006-01-23 2009-07-14 Amgen Inc. Aurora kinase modulators and method of use
BRPI0707794A2 (en) 2006-02-14 2011-05-10 Intercept Pharmaceuticals Inc bile acid derivatives, formulations and pharmaceutical compositions, as well as use of said compounds
EP2007759A4 (en) 2006-04-17 2010-12-22 Neuromed Pharmaceuticals Ltd Isoxazole derivatives as calcium channel blockers
ATE549338T1 (en) 2006-05-24 2012-03-15 Boehringer Ingelheim Int SUBSTITUTED PTERIDINES SUBSTITUTED WITH A FOUR-MEMBER HETEROCYCLE
CN101448798A (en) * 2006-05-24 2009-06-03 伊莱利利公司 Compounds and methods for modulating FXR
DK2040713T3 (en) 2006-06-27 2014-09-29 Intercept Pharmaceuticals Inc BILIC ACID DERIVATIVES AS FXR LIGANDS FOR THE PREVENTION OR TREATMENT OF FXR-MEDIATED DISEASES OR CONDITIONS
US8222256B2 (en) 2006-07-05 2012-07-17 Exelixis, Inc. Methods of using IGFIR and ABL kinase modulators
EA200900072A1 (en) 2006-07-07 2009-06-30 Бёрингер Ингельхайм Интернациональ Гмбх PHENYL-SUBSTITUTED HETEROARRYNE DERIVATIVES AND THEIR APPLICATION AS ANTI-TUMOR MEDIA
US20080032990A1 (en) 2006-07-07 2008-02-07 Khalifah Raja G Inhibitors of advanced glycation end products
US8193225B2 (en) 2006-10-13 2012-06-05 The Board Of Regents Of The University Of Texas System Isoxazole amides, derivatives and methods of chemical induction of neurogenesis
US8501933B2 (en) 2006-11-09 2013-08-06 Roche Palo Alto Llc Thiazole and oxazole-substituted arylamides as P2X3 and P2X2/3 antagonists
CN101679297B (en) 2006-12-08 2012-01-11 埃克塞利希斯股份有限公司 LXR and FXR modulators
GB0625842D0 (en) 2006-12-22 2007-02-07 Argenta Discovery Ltd Indolizine derivatives
US20090105251A1 (en) 2007-01-25 2009-04-23 Benjamin Jones Renin inhibitors
JP5254999B2 (en) 2007-02-09 2013-08-07 ダウ アグロサイエンシィズ エルエルシー Method for oxidizing certain substituted sulfilimines to insecticidal sulfoximines
US7511149B2 (en) 2007-02-09 2009-03-31 Dow Agrosciences Llc Process for the oxidation of certain substituted sulfilimines to insecticidal sulfoximines
AU2008219748B2 (en) 2007-02-26 2012-05-03 Corteva Agriscience Llc Process for the preparation of certain substituted sulfilimines
JP2008308448A (en) 2007-06-15 2008-12-25 Sankyo Agro Kk (3-sulfur atom-substituted phenyl)heteroaryl derivative
WO2008155054A1 (en) 2007-06-20 2008-12-24 F. Hoffmann-La Roche Ag Farnesoid-x-receptor mutants, and crystallisation thereof
EP2176249A2 (en) 2007-07-02 2010-04-21 Boehringer Ingelheim International GmbH New chemical compounds
US20090197880A1 (en) 2007-07-13 2009-08-06 Genelabs Technologies, Inc. Anti-viral compounds, compositions, and methods of use
EA201000051A1 (en) 2007-07-13 2010-08-30 ГЛАКСОСМИТКЛАЙН ЭлЭлСи ANTI-VIRUS COMPOUNDS, COMPOSITIONS AND METHODS OF USE
US20100256145A1 (en) 2007-08-01 2010-10-07 H. Lundbeck A/S Use of kcnq potassium channel openers for reducing symptoms of or treating disorders or conditions wherein the dopaminergic system is disrupted
US8188080B2 (en) 2007-10-17 2012-05-29 Sanford-Burnham Medical Research Institute VHR protein tyrosine phosphatase inhibitors, compositions and methods of use
US20090143451A1 (en) 2007-11-14 2009-06-04 Andrews William H Compounds that increase telomerase reverse transcriptase (tert) expression and methods for using the same
EP2110374A1 (en) 2008-04-18 2009-10-21 Merck Sante Benzofurane, benzothiophene, benzothiazol derivatives as FXR modulators
CA2724232A1 (en) 2008-05-13 2009-11-19 Boehringer Ingelheim International Gmbh Sulfone compounds which modulate the cb2 receptor
AU2009249180A1 (en) 2008-05-19 2009-11-26 Burnham Institute For Medical Research Intestinal Alkaline Phosphatase modulators and uses thereof
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2128158A1 (en) 2008-05-26 2009-12-02 Phenex Pharmaceuticals AG Heterocyclic cyclopropyl-substituted FXR binding compounds
AU2009257635A1 (en) 2008-06-10 2009-12-17 Plexxikon, Inc. 5H-Pyrrolo [2,3-b] pyrazine derivatives for kinase modulation, and indications therefor
US8822513B2 (en) 2010-03-01 2014-09-02 Gtx, Inc. Compounds for treatment of cancer
KR20110036586A (en) 2008-06-23 2011-04-07 바스프 에스이 Sulfoximinamide compounds for combating animal pests
WO2010006096A1 (en) 2008-07-11 2010-01-14 Smithkline Beecham Corporation Processes for the preparation of anti-viral compounds and compositions containing them
EP2318381A1 (en) 2008-08-25 2011-05-11 Dow Global Technologies LLC Process for preparing isoxazole compounds
US20120021519A1 (en) 2008-09-19 2012-01-26 Presidents And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
KR101444988B1 (en) 2008-09-25 2014-09-26 에프. 호프만-라 로슈 아게 3-amino-indazole or 3-amino-4,5,6,7-tetrahydro-indazole derivatives
AU2009296048A1 (en) 2008-09-25 2010-04-01 F. Hoffmann-La Roche Ag 2,3-substituted indazole or 4,5,6,7-tetrahydro-indazoles as FXR modulators against dyslipidemia and related diseases
WO2010036362A1 (en) 2008-09-26 2010-04-01 Wyeth 1,2,3,6-tetrahydroazepino[4,5-b]indole-5-carboxylate nuclear receptor inhibitors
KR20110082145A (en) 2008-10-21 2011-07-18 메타볼렉스, 인코포레이티드 Aryl gpr120 receptor agonists and uses thereof
CA2744946A1 (en) 2009-02-04 2010-08-12 Boehringer Ingelheim International Gmbh Cyclic inhibitors of 11.beta.-hydroxysteroid dehydrogenase 1
WO2010093191A2 (en) 2009-02-13 2010-08-19 Lg Life Sciences Ltd. Novel compounds effective as xanthine oxidase inhibitors, method for preparing the same, and pharmaceutical composition containing the same
FR2943059A1 (en) 2009-03-16 2010-09-17 Sanofi Aventis N-6-AZA-BICYCLO® 3.2.1.0-OCT-5-YL) -ARYL-METHYL-HETEROBENZAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
US8883832B2 (en) 2009-07-06 2014-11-11 Aerpio Therapeutics Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
WO2011003793A1 (en) 2009-07-06 2011-01-13 Basf Se Pyridazine compounds for controlling invertebrate pests
BR112012001597A2 (en) 2009-07-24 2015-09-01 Basf Se Method for controlling and / or combating invertebrate pests, method for protecting plant propagating material and / or growing plants, plant propagating material and method for treating or protecting an animal from infestation or parasite infection.
US9212177B2 (en) 2009-08-05 2015-12-15 Versitech Limited Antiviral compounds and methods of making and using thereof
CN102656151A (en) 2009-09-04 2012-09-05 扎里卡斯药品有限公司 Substituted heterocyclic derivatives for the treatment of pain and epilepsy
US9095596B2 (en) 2009-10-15 2015-08-04 Southern Research Institute Treatment of neurodegenerative diseases, causation of memory enhancement, and assay for screening compounds for such
KR20120130777A (en) 2010-03-01 2012-12-03 유니버시티 오브 테네시 리서치 파운데이션 Compounds for treatment of cancer
WO2011156640A2 (en) 2010-06-09 2011-12-15 Afraxis, Inc. 8-(HETEROARYLMETHYL)PYRIDO[2,3-d]PYRIMIDIN-7(8H)-ONES FOR THE TREATMENT OF CNS DISORDERS
WO2012058531A2 (en) 2010-10-29 2012-05-03 North Carolina State University Modulation of response regulators by imidazole derivatives
TWI408128B (en) 2010-12-03 2013-09-11 Nat Univ Tsing Hua M-terphenyl compound derivatives and application for organic light emitting diode
CN103391937A (en) 2010-12-20 2013-11-13 Irm责任有限公司 Compositions and methods for modulating farnesoid x receptors
US20130261108A1 (en) 2010-12-20 2013-10-03 Irm Llc Compositions and methods for modulating farnesoid x receptors
CU24152B1 (en) 2010-12-20 2016-02-29 Irm Llc 1,2 OXAZOL-8-AZABICICLO [3,2,1] OCTANO 8 IL AS FXR MODULATORS
EP2545964A1 (en) 2011-07-13 2013-01-16 Phenex Pharmaceuticals AG Novel FXR (NR1H4) binding and activity modulating compounds
WO2013037482A1 (en) 2011-09-15 2013-03-21 Phenex Pharmaceuticals Ag Farnesoid x receptor agonists for cancer treatment and prevention
CA3028207A1 (en) 2012-06-19 2013-12-27 Intercept Pharmaceuticals, Inc. Preparation, uses and solid forms of obeticholic acid
TWI621618B (en) 2013-03-13 2018-04-21 比利時商健生藥品公司 Substituted 2-azabicycles and their use as orexin receptor modulators
WO2014181287A1 (en) 2013-05-09 2014-11-13 Piramal Enterprises Limited Heterocyclyl compounds and uses thereof
UA118673C2 (en) 2013-05-14 2019-02-25 Інтерсепт Фармасутікалз, Інк. 11-hydroxyl-derivatives of bile acids and amino acid conjugates thereof as farnesoid x receptor modulators
CN105593237B (en) 2013-08-01 2019-06-04 美国卫生和人力服务部 The inhibitor of farnesoid X receptor and purposes in medicine
BR112016005507B1 (en) 2013-09-11 2023-02-07 Centre National De La Recherche Scientifique (Cnrs) USE OF A FARNESOID X RECEPTOR AGONIST (FXR)
US20150082981A1 (en) 2013-09-20 2015-03-26 E I Du Pont De Nemours And Company Capture of trifluoromethane using ionic liquids
CN104513213A (en) 2013-09-28 2015-04-15 山东亨利医药科技有限责任公司 Fxr agonist
WO2015065983A1 (en) 2013-10-29 2015-05-07 Lumena Pharmaceuticals, Inc. Bile acid recycling inhibitors for treatment of gastrointestinal infections
TN2016000143A1 (en) 2013-11-05 2017-10-06 Novartis Ag Compositions and methods for modulating farnesoid x receptors.
WO2015116856A2 (en) 2014-01-29 2015-08-06 City Of Hope Farnesoid x receptor antagonists
EP3116851B1 (en) 2014-03-13 2023-07-26 Salk Institute for Biological Studies Analogs of fexaramine and methods of making and using
US10077268B2 (en) 2014-03-13 2018-09-18 Salk Institute For Biological Studies FXR agonists and methods for making and using
CA2942403A1 (en) 2014-03-13 2015-09-17 Salk Institute For Biological Studies Fxr agonists and methods for making and using
JP6673850B2 (en) 2014-04-14 2020-03-25 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Heteroaryl-substituted heterocyclyl sulfone
WO2015162538A1 (en) 2014-04-21 2015-10-29 Lupin Limited Heterocyclic compounds as calcium sensing receptor modulators for the treatment of hyperparathyroidism, chronic renal failure and chronic kidney disease
WO2015162244A1 (en) 2014-04-25 2015-10-29 Basf Se N-acylamidine compounds
WO2015165960A1 (en) 2014-04-30 2015-11-05 Basf Se N-acylamidine compounds
SI3626725T1 (en) 2014-05-29 2023-04-28 Bar Pharmaceuticals S.R.L. Cholane derivatives for use in the treatment and/or prevention of fxr and tgr5/gpbar1 mediated diseases
CN104045635A (en) 2014-06-23 2014-09-17 华东理工大学 3,4,5-tri-substituted isoxazole compounds and applications thereof
WO2016054560A1 (en) 2014-10-02 2016-04-07 Flatley Discovery Lab Isoxazole compounds and methods for the treatment of cystic fibrosis
EP3006939A1 (en) 2014-10-06 2016-04-13 Gilead Sciences, Inc. Histidine-rich Glycoprotein as a marker for hepatic Farnesoid X receptor activation
CN106716665B9 (en) 2014-10-27 2018-12-07 株式会社Lg化学 Organic electroluminescent device
MX2017005935A (en) 2014-11-06 2018-01-11 Enanta Pharm Inc Bile acid analogs an fxr/tgr5 agonists and methods of use thereof.
RU2706007C2 (en) 2014-11-21 2019-11-13 Акарна Терапьютикс, Лтд. Condensed bicyclic compounds for treating disease
WO2016086134A1 (en) 2014-11-26 2016-06-02 Enanta Pharmaceuticals, Inc. Bile acid derivatives as fxr/tgr5 agonists and methods of use thereof
US10208081B2 (en) 2014-11-26 2019-02-19 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
CA2968404A1 (en) 2014-11-26 2016-06-02 Enanta Pharmaceuticals, Inc. Bile acid analogs as fxr/tgr5 agonists and methods of use thereof
WO2016086115A1 (en) 2014-11-26 2016-06-02 Enanta Pharmaceuticals, Inc. Tetrazole derivatives of bile acids as fxr/tgr5 agonists and methods of use thereof
EP3034501A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Hydroxy containing FXR (NR1H4) modulating compounds
EP3034499A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Novel FXR (NR1H4) modulating compounds
WO2016097933A1 (en) 2014-12-18 2016-06-23 Novartis Ag Azabicyclooctane derivatives as fxr agonists for use in the treatment of liver and gastrointestinal diseases
CA2972919A1 (en) 2015-01-09 2016-07-14 Gilead Apollo, Llc Acc inhibitor combination therapy for the treatment of non-alcoholic fatty liver disease
AU2016209321B2 (en) 2015-01-20 2019-05-09 Boehringer Ingelheim Animal Health USA Inc. Anthelmintic compounds, compositions and method of using thereof
TWI698430B (en) 2015-02-13 2020-07-11 南北兄弟藥業投資有限公司 Tricyclic compounds and uses thereof in medicine
US10100285B2 (en) 2015-04-03 2018-10-16 Propagenix Inc. Ex vivo proliferation of epithelial cells
CN106146483A (en) 2015-04-23 2016-11-23 上海迪诺医药科技有限公司 Heterocyclic method Buddhist nun's ester derivant X receptor modulators
GB201507340D0 (en) 2015-04-29 2015-06-10 Univ St Andrews Light emitting devices and compounds
ES2550374B1 (en) * 2015-06-30 2016-09-08 Universidad De La Rioja Analog photoprotective compounds of MAA, synthesis procedure and composition comprising the same
WO2017011466A1 (en) 2015-07-13 2017-01-19 Zwiebel Laurence J Thermal volatilization of orco agonists
EP3892718A1 (en) 2015-09-11 2021-10-13 Propagenix Inc. Ex vivo proliferation of epithelial cells
MX2018006217A (en) 2015-12-04 2018-08-01 Squibb Bristol Myers Co Apelin receptor agonists and methods of use.
WO2017097870A1 (en) 2015-12-11 2017-06-15 Bayer Cropscience Aktiengesellschaft Substituted malonic acid amides as insecticides
CN106946867B (en) 2016-01-06 2019-11-12 广州市恒诺康医药科技有限公司 FXR receptor modulators and its preparation method and application
EP3190103A1 (en) 2016-01-08 2017-07-12 Rijksuniversiteit Groningen Inhibitors of the pd-1/pd-l1 protein/protein interaction
WO2017117687A1 (en) 2016-01-10 2017-07-13 British Columbia Cancer Agency Branch 18/19f-labelled compounds which target the prostate specific membrane antigen
WO2017122209A2 (en) 2016-01-12 2017-07-20 Yeda Research And Development Co. Ltd. NF-kappaB INHIBITORS
WO2017128896A1 (en) 2016-01-26 2017-08-03 江苏豪森药业集团有限公司 Fxr agonist and preparation method and use thereof
CN107021958A (en) 2016-02-01 2017-08-08 山东轩竹医药科技有限公司 FXR receptor stimulating agents
CN107021957A (en) 2016-02-01 2017-08-08 山东轩竹医药科技有限公司 FXR receptor stimulating agents
CN108602811B (en) 2016-02-01 2021-11-16 轩竹生物科技有限公司 FXR receptor agonists
TW201741307A (en) 2016-02-22 2017-12-01 艾洛斯生物製藥公司 FXR modulators and methods of their use
CN107224583A (en) 2016-03-24 2017-10-03 中美华世通生物医药科技(武汉)有限公司 Medical composition and its use
US10080742B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2017189663A1 (en) 2016-04-26 2017-11-02 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
US10080741B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2017201152A1 (en) 2016-05-18 2017-11-23 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
WO2017201155A1 (en) 2016-05-18 2017-11-23 Enanta Pharmaceuticals, Inc. lSOXAZOLE DERIVATIVES AS FXR AGONISTS AND METHODS OF USE THEREOF
US10144729B2 (en) 2016-05-18 2018-12-04 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
US10682344B2 (en) 2016-06-03 2020-06-16 Chemocentryx, Inc. Method of treating liver fibrosis
CA2968836A1 (en) 2016-06-13 2017-12-13 Gilead Sciences, Inc. Fxr (nr1h4) modulating compounds
JP6678779B2 (en) 2016-06-13 2020-04-08 ギリアード サイエンシーズ, インコーポレイテッド FXR (NR1H4) modulating compound
TW201808283A (en) 2016-08-05 2018-03-16 廣東東陽光藥業有限公司 Nitrogen-containing tricyclic compounds and uses thereof in medicine
EP3954684A1 (en) 2016-08-23 2022-02-16 Ardelyx, Inc. Process for the preparation of hormone receptor modulators for treating metabolic conditions and disorders
WO2018039384A1 (en) 2016-08-23 2018-03-01 Ardelyx, Inc. Isoxazolyl-carbonyloxy azabicyclo[3.2.1]octanyl compounds as fxr activators
CN108430998B (en) 2016-09-28 2021-07-09 四川科伦博泰生物医药股份有限公司 Azabicyclo derivatives, preparation method and application thereof
US20200045972A1 (en) 2016-09-29 2020-02-13 Bayer Cropscience Aktiengesellschaft Novel 5-substituted imidazolylmethyl derivatives
KR20190056436A (en) 2016-10-04 2019-05-24 이난타 파마슈티칼스, 인코포레이티드 Isoxazole analogs as FXR agonists and methods for their use
CN107973790A (en) 2016-10-22 2018-05-01 合帕吉恩治疗公司 Heterocyclic FXR conditioning agent
CN110191724A (en) 2016-11-10 2019-08-30 盖尔梅德研究与发展有限公司 The treatment of fibrosis
AU2017356160A1 (en) 2016-11-11 2019-05-23 Gilead Sciences, Inc. Methods of treating liver disease
CN106588804B (en) 2016-12-09 2018-11-09 都创(上海)医药科技有限公司 A kind of preparation method of compound as Farnesoid X receptor (FXR)
CN106632294A (en) 2016-12-15 2017-05-10 宁波百纳西药业有限公司 Spiro compound and medicinal use thereof
CN106748922B (en) 2017-01-12 2019-02-01 中国药科大学 A kind of novel sulfone acid derivative, preparation method and its purposes as drug

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000037077A1 (en) 1998-12-23 2000-06-29 Glaxo Group Limited Assays for ligands for nuclear receptors
WO2003015771A1 (en) 2001-08-13 2003-02-27 Lion Bioscience Ag Fxr nr1h4 nuclear receptor binding compounds
WO2003080803A2 (en) 2002-03-21 2003-10-02 Smithkline Beecham Corporation Methods of using farnesoid x receptor (fxr) agonists
WO2004048349A1 (en) 2002-11-22 2004-06-10 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2004087076A2 (en) 2003-03-31 2004-10-14 The Rockefeller University Methods for inhibiting adipogenesis and for treating type 2 diabetes
WO2007076260A2 (en) 2005-12-19 2007-07-05 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2007092751A2 (en) 2006-02-03 2007-08-16 Eli Lilly And Company Compounds and methods for modulating fx-receptors
WO2007140183A1 (en) 2006-05-24 2007-12-06 Eli Lilly And Company Fxr agonists
WO2007140174A2 (en) 2006-05-24 2007-12-06 Eli Lilly And Company Compounds and methods for modulating fxr
EP1894924A1 (en) * 2006-08-29 2008-03-05 Phenex Pharmaceuticals AG Heterocyclic FXR binding compounds
WO2008025540A1 (en) 2006-08-29 2008-03-06 Phenex Pharmaceuticals Ag Heterocyclic fxr binding compounds
WO2008025539A1 (en) 2006-08-29 2008-03-06 Phenex Pharmaceuticals Ag Heterocyclic fxr binding compounds
WO2008051942A2 (en) 2006-10-24 2008-05-02 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2008157270A1 (en) 2007-06-13 2008-12-24 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2009005998A1 (en) 2007-07-02 2009-01-08 Smithkline Beecham Corporation Farnesoid x receptor agonists
WO2009012125A1 (en) 2007-07-16 2009-01-22 Eli Lilly And Company Compounds and methods for modulating fxr
WO2011020615A1 (en) 2009-08-19 2011-02-24 Phenex Pharmaceuticals Ag Novel fxr (nr1h4 ) binding and activity modulating compounds

Non-Patent Citations (73)

* Cited by examiner, † Cited by third party
Title
"March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", JOHN WILEY & SONS
A. ARANDA; A. PASCUAL, PHYSIOL. REV., vol. 81, 2001, pages 1269
A. FIGGE ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 2790
A. M. BRZOZOWSKI ET AL., NATURE, vol. 389, 1997, pages 753
A. MOSCHETTA ET AL., NATURE MEDICINE, vol. 10, 2004, pages 1352
B. CARIOU ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 11039
B. GOODWIN ET AL., MOL. CELL, vol. 6, 2000, pages 517
B. M. FORMAN ET AL., CELL, vol. 81, 1995, pages 687
C. J. SINAL ET AL., CELL, vol. 102, 2000, pages 731
C. LIHONG ET AL., AMERICAN DIABETES ASSOCIATION (ADA) 66TH ANNUAL SCIENTIFIC SESSIONS, June 2006 (2006-06-01)
D. DURAN-SANDOVAL ET AL., BIOCHIMIE, vol. 87, 2005, pages 93
D. J. MANGELSDORF ET AL., CELL, vol. 83, 1995, pages 835
D. J. PARKS ET AL., SCIENCE, vol. 284, 1999, pages 1365
D. M. HEERY ET AL., NATURE, vol. 387, 1997, pages 733
D. MERK ET AL., FUTURE MED. CHEM., vol. 4, 2012, pages 1015
DYER ET AL., ANAL. BIOCHEM., vol. 282, 2000, pages 158 - 161
E. A. HANNIMAN ET AL., J. LIPID RES., vol. 46, 2005, pages 2595
E. TOMLINSON ET AL., ENDOCRINOLOGY, vol. 143, 2002, pages 1741
F. CHEN ET AL., GASTROENTEROLOGY, vol. 126, 2004, pages 756
F. HE ET AL., CIRC. RES., vol. 98, 2006, pages 192
F. JOURNE ET AL., BREAST CANCER RES. TREAT., vol. 115, 2009, pages 523
F. YANG ET AL., CANCER RES., vol. 67, 2007, pages 863
FIESER ET AL.: "Fiesers' Reagents for organic Synthesis", 2000, JOHN WILEY & SONS
G. LAMBERT ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 2563
G. RIZZO ET AL., CURR. DRUG TARGETS IMMUNE ENDOCR. METABOL. DISORD., vol. 5, 2005, pages 289
G. RIZZO ET AL., CURR. DRUG TARGETS IMMUNE ENDOER. METABOL. DISORD., vol. 5, 2005, pages 289
G. ZOLLNER ET AL., MOL. PHARM., vol. 3, 2006, pages 231
H. R. KAST ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 2908
H. R. KAST ET AL., MOL. ENDOCRINOL., vol. 15, 2001, pages 1720
H. WITTENBURG, GASTROENTEROLOGY, vol. 125, 2003, pages 868
I. KIM ET AL., CARCINOGENESIS, vol. 28, 2007, pages 940
J. A. HOLT ET AL., GENES DEV., vol. 17, 2003, pages 1581
J. HOLT ET AL., GENES DEV., vol. 17, 2003, pages 1581
J. R. PLASS ET AL., HEPATOLOGY, vol. 35, 2002, pages 589
K. E. SWALES ET AL., CANCER RES., vol. 66, 2006, pages 10120
K. MA ET AL., J. CLIN. INVEST., vol. 116, 2006, pages 1102
K. R. STAYROOK ET AL., ENDOCRINOLOGY, vol. 146, 2005, pages 984
K. W. NETTLES; G. L. GREENE, ANNU. REV. PHYSIOL., vol. 67, 2005, pages 309
L. ALVAREZ ET AL., HUM. MOL. GENET., vol. 13, 2004, pages 2451
L. HUANG ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 51085
L. HUANG; J. DONG; S. KARKI: "Evaluation of drug candidates for preclinical development", 2010, WILEY & SONS, pages: 187 - 217
M. ANANTHANARAYANAN ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 28857
M. L. CRAWLEY, EXPERT OPIN THER. PAT., vol. 20, 2010, pages 1047
M. MAKISHIMA ET AL., SCIENCE, vol. 284, 1999, pages 1362
M. MIYATA, J. PHARMACOL. EXP. THER., vol. 312, 2005, pages 759
M. SCHENA; K. R. YAMAMOTO, SCIENCE, vol. 241, 1988, pages 965
M. WATANABE ET AL., J. CLIN. INVEST., vol. 113, 2004, pages 1408
N. L. URIZAR ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 39313
N. L. URIZAR ET AL., SCIENCE, vol. 296, 2002, pages 1703
P. MALONEY ET AL., J. MED. CHEM., vol. 43, 2000, pages 2971
P. R. MALONEY ET AL., J. MED. CHEM., vol. 43, 2000, pages 2971
R. M. EVANS, MOL. ENDOCRINOL., vol. 19, 2005, pages 1429
R. PELLICCIARI ET AL., J. MED. CHEM., vol. 45, 2002, pages 3569
R. R. MARAN ET AL., J. PHARMACOL. EXP. THER., vol. 328, 2009, pages 469
S. A. DOGGRELL, CURR. OPIN. INVESTIG. DRUGS, vol. 7, 2006, pages 344
S. BILZ ET AL., AM. J. PHYSIOL. ENDOCRINOL. METAB., vol. 290, 2006, pages 716
S. FIORUCCI ET AL., GASTROENTEROLOGY, vol. 127, 2004, pages 1497
S. FIORUCCI ET AL., J. PHARMACOL. EXP. THER., vol. 313, 2005, pages 604
S. FIORUCCI ET AL., J. PHARMACOL. EXP. THER., vol. 314, 2005, pages 584
S. MODICA ET AL., CANCER RES., vol. 68, 2008, pages 9589
S. Y. CAI ET AL., EXPERT OPIN. THER. TARGETS, vol. 10, 2006, pages 409
T. CLAUDEL ET AL., ARTERIOSCLER. THROMB. VASC. BIOL., vol. 25, 2005, pages 2020
T. EICHER; S. HAUPTMANN: "The Chemistry of Heterocycles; Structures, Reactions, Synthesis and Application", 2003, WILEY-VCH
T. HEINZEL ET AL., NATURE, vol. 387, 1997, pages 43
T. INAGAKI ET AL., CELL METAB., vol. 2, 2005, pages 217
T. INAGAKI ET AL., PNAS, vol. 103, 2006, pages 3920
T. M. WILLSON ET AL., MED. RES. REV., vol. 21, 2001, pages 513
T. T. LU ET AL., MOL. CELL, vol. 6, 2000, pages 507
T. WILLSON ET AL., MED. RES. REV., vol. 21, 2001, pages 513
W. HUANG ET AL., SCIENCE, vol. 312, 2006, pages 233
Y. D. WANG ET AL., CELL RES., vol. 18, 2008, pages 1087
Y. LIU ET AL., J. CLIN. INVEST., vol. 112, 2003, pages 1678
Y. ZHANG ET AL., PNAS, vol. 103, 2006, pages 1006

Cited By (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10485795B2 (en) 2011-07-13 2019-11-26 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US10220027B2 (en) 2011-07-13 2019-03-05 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
EP3711762A1 (en) 2013-09-11 2020-09-23 INSERM (Institut National de la Santé et de la Recherche Médicale) A farnesoid x receptor agonsits foruse and pharmaceutical compositions for the treatment of chronic hepatitis b virus infection
EP3006939A1 (en) 2014-10-06 2016-04-13 Gilead Sciences, Inc. Histidine-rich Glycoprotein as a marker for hepatic Farnesoid X receptor activation
US11718641B2 (en) 2014-11-26 2023-08-08 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
WO2016086218A1 (en) 2014-11-26 2016-06-02 Enanta Pharmaceuticals, Inc. Bile acid derivatives as fxr/tgr5 agonists and methods of use thereof
US10968249B2 (en) 2014-11-26 2021-04-06 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
KR20170095955A (en) * 2014-12-17 2017-08-23 길리애드 사이언시즈, 인코포레이티드 Hydroxy containing fxr (nr1h4) modulating compounds
AU2015365945B2 (en) * 2014-12-17 2018-09-27 Gilead Sciences, Inc. Novel FXR (NR1H4) modulating compounds
EA033642B1 (en) * 2014-12-17 2019-11-12 Gilead Sciences Inc Fxr (nr1h4) modulating compounds
CN107108595A (en) * 2014-12-17 2017-08-29 吉利德科学公司 New FXR (NR1H4) modulating compound
US9751874B2 (en) 2014-12-17 2017-09-05 Gilead Sciences, Inc. Hydroxy containing FXR (NR1H4) modulating compounds
KR101998007B1 (en) 2014-12-17 2019-07-08 길리애드 사이언시즈, 인코포레이티드 Hydroxy containing fxr (nr1h4) modulating compounds
EP3034499A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Novel FXR (NR1H4) modulating compounds
JP2018500304A (en) * 2014-12-17 2018-01-11 ギリアード サイエンシーズ, インコーポレイテッド Hydroxy-containing FXR (NR1H4) modulating compound
JP2018500305A (en) * 2014-12-17 2018-01-11 ギリアード サイエンシーズ, インコーポレイテッド Novel FXR (NR1H4) modulating compound
US9938278B2 (en) 2014-12-17 2018-04-10 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
CN108064223A (en) * 2014-12-17 2018-05-22 吉利德科学公司 FXR (NR1H4) modulating compound of hydroxyl
TWI697483B (en) * 2014-12-17 2020-07-01 美商基利科學股份有限公司 Hydroxy containing fxr (nr1h4) modulating compounds
KR101955840B1 (en) 2014-12-17 2019-03-07 길리애드 사이언시즈, 인코포레이티드 Novel fxr (nr1h4) modulating compounds
EP3034501A1 (en) 2014-12-17 2016-06-22 Gilead Sciences, Inc. Hydroxy containing FXR (NR1H4) modulating compounds
WO2016096116A1 (en) * 2014-12-17 2016-06-23 Gilead Sciences, Inc. Novel fxr (nr1h4) modulating compounds
KR20170093969A (en) * 2014-12-17 2017-08-16 길리애드 사이언시즈, 인코포레이티드 Novel fxr (nr1h4) modulating compounds
CN107108595B (en) * 2014-12-17 2021-04-06 吉利德科学公司 Novel FXR (NR1H4) modulating compounds
CN108064223B (en) * 2014-12-17 2021-06-01 吉利德科学公司 Fxr (nr1h4) modulating compounds containing hydroxy groups
AU2015365944B2 (en) * 2014-12-17 2019-01-17 Gilead Sciences, Inc. Hydroxy containing FXR (NR1H4) modulating compounds
WO2016096115A1 (en) * 2014-12-17 2016-06-23 Gilead Sciences, Inc. Hydroxy containing fxr (nr1h4) modulating compounds
WO2016127924A1 (en) 2015-02-13 2016-08-18 Sunshine Lake Pharma Co., Ltd. Tricyclic compounds and uses thereof in medicine
US11040998B2 (en) 2015-03-31 2021-06-22 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
US11958879B2 (en) 2015-03-31 2024-04-16 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
US10080741B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10080742B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10080743B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10149835B2 (en) 2016-05-18 2018-12-11 Elmore Patent Law Group, P.C. Isoxazole derivatives as FXR agonists and methods of use thereof
US10144729B2 (en) 2016-05-18 2018-12-04 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
US10138228B2 (en) 2016-05-18 2018-11-27 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use therof
WO2017201150A1 (en) * 2016-05-18 2017-11-23 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
KR102276042B1 (en) * 2016-06-13 2021-07-14 길리애드 사이언시즈, 인코포레이티드 FXR (NR1H4) modulating compound
KR20190017028A (en) * 2016-06-13 2019-02-19 길리애드 사이언시즈, 인코포레이티드 FXR (NR1H4) adjusting compound
KR102269305B1 (en) * 2016-06-13 2021-06-25 길리애드 사이언시즈, 인코포레이티드 FXR (NR1H4) modulating compound
EA037744B1 (en) * 2016-06-13 2021-05-17 Джилид Сайэнс, Инк. Fxr (nr1h4) modulating compounds
US10981881B2 (en) 2016-06-13 2021-04-20 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10329286B2 (en) 2016-06-13 2019-06-25 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
KR20190017027A (en) * 2016-06-13 2019-02-19 길리애드 사이언시즈, 인코포레이티드 FXR (NR1H4) adjusting compound
US20190315729A1 (en) * 2016-06-13 2019-10-17 Gilead Sciences, Inc. Fxr (nr1h4) modulating compounds
AU2017284160B2 (en) * 2016-06-13 2019-12-19 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
EP3730487A1 (en) 2016-06-13 2020-10-28 Gilead Sciences, Inc. Azetidine derivatives as fxr (nr1h4) modulators
US11247986B2 (en) 2016-06-13 2022-02-15 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10774054B2 (en) 2016-06-13 2020-09-15 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11739065B2 (en) 2016-06-13 2023-08-29 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
WO2017218337A1 (en) 2016-06-13 2017-12-21 Gilead Sciences, Inc. Fxr (nr1h4) modulating compounds
US11034684B2 (en) 2016-10-04 2021-06-15 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
US10450306B2 (en) 2016-10-04 2019-10-22 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
JP2019535811A (en) * 2016-10-22 2019-12-12 へパジーン セラピューティクス インコーポレイテッド Heterocyclic FXR modulator
JP7065107B2 (en) 2016-10-22 2022-05-11 へパジーン セラピューティクス (エイチケイ) リミテッド Heterocyclic FXR modulator
US10597391B2 (en) 2016-10-26 2020-03-24 Enanta Pharmaceuticals, Inc. Urea-containing isoxazole derivatives as FXR agonists and methods of use thereof
CN106588804A (en) * 2016-12-09 2017-04-26 都创(上海)医药科技有限公司 Preparation method of compound serving as farnesoid X receptor (FXR)
WO2018153933A1 (en) 2017-02-21 2018-08-30 Genfit Combination of a ppar agonist with a fxr agonist
US11833150B2 (en) 2017-03-28 2023-12-05 Gilead Sciences, Inc. Methods of treating liver disease
CN110461328A (en) * 2017-03-28 2019-11-15 吉利德科学公司 The therapeutic combination for treating liver disease
WO2018178260A1 (en) 2017-03-30 2018-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for reducing persistence and expression of episomal viruses
US10676500B2 (en) 2017-04-07 2020-06-09 Enanta Pharmaceuticals, Inc. Process for preparation of sulfonyl carbamate bile acid derivatives
US10961272B2 (en) 2017-04-07 2021-03-30 Enanta Pharmaceuticals, Inc. Process for preparation of sulfonyl carbamate bile acid derivatives
US10988449B2 (en) 2017-04-12 2021-04-27 Il Dong Pharmaceutical Co., Ltd. Isoxazole derivatives as nuclear receptor agonists and uses thereof
US20180333401A1 (en) * 2017-04-12 2018-11-22 Gilead Sciences, Inc. Methods of treating liver disease
WO2018231851A1 (en) 2017-06-13 2018-12-20 Gilead Sciences, Inc. Methods of treating liver fibrosis
US10495648B2 (en) 2017-06-13 2019-12-03 Gilead Sciences, Inc. Methods of treating liver fibrosis
US11286252B2 (en) 2017-11-01 2022-03-29 Bristol-Myers Squibb Company Alkene spirocyclic compounds as farnesoid X receptor modulators
US11168079B2 (en) 2017-11-01 2021-11-09 Bristol-Myers Squibb Company Alkene compounds as farnesoid x receptor modulators
US11370785B2 (en) 2017-11-01 2022-06-28 Bristol-Myers Squibb Company Multicyclic compounds as farnesoid X receptor modulators
WO2019094777A1 (en) 2017-11-13 2019-05-16 Gilead Sciences, Inc. Compositions and methods for identifying and treating liver diseases and monitoring treatment outcomes
US10689391B2 (en) 2017-12-12 2020-06-23 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
WO2019149158A1 (en) 2018-02-02 2019-08-08 Sunshine Lake Pharma Co., Ltd. Nitrogenous tricyclic compounds and uses thereof in medicine
US10829486B2 (en) 2018-02-14 2020-11-10 Enanta Pharmacueticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2020065597A1 (en) 2018-09-28 2020-04-02 Richter Gedeon Nyrt. Bicyclic derivatives as gabaa α5 receptor modulators
US11225473B2 (en) 2019-01-15 2022-01-18 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11524005B2 (en) 2019-02-19 2022-12-13 Gilead Sciences, Inc. Solid forms of FXR agonists
CN113573700A (en) * 2019-03-11 2021-10-29 吉利德科学公司 Formulations of compounds and uses thereof
US11555032B2 (en) 2019-05-13 2023-01-17 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2021009332A1 (en) 2019-07-18 2021-01-21 Enyo Pharma Method for decreasing adverse-effects of interferon
WO2021144330A1 (en) 2020-01-15 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of fxr agonists for treating an infection by hepatitis d virus
WO2021191837A1 (en) 2020-03-26 2021-09-30 Richter Gedeon Nyrt. 1,3-dihydro-2h-pyrrolo[3,4-c]pyridine derivatives as gabaa α5 receptor modulators
US11478533B2 (en) 2020-04-27 2022-10-25 Novo Nordisk A/S Semaglutide for use in medicine
WO2022134146A1 (en) * 2020-12-22 2022-06-30 江苏天士力帝益药业有限公司 Novel fxr agonist having pyrazine structure, and preparation method and use
WO2022152770A1 (en) 2021-01-14 2022-07-21 Enyo Pharma Synergistic effect of a fxr agonist and ifn for the treatment of hbv infection
WO2022229302A1 (en) 2021-04-28 2022-11-03 Enyo Pharma Strong potentiation of tlr3 agonists effects using fxr agonists as a combined treatment
US11919879B2 (en) 2021-06-16 2024-03-05 Celgene Corporation Carboxylic acid containing azetidinyl compounds for the treatment of neurodegenerative diseases
WO2022266444A1 (en) 2021-06-18 2022-12-22 Gilead Sciences, Inc. Il-31 modulators for treating fxr-induced pruritis
WO2024089582A1 (en) 2022-10-25 2024-05-02 Assia Chemical Industries Ltd. Solid state forms of cilofexor salts

Also Published As

Publication number Publication date
CN107252424B (en) 2020-03-27
MX2019011629A (en) 2019-12-05
EP2545964A1 (en) 2013-01-16
AU2019261667A1 (en) 2019-11-28
US10485795B2 (en) 2019-11-26
US9139539B2 (en) 2015-09-22
JP6321097B2 (en) 2018-05-09
ES2632492T3 (en) 2017-09-13
CN103702719A (en) 2014-04-02
JP2017014249A (en) 2017-01-19
BR122020008872B1 (en) 2022-04-12
DK3246070T3 (en) 2024-04-02
BR112014000260A2 (en) 2017-03-01
HK1192181A1 (en) 2014-08-15
AU2018203613B2 (en) 2019-08-08
US9820979B2 (en) 2017-11-21
PL2731676T3 (en) 2016-08-31
US10220027B2 (en) 2019-03-05
RS54786B1 (en) 2016-10-31
UY34196A (en) 2013-01-31
CN103702719B (en) 2017-09-08
MX368371B (en) 2019-09-30
ME02434B (en) 2016-09-20
SMT201600169B (en) 2016-07-01
US20170143684A1 (en) 2017-05-25
EP2731676A1 (en) 2014-05-21
KR20180053771A (en) 2018-05-23
TW201302719A (en) 2013-01-16
AU2018203613A1 (en) 2018-06-14
JP2014520823A (en) 2014-08-25
BR112014000260B1 (en) 2020-08-11
KR20140051932A (en) 2014-05-02
KR20170093258A (en) 2017-08-14
TWI439455B (en) 2014-06-01
KR101859533B1 (en) 2018-05-18
AU2012283387A1 (en) 2014-01-09
KR101934335B1 (en) 2019-01-02
LT3246070T (en) 2024-04-25
AU2012283387C1 (en) 2020-03-12
US9539244B2 (en) 2017-01-10
PT2987532T (en) 2017-07-13
EP3246070B1 (en) 2024-03-20
KR20170007543A (en) 2017-01-18
NZ620177A (en) 2015-10-30
ES2569718T3 (en) 2016-05-12
JP5986633B2 (en) 2016-09-06
EP2987532B1 (en) 2017-04-12
EP2987532A1 (en) 2016-02-24
MX2014000534A (en) 2014-05-13
IL229944A (en) 2017-07-31
CA2839357C (en) 2017-10-24
MY161158A (en) 2017-04-14
CY1117453T1 (en) 2017-04-26
AR087127A1 (en) 2014-02-12
EA201391674A1 (en) 2014-08-29
KR101766323B1 (en) 2017-08-08
EA024843B1 (en) 2016-10-31
HK1221680A1 (en) 2017-06-09
BR122019026062B1 (en) 2020-12-15
US20150342930A1 (en) 2015-12-03
EP2731676B1 (en) 2016-02-03
EP3246070A1 (en) 2017-11-22
NZ710770A (en) 2017-02-24
KR101722410B1 (en) 2017-04-03
PL2987532T3 (en) 2017-10-31
HUE027931T2 (en) 2016-11-28
AU2016265993A1 (en) 2016-12-15
HRP20160442T1 (en) 2016-05-20
AU2016265993B2 (en) 2018-03-15
DK2731676T3 (en) 2016-05-02
US20180200243A1 (en) 2018-07-19
SI2987532T1 (en) 2017-07-31
SI2731676T1 (en) 2016-06-30
ZA201309521B (en) 2014-08-27
US20140221659A1 (en) 2014-08-07
AU2019261667B2 (en) 2021-03-11
FI3246070T3 (en) 2024-04-18
US20190142814A1 (en) 2019-05-16
CN107252424A (en) 2017-10-17
AU2012283387B2 (en) 2016-09-08
CA2839357A1 (en) 2013-01-17

Similar Documents

Publication Publication Date Title
AU2018203613B2 (en) Novel FXR (NR1H4) binding and activity modulating compounds
JP2019214611A (en) Novel FXR (NR1H4) binding and activity modulating compounds
NZ710770B2 (en) Novel fxr (nr1h4) binding and activity modulating compounds
NZ620177B2 (en) Novel fxr (nr1h4) binding and activity modulating compounds

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12735100

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 201391674

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2839357

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2012283387

Country of ref document: AU

Date of ref document: 20120712

Kind code of ref document: A

Ref document number: 2014519453

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/000534

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2012735100

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20147002937

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14232118

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014000260

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: P-2016/0266

Country of ref document: RS

ENP Entry into the national phase

Ref document number: 112014000260

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140106