WO2016168553A1 - Deuterated obeticholic acid - Google Patents

Deuterated obeticholic acid Download PDF

Info

Publication number
WO2016168553A1
WO2016168553A1 PCT/US2016/027688 US2016027688W WO2016168553A1 WO 2016168553 A1 WO2016168553 A1 WO 2016168553A1 US 2016027688 W US2016027688 W US 2016027688W WO 2016168553 A1 WO2016168553 A1 WO 2016168553A1
Authority
WO
WIPO (PCT)
Prior art keywords
deuterium
compound
hydrogen
same
formula
Prior art date
Application number
PCT/US2016/027688
Other languages
French (fr)
Other versions
WO2016168553A8 (en
Inventor
I. Robert Silverman
Roger D. Tung
Original Assignee
Concert Pharmaceuticals, Inc.
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
Application filed by Concert Pharmaceuticals, Inc. filed Critical Concert Pharmaceuticals, Inc.
Publication of WO2016168553A1 publication Critical patent/WO2016168553A1/en
Publication of WO2016168553A8 publication Critical patent/WO2016168553A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/001Acyclic or carbocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • C07J9/005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane containing a carboxylic function directly attached or attached by a chain containing only carbon atoms to the cyclopenta[a]hydrophenanthrene skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J7/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms
    • C07J7/0005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms not substituted in position 21
    • C07J7/001Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms not substituted in position 21 substituted in position 20 by a keto group
    • C07J7/0015Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms not substituted in position 21 substituted in position 20 by a keto group not substituted in position 17 alfa
    • C07J7/002Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms not substituted in position 21 substituted in position 20 by a keto group not substituted in position 17 alfa not substituted in position 16

Definitions

  • ADME absorption, distribution, metabolism and/or excretion
  • ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites.
  • some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent.
  • modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
  • a metabolic inhibitor will be co- administered with a drug that is cleared too rapidly.
  • a drug that is cleared too rapidly.
  • the FDA recommends that these drugs be co-dosed with ritonavir, an inhibitor of cytochrome P450 enzyme 3A4 (CYP3A4), the enzyme typically responsible for their metabolism (see Kempf, D.J. et al., Antimicrobial agents and chemotherapy, 1997, 41(3): 654-60).
  • CYP3A4 cytochrome P450 enzyme 3A4
  • Ritonavir causes adverse effects and adds to the pill burden for HIV patients who must already take a combination of different drugs.
  • the CYP2D6 inhibitor quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism of dextromethorphan in a treatment of pseudobulbar affect.
  • Quinidine has unwanted side effects that greatly limit its use in potential combination therapy (see Wang, L et al., Clinical Pharmacology and Therapeutics, 1994, 56(6 Pt 1): 659-67; and FDA label for quinidine at www.accessdata.fda.gov).
  • a potentially attractive strategy for improving a drug's metabolic properties is deuterium modification.
  • Deuterium is a safe, stable, nonradioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability.
  • the size and shape of deuterium are essentially identical to those of hydrogen, replacement of hydrogen by deuterium would not be expected to affect the biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen.
  • This invention relates to deuterated forms of obeticholic acid
  • the invention provides a
  • each of Y , Y , Y , Y , Y , Y 3b , Y 4 , Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 , Y 9 , and Y 10 is independently selected from hydrogen and deuterium; and at least one of Y la , Y lb , Y 2a , Y 2b , Y 3a , Y 3b , Y 4 , Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 , Y 9 , or Y 10 is deuterium.
  • This invention also provides compositions comprising a compound of the invention, including pharmaceutical compositions comprising a compound of this invention and a pharmaceutically acceptable carrier.
  • the invention also provides the use of such compounds and compositions in methods of treating diseases and conditions that are beneficially treated by administering an agonist of the farnesoid X receptor (FXR).
  • FXR farnesoid X receptor
  • Some exemplary embodiments include a method of treating a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL- cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia and hyperlipidemia, the method comprising the step of administering to a subject in need thereof a pharmaceutically acceptable composition of the present invention.
  • PBC primary biliary cirrhosis
  • NASH non-alcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • AH alcoholic hepatitis
  • AH hepati
  • Obeticholic acid also known as (3a,5B,6a,7a)-6-ethyl-3,7- dihydroxycholan-24-oic acid, and as 6-ethylchenodeoxycholic acid, and as INT-747) is a potent and selective agonist of farnesoid X receptor (FXR).
  • FXR plays an integral role in bile acid synthesis and transport and its activation regulates the expression of a number of genes involved in bile acid and cholesterol homeostasis.
  • FXR is considered a prime target for treatment of hepatobiliary, metabolic and intestinal disorders.
  • Obeticholic acid has been submitted for regulatory approval in the U.S. for the treatment of primary biliary cirrhosis (PBC) as monotherapy or in combination with
  • ursodeoxycholic acid is also in phase III clinical trials for treatment of non-alcoholic steatohepatitis (NASH), in phase II clinical trials for sclerosing cholangitis, diabetes and non-alcoholic fatty liver disease (NAFLD).
  • NASH non-alcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • treat means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
  • a disease e.g., a disease or disorder delineated herein
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen”
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • a position is designated specifically as “D” or “deuterium”
  • the position is understood to have deuterium at an abundance that is at least 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1% incorporation of deuterium).
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • isotopologue refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
  • a compound represented by a particular chemical structure containing indicated deuterium atoms will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure.
  • the relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
  • the invention also provides salts of the compounds of the invention.
  • a salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • the compound is a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylprop
  • pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
  • the pharmaceutically acceptable salt may also be a salt of a compound of the present invention having an acidic functional group, such as a carboxylic acid functional group, and a base.
  • exemplary bases include, but are not limited to, hydroxide of alkali metals including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydro xyl- substituted mono- , di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N- ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-(Ci-C6)- alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2- hydroxyethyl)amine; N-
  • the compounds of the present invention may contain an asymmetric carbon atom, for example, as the result of deuterium substitution or otherwise.
  • compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers.
  • a compound of the present invention may exist as either a racemic mixture or a scalemic mixture, or as individual respective stereoisomers that are substantially free from another possible stereoisomer.
  • substantially free of other stereoisomers as used herein means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present.
  • stable compounds refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
  • Substituted with deuterium refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
  • each Y may be referred to specifically (e.g., Y l , Y 4 , Y 6 , etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
  • the present invention provides a compound of Formula A:
  • R 1 , R2 and R 3 is independently selected from CD 3 , CD 2 H, CDH 2 , and
  • each of R 1 , R2 and R 3 is independently CD 3 or CH 3 .
  • R 1 is CD 3 , and each of R 2 and R 3 is independently CD 3 or CH 3 . In one aspect of these embodiments, R 1 is CD 3 , and each of
  • R 2 and R 3 is CH 3 .
  • R 1 is CD 3
  • R2 is CD 3
  • R 3 is
  • R 1 is CD 3
  • R2 is CH 3 and R 3 is CD 3
  • R 1 is CD 3
  • each of R 2 and R 3 is CD 3 .
  • R 1 is CH 3 , and each of R 2 and R 3 is independently CD 3 or CH 3 . In one aspect of these embodiments, R 1 is CH 3 , and each of
  • R 2 and R 3 is CH 3 .
  • R 1 is CH 3
  • R2 is CD 3
  • R 3 is
  • R 1 is CH 3 .
  • R2 is CH 3 and R 3 is CD 3 .
  • R 1 is CH 3 , and each of R 2 and R 3 is CD 3 .
  • Y l and Y lb are the same; Y 2 and
  • Y 2b are the same; Y 3 and Y 3b are the same; each Y 4 is the same; Y 5 and Y 5b are the same; Y 7a and Y 7b are the same; Y 13a and Y 13b are the same; Y 14a and Y 14b are the same; Y 17a and Y 17b are the same; Y 18a and Y 18b are the same; and Y 19a and Y 19b are the same.
  • the present invention provides a compound of
  • each of Y la , Y lb , Y 2a , Y 2b , Y 3a , Y 3b , Y 4 , Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 , Y 9 , and Y 10 is independently selected from hydrogen and deuterium; and [00037] at least one of Y la , Y lb , Y 2a , Y 2b , Y 3a , Y 3b , Y 4 , Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 ,
  • Y 9 , or Y 10 is deuterium.
  • Y l and Y lb are the same; Y 2 and Y 2b are the same; Y 3 and Y 3b are the same; each Y 4 is the same; Y 5 and Y 5b are the same; and Y 7 and Y 7b are the same.
  • Y l and Y lb are deuterium. In one aspect of these embodiments, Y l and Y lb are deuterium; and Y 2 and Y 2b are hydrogen. In an alternate aspect of these embodiments, Y l and Y lb are deuterium; and Y 2 and Y 2b are deuterium.
  • Y l and Y lb are hydrogen. In one aspect of these embodiments, Y l and Y lb are hydrogen; and Y 2 and Y 2b are hydrogen. In an alternate aspect of these embodiments, Y l and Y lb are hydrogen; and Y 2 and Y 2b are deuterium.
  • Y 3 and Y 3b are deuterium. In one aspect of these embodiments, Y 3 and Y 3b are deuterium; and each Y 4 is hydrogen. In an alternate aspect of these embodiments, Y 3 and Y 3b are deuterium; and each Y 4 is deuterium.
  • each Y 4 is deuterium.
  • Y 3 and Y 3b are hydrogen.
  • Y 5 and Y 5b are hydrogen.
  • Y 5 and Y 5b are hydrogen; Y 6 is deuterium; and each of Y 7 and Y 7b is deuterium.
  • Y 5 and Y 5b are hydrogen; Y 6 is hydrogen; and each of Y 7 and Y 7b is deuterium.
  • Y 5 and Y 5b are hydrogen; Y 6 is deuterium; and each of Y 7 and Y 7b is hydrogen.
  • Y 5 and Y 5b are hydrogen; Y 6 is hydrogen; and each of Y 7 and Y 7b is hydrogen.
  • Y 5 and Y 5b are deuterium.
  • Y 5 and Y 5b are deuterium; Y 6 is deuterium; and each of Y 7 and Y 7b is deuterium.
  • Y 5 and Y 5b are deuterium; Y 6 is hydrogen; and each of Y 7 and Y 7b is deuterium.
  • Y 5 and Y 5b are deuterium; Y 6 is deuterium; and each of Y 7 and Y 7b is hydrogen.
  • Y 5 and Y 5b are deuterium; Y 6 is hydrogen; and each of Y 7 and Y 7b is hydrogen.
  • Y 6 is deuterium
  • Y 7 and Y 7b are hydrogen. In one aspect of these embodiments, Y 5 , Y 5b , Y 7 and Y 7b are hydrogen.
  • Y is hydrogen
  • Y 9 is deuterium. In one aspect of these embodiments, Y 6 and Y 9 are deuterium.
  • Y 10 is hydrogen
  • At least one of Y l and Y lb ; Y 2a and Y 2b ; Y 3a and Y 3b ; or each Y 4 are deuterium.
  • at least one of Y la and Y lb ; Y 2a and Y 2b ; Y 3a and Y 3b ; or each Y 4 are deuterium; and each of Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 , Y 9 , and Y 10 is hydrogen.
  • Y l , Y lb , Y 2 and Y 2b are deuterium; and each of Y 5 , Y 5b , Y 6 , Y 7 , Y 7b , Y 8 , Y 9 , and Y 10 is hydrogen.
  • Y 3 , Y 3b , and each Y 4 are deuterium; and each of Y 5a , Y 5b , Y 6 , Y 7a , Y 7b , Y 8 , Y 9 , and Y 10 is hydrogen.
  • Y 8 , and Y 10 are hydrogen.
  • Y 5a , Y 5b , Y 7a , Y 7b , Y 8 , and Y 10 are hydrogen and Y 6 is deuterium.
  • Y 5a , Y 5b , Y 7a , Y 7b , Y 8 , and Y 10 are hydrogen and Y 9 is deuterium.
  • Y 5a , Y 5b , Y 7a , Y 7b , Y 8 , and Y 10 are hydrogen; and Y la , Y lb , Y 2a and Y 2b are deuterium.
  • Y 5 , Y 5b , Y 7 , Y 7b , Y 8 , and Y 10 are hydrogen; and Y 3 , Y 3b , and each Y 4 are deuterium.
  • Y 5a , Y 5b , Y 7a , Y 7b , Y 8 , and Y 10 are hydrogen; Y l and Y lb are the same; Y 2 and Y 2b are the same; Y 3 and Y 3b are the same; each Y 4 is the same; and the compound is selected from any one of the compounds set forth in Table 1 (below):
  • any atom not designated as deuterium is present at its natural isotopic abundance.
  • the invention does not include a compound wherein each of Y la , Y lb , Y 2a , Y 2b , Y 3a , Y 3b , Y 4 , Y 5a , Y 5b , Y 6 , Y 7a , y7b ⁇ 8 ⁇ 9 ⁇ ⁇ ⁇ ⁇ 12 yl3a yl3b yl4a yl4b ⁇ 15 ⁇ 16 yl7a yl7b yl8a yl8b yl9a
  • Y 19b and Y 20 is deuterium; and each of R 1 , R 2 and R 3 is CD 3 .
  • the level of deuterium incorporation at each Y l or Y lb is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
  • the level of deuterium incorporation at each Y 2 or Y 2b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, or at least 99%.
  • the level of deuterium incorporation at each Y 3 or Y 3b is at least 52.5%, is at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
  • the level of deuterium incorporation at each Y 4 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
  • the level of deuterium incorporation at each Y 5 or Y 5b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, is at least 95%, at least 97%, or at least 99%.
  • the level of deuterium incorporation at Y 6 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, at least 99%.
  • the level of deuterium incorporation at each Y 7 or Y 7b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • the level of deuterium incorporation at Y is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • the level of deuterium incorporation at Y 9 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • the level of deuterium incorporation at Y 10 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
  • the present invention also provides deuterated intermediates useful, e.g., in the preparation of the compounds of Formula I, and as provided in the Exemplary
  • any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
  • Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure.
  • Reagents and conditions (a) MeOH; (b) TMS-C1, TEA; (c) TMS-C1, LDA; (d) BF 3 ; (e) NaOH; (f) NaOD, D 2 0, H 2 or D 2 , Pd/C; (g) NaBH 4 or NaBD 4 , NaOD.
  • esterification of the carboxylic acid moiety of 3a, 7-keto cholanic acid intermediate (1) using methanol in the presence of acid catalysis and at elevated temperature produces appropriately deuterated methyl ester intermediate (2), 3a-hydroxy moiety of which is subsequently treated with a chlorosilane such as TMS-C1 in the presence of a base such as TEA providing appropriately deuterated silyl protected intermediate (3).
  • a chlorosilane such as TMS-C1
  • a base such as TEA
  • Further treatment of intermediate (3) with a strong base such as LDA followed by treatment with a chlorosilane such as TMS-C1 produces appropriately deuterated silylenolether intermediate (4).
  • Aldol condensation reaction of enol ether intermediate (4) with appropriately deuterated aldehyde intermediate (5) at low reaction temperature produces appropriately deuterated alkylidine intermediate (6) which is comprised of a mixture of E and Z isomers.
  • Saponification of the methyl ester moiety of intermediate (6) at elevated temperature furnishes appropriately deuterated alkylidine carboxylic acid intermediate (7), which is subsequently treated with Pd/C under standard hydrogenation conditions using hydrogen or deuterium gas, followed by isomerization at elevated temperature to produce corresponding and appropriately deuterated alky intermediate (8).
  • selective reduction of the 7-keto moiety of intermediate (8) using NaBD 4 or NaBH 4 at reflux furnishes appropriately deuterated compounds of Formula I.
  • compounds of Formula I can be prepared with greater than 90% or greater than 95% or greater than 97% or greater than 99% deuterium incorporation at each position designated as D (for example, at positions Y l , y lb Y 2 ⁇ y 2b y 3a y 3b y4 ⁇ y 5a y 5b ⁇ 6 ⁇ ⁇ 7 ⁇ y 7b ⁇ 8 ⁇ ⁇ 9 ⁇ Qr ⁇ 10 ⁇ ⁇ p ormula j Qr any appropriate intermediate herein, see below for details).
  • deuterated intermediate (5) for use in the preparation of compounds of Formula I according to Scheme 1, are commercially available (e shown below) or may be prepared from corresponding deuterated reagents .
  • deuterated intermediates (5) are commercially available: Acetaldehyde-d 4 (98 atom %D) (5a), Acetaldehyde-l-di (98 atom %D) (5b), Acetaldehyde-2,2,2-d 3 (98 atom %D) (5c).
  • deuterated reagents for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods
  • deuterium incorporation at the Y 3 and Y 4 positions of intermediate (5a), (5b) or (5c) allows deuterium incorporation at the Y 3 and Y 4 positions of intermediate (5a), (5b) or (5c), e.g., 90, 95, 97, or 99% deuterium incorporation at any Y 3 and/or Y 4.
  • deuterated intermediate (1) for use in the preparation of compounds of Formula I according to Scheme 1 may be prepared from corresponding deuterated reagents exemplified in Scheme 2.
  • Appropriately deuterated 7oc-hydroxyl keto ester intermediate (9) is hydro lyzed with a base such as KOH and then deprotonated with sodium methoxide and treated with D 2 0 to afford appropriate deuterated ketone intermediate (10) which is subsequently treated with a reducing agent such as NaBH 4 in the presence of D 2 0 to furnish appropriately deuterated 3oc,7oc-dihydroxy cholanoic acid intermediate (11) in a manner analogous to a procedure described by Aragozzini, F., et al., Biochemical J. (1985), 230, 451-455.
  • Intermediate (11) is submitted to regio selective microbial oxidation using D-glucose in a manner analogous to a procedure described by Fantin, G. et al., Tetrahedron, 54(9), 1937-1942; 1998, producing appropriately deuterated dione intermediate (12).
  • Intermediate (12) is treated with methanol to produce 3-dimethyl ketal intermediate which is not isolated but directly treated with sodium and deuterated methanol, followed by reduction with reducing agent such as NaBD 4 in the presence D 2 0 to furnish appropriately deuterated 7oc-hydroxyl intermediate (13) in a manner analogous to procedures described by Fantin, G. et al., Steroids, 58(11), 524-6; 1993, and Lai, C. et al.
  • intermediate (16) is oxidized using suitable oxidizing agent such as NBS to furnish corresponding and appropriately deuterated 3oc-hydroxy-7-keto-5P-cholanic acid intermediate (1) in a manner analogous to a procedure described in CN103319560.
  • suitable oxidizing agent such as NBS
  • microbial oxidation using D-glucose may be employed to furnish intermediate (1) in a manner analogous to a procedure described by Fantin, G. et al., Tetrahedron, 54(9), 1937-1942; 1998.
  • deuterated intermediate (9), for use in the preparation of compounds of Formula I according to Scheme 1 may be prepared from corresponding deuterated reagents exemplified in Scheme 3.
  • deuterated reagents for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods
  • Appropriately deuterated starting material (20c) for use in the preparation of intermediate (18c) is prepared by treating ethyl cyanoacetate with NaBD 4 in a manner analogous to a procedure described by Van den Berg, E. et al., Synthetic Communications, 17(10), 1189-98; 1987.
  • deuterated reagents for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods
  • the invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formula A or Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier e.g., including any of the formulae herein
  • the carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • Pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates (e.g., phosphate buffered saline, etc.), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • ion exchangers e.g.,
  • the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley- Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • topical including buccal and sublingual
  • vaginal or parenteral including subcutaneous, intramuscular, intravenous and intradermal
  • the compound of the formulae herein is administered
  • transdermally e.g., using a transdermal patch or iontophoretic techniques.
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
  • Such preparative methods include the step of bringing into association with the compound to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • the compound is administered orally.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit- dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
  • Application of the subject therapeutics may be local, so as to be administered at the site of interest.
  • Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • a composition of this invention further comprises a second therapeutic agent.
  • the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties in the treatment of any of primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia or hyperlipidemia.
  • PBC primary biliary cirrhosis
  • NASH nonalcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • AH alcoholic hepatitis
  • gallstones obesity, hyper
  • Such second therapeutic agents are set forth in one or more of the following: United States patent publications US20140186438, US20140148428, US20130345188, US20110257139, and US20060252670; United States Patent Nos. US 7,994,352 and US 7,786,102; and PCT patent publication No. WO/2003080803.
  • the second therapeutic agent is an agent useful in the treatment of a disease or condition selected from primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, and obesity.
  • PBC primary biliary cirrhosis
  • NASH nonalcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • hypertension chronic diarrhea
  • bile acid malabsorption alcoholic hepatitis
  • gallstones gallstones
  • the second therapeutic agent is eicosapentanoic acid. In another embodiment, the second therapeutic agent is ursodeoxycholic acid (URSO).
  • URSO ursodeoxycholic acid
  • the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another.
  • association with one another means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • the compound of the present invention is present in an effective amount.
  • the term is a pharmaceutical composition of the invention.
  • an “effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
  • an effective amount of a compound of this invention can range from about 0.1 mg to about 50 mg/day. In more specific aspects of this embodiments, an effective amount of a compound of this invention ranges from about 0.1 mg - 25 mg/day, from about 1 mg - 50 mg/day, from about 1 mg - 25 mg/day, from about 5 mg - 25 mg/day, from about 1 mg - 5 mg/day, from about 1 mg - 10 mg/day, and from about 5 mg - 10 mg/day.
  • an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
  • an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds.,
  • the invention provides a method of activating the farnesoid X receptor (FXR) in a cell, comprising contacting a cell with one or more compounds of Formula A or Formula I herein, or a pharmaceutically acceptable salt thereof.
  • FXR farnesoid X receptor
  • the invention provides a method of treating a disease or condition selected from primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia or hyperlipidemia, in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound or a composition of this invention.
  • PBC primary biliary cirrhosis
  • NASH nonalcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • AH alcoholic hepatitis
  • AH
  • the method of this invention is used to treat a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, non-alcoholic fatty liver disease (NAFLD), chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, and obesity in a subject in need thereof.
  • PBC primary biliary cirrhosis
  • NASH non-alcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • chronic diarrhea bile acid malabsorption
  • AH alcoholic hepatitis
  • gallstones gallstones
  • the method of this invention is used to treat a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, and non-alcoholic fatty liver disease (NAFLD) in a subject in need thereof.
  • PBC primary biliary cirrhosis
  • NASH non-alcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
  • any of the above methods of treatment comprises the further step of co- administering to the subject in need thereof one or more second therapeutic agents.
  • the choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for the treatment of any of primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection,
  • PBC primary biliary cirrhosis
  • NASH non-alcoholic steatohepatitis
  • NAFLD non-alcoholic fatty liver disease
  • AH alcoholic hepatitis
  • gallstones
  • hypercholesterolemia or hyperlipidemia is also dependent upon the particular disease or condition to be treated.
  • the combination therapies of this invention include co- administering a compound of Formula A or Formula I and either
  • the invention provides a method of treating PBC, wherein the second therapeutic agent is URSO.
  • co- administered means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms.
  • the additional agent may be
  • both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods.
  • the administration of a composition of this invention, comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • the invention provides the use of a compound of Formula A or Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment in a subject of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula A or Formula I for use in the treatment in a subject of a disease, disorder or symptom thereof delineated herein.
  • Microsomal Assay Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). ⁇ -nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCl 2 ), and dimethyl sulfoxide
  • DMSO DMSO
  • 7.5 mM stock solutions of test compounds are prepared in DMSO.
  • the 7.5 mM stock solutions are diluted to 12.5-50 ⁇ in acetonitrile (ACN).
  • ACN acetonitrile
  • the 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl 2 .
  • the diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate. A 10 aliquot of the 12.5-50 ⁇ test compound is added to the
  • reaction mixtures are pre-warmed for 10 minutes. Reactions are initiated by addition of pre-warmed NADPH solution.
  • the final reaction volume is 0.5 mL and contains 0.5 mg/mL human liver microsomes, 0.25-1.0 ⁇ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl 2 .
  • the reaction mixtures are incubated at 37 °C, and 50 ⁇ aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 ⁇ of ice-cold ACN with internal standard to stop the reactions.
  • the plates are stored at 4 °C for 20 minutes after which 100 ⁇ of water is added to the wells of the plate before centrifugation to pellet precipitated proteins.
  • Supernatants are transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Bio-systems API 4000 mass spectrometer. The same procedure is followed for the non-deuterated counterpart of the compound of Formula I and the positive control, 7-ethoxycoumarin (1 ⁇ ). Testing is done in triplicate.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

This invention relates to deuterated forms of obeticholic acid, and pharmaceutically acceptable salts thereof. This invention also provides pharmaceutical compositions comprising a compound of this invention and the use of such compositions in methods of treating diseases and conditions that are beneficially treated by administering an agonist of the farnesoid X receptor (FXR).

Description

DEUTERATED OBETICHOLIC ACID
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 62/148,900, filed April 17, 2015, the contents of which are
incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
[0002] Many current medicines suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties that prevent their wider use or limit their use in certain indications. Poor ADME properties are also a major reason for the failure of drug candidates in clinical trials. While formulation technologies and prodrug strategies can be employed in some cases to improve certain ADME properties, these approaches often fail to address the underlying ADME problems that exist for many drugs and drug candidates. One such problem is rapid metabolism that causes a number of drugs, which otherwise would be highly effective in treating a disease, to be cleared too rapidly from the body. A possible solution to rapid drug clearance is frequent or high dosing to attain a sufficiently high plasma level of drug. This, however, introduces a number of potential treatment problems such as poor patient compliance with the dosing regimen, side effects that become more acute with higher doses, and increased cost of treatment. A rapidly metabolized drug may also expose patients to undesirable toxic or reactive metabolites.
[0003] Another ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites. As a result, some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent. In certain cases, modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
[0004] In some select cases, a metabolic inhibitor will be co- administered with a drug that is cleared too rapidly. Such is the case with the protease inhibitor class of drugs that are used to treat HIV infection. The FDA recommends that these drugs be co-dosed with ritonavir, an inhibitor of cytochrome P450 enzyme 3A4 (CYP3A4), the enzyme typically responsible for their metabolism (see Kempf, D.J. et al., Antimicrobial agents and chemotherapy, 1997, 41(3): 654-60). Ritonavir, however, causes adverse effects and adds to the pill burden for HIV patients who must already take a combination of different drugs. Similarly, the CYP2D6 inhibitor quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism of dextromethorphan in a treatment of pseudobulbar affect. Quinidine, however, has unwanted side effects that greatly limit its use in potential combination therapy (see Wang, L et al., Clinical Pharmacology and Therapeutics, 1994, 56(6 Pt 1): 659-67; and FDA label for quinidine at www.accessdata.fda.gov).
[0005] In general, combining drugs with cytochrome P450 inhibitors is not a satisfactory strategy for decreasing drug clearance. The inhibition of a CYP enzyme's activity can affect the metabolism and clearance of other drugs metabolized by that same enzyme. CYP inhibition can cause other drugs to accumulate in the body to toxic levels.
[0006] A potentially attractive strategy for improving a drug's metabolic properties is deuterium modification. In this approach, one attempts to slow the CYP-mediated metabolism of a drug or to reduce the formation of undesirable metabolites by replacing one or more hydrogen atoms with deuterium atoms. Deuterium is a safe, stable, nonradioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability. At the same time, because the size and shape of deuterium are essentially identical to those of hydrogen, replacement of hydrogen by deuterium would not be expected to affect the biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen.
[0007] Over the past 35 years, the effects of deuterium substitution on the rate of metabolism have been reported for a very small percentage of approved drugs (see, e.g., Blake, MI et al, J Pharm Sci, 1975, 64:367-91; Foster, AB, Adv Drug Res 1985, 14: 1-40 ("Foster"); Kushner, DJ et al, Can J Physiol Pharmacol 1999, 79-88; Fisher, MB et al, Curr Opin Drug Discov Devel, 2006, 9: 101-09 ("Fisher")). The results have been variable and unpredictable. For some compounds deuteration caused decreased metabolic clearance in vivo. For others, there was no change in metabolism. Still others demonstrated increased metabolic clearance. The variability in deuterium effects has also led experts to question or dismiss deuterium modification as a viable drug design strategy for inhibiting adverse metabolism (see Foster at p. 35 and Fisher at p. 101).
[0008] The effects of deuterium modification on a drug's metabolic properties are not predictable even when deuterium atoms are incorporated at known sites of metabolism. Only by actually preparing and testing a deuterated drug can one determine if and how the rate of metabolism will differ from that of its non-deuterated counterpart. See, for example, Fukuto et al. (J. Med. Chem. 1991, 34, 2871-76). Many drugs have multiple sites where metabolism is possible. The site(s) where deuterium substitution is required and the extent of deuteration necessary to see an effect on metabolism, if any, will be different for each drug.
SUMMARY OF THE INVENTION
[0009] This invention relates to deuterated forms of obeticholic acid, and
pharmaceutically acceptable salts thereof. In one aspect, the invention provides a
compound of Formula I:
Figure imgf000004_0001
(I), or a pharmaceutically acceptable salt thereof, wherein each of Y , Y , Y , Y , Y , Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and Y10 is independently selected from hydrogen and deuterium; and at least one of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, or Y10 is deuterium.
[00010] This invention also provides compositions comprising a compound of the invention, including pharmaceutical compositions comprising a compound of this invention and a pharmaceutically acceptable carrier. The invention also provides the use of such compounds and compositions in methods of treating diseases and conditions that are beneficially treated by administering an agonist of the farnesoid X receptor (FXR). Some exemplary embodiments include a method of treating a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL- cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia and hyperlipidemia, the method comprising the step of administering to a subject in need thereof a pharmaceutically acceptable composition of the present invention.
DETAILED DESCRIPTION OF THE INVENTION
[00011] Obeticholic acid, also known as (3a,5B,6a,7a)-6-ethyl-3,7- dihydroxycholan-24-oic acid, and as 6-ethylchenodeoxycholic acid, and as INT-747) is a potent and selective agonist of farnesoid X receptor (FXR). FXR plays an integral role in bile acid synthesis and transport and its activation regulates the expression of a number of genes involved in bile acid and cholesterol homeostasis. FXR is considered a prime target for treatment of hepatobiliary, metabolic and intestinal disorders.
Obeticholic acid has been submitted for regulatory approval in the U.S. for the treatment of primary biliary cirrhosis (PBC) as monotherapy or in combination with
ursodeoxycholic acid. Obeticholic acid is also in phase III clinical trials for treatment of non-alcoholic steatohepatitis (NASH), in phase II clinical trials for sclerosing cholangitis, diabetes and non-alcoholic fatty liver disease (NAFLD). Despite the beneficial activities of obeticholic acid, reports of high incidence of pruritus was reported in the phase II clinical trial for PBC. Seventy percent of patients receiving a 10 mg dose experience pruritus with 15% discontinuing the trial as a result. Thus, there is a continuing need for new FXR agonists to treat the aforementioned diseases and conditions.
Definitions
[00012] The term "treat" means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
[00013] "Disease" means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. [00014] It will be recognized that some variation of natural isotopic abundance occurs in a synthesized compound depending upon the origin of chemical materials used in the synthesis. Thus, a preparation of obeticholic acid will inherently contain small amounts of deuterated isotopologues. The concentration of naturally abundant stable hydrogen and carbon isotopes, notwithstanding this variation, is small and immaterial as compared to the degree of stable isotopic substitution of compounds of this invention. See, for instance, Wada, E et al., Seikagaku, 1994, 66: 15; Gannes, LZ et al., Comp Biochem Physiol Mol Integr Physiol, 1998, 119:725.
[00015] In the compounds of this invention any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Also unless otherwise stated, when a position is designated specifically as "D" or "deuterium", the position is understood to have deuterium at an abundance that is at least 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1% incorporation of deuterium).
[00016] The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
[00017] In other embodiments, a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
[00018] The term "isotopologue" refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
[00019] The term "compound," when referring to a compound of this invention, refers to a collection of molecules having an identical chemical structure, except that there may be isotopic variation among the constituent atoms of the molecules. Thus, it will be clear to those of skill in the art that a compound represented by a particular chemical structure containing indicated deuterium atoms, will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure. The relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
[00020] The invention also provides salts of the compounds of the invention.
[00021] A salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group. According to another embodiment, the compound is a pharmaceutically acceptable acid addition salt.
[00022] The term "pharmaceutically acceptable," as used herein, refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention. A "pharmaceutically acceptable counterion" is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
[00023] Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids. Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, β-hydroxybutyrate, glycolate, maleate, tartrate, methanesulfonate, propanesulfonate, naphthalene- 1-sulfonate, naphthalene-2- sulfonate, mandelate and other salts. In one embodiment,
pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
[00024] The pharmaceutically acceptable salt may also be a salt of a compound of the present invention having an acidic functional group, such as a carboxylic acid functional group, and a base. Exemplary bases include, but are not limited to, hydroxide of alkali metals including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydro xyl- substituted mono- , di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N- ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-(Ci-C6)- alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2- hydroxyethyl)amine; N-methyl-D-glucamine; morpholine; thiomorpholine; piperidine; pyrrolidine; and amino acids such as arginine, lysine, and the like.
[00025] The compounds of the present invention (e.g., compounds of Formula I), may contain an asymmetric carbon atom, for example, as the result of deuterium substitution or otherwise. As such, compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers. Accordingly, a compound of the present invention may exist as either a racemic mixture or a scalemic mixture, or as individual respective stereoisomers that are substantially free from another possible stereoisomer. The term "substantially free of other stereoisomers" as used herein means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present. Methods of obtaining or synthesizing an individual enantiomer for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates.
[00026] Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.
[00027] The term "stable compounds," as used herein, refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
[00028] "D" and "d" both refer to deuterium. "Stereoisomer" refers to both enantiomers and diastereomers. "Tert" and "t-" each refer to tertiary. "US" refers to the United States of America.
[00029] "Substituted with deuterium" refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
[00030] Throughout this specification, a variable may be referred to generally
(e.g., "each Y") or may be referred to specifically (e.g., Yl , Y4, Y6, etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
Therapeutic Compounds
[00031] The present invention provides a compound of Formula A:
Figure imgf000009_0001
pharmaceutically acceptable salt thereof, wherein:
each of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, Y10, Y11,
Y125 y13a y13b y14a y14b yl5 > γ16> γ17 y17b ylta y18b γ19 y19b ^ γ20 [& independently hydrogen or deuterium;
eeaach of R 1 , R2 and R 3 is independently selected from CD3, CD2H, CDH2, and
CH3; and when each of Y11, Y12, Y13a, Y13b, Y14a, Y14b, Y15, Y16, Y17a, Y17b, Y18a, Y18b, Y19a, Y19b and Y20 is hydrogen and each of R1, R2, and R3 is CH3, then at least one of y ylb Y2^ y2b y3a y3b γ4^ γ5^ y5b γ6 ^ y7b γ8^ γ9^ Qr γ10 ^ deuterium
[00032] In some embodiments of Formula A, each of R 1 , R2 and R 3 is independently CD3 or CH3.
[00033] In some embodiments of Formula A, R 1 is CD3, and each of R 2 and R 3 is independently CD3 or CH3. In one aspect of these embodiments, R1 is CD3, and each of
R 2 and R 3 is CH3. In one aspect of these embodiments, R 1 is CD3, R2 is CD3 and R 3 is
CH3. In one aspect of these embodiments, R 1 is CD3, R2 is CH3 and R 3 is CD3. In one aspect of these embodiments, R 1 is CD3, and each of R 2 and R 3 is CD3.
[00034] In some embodiments of Formula A, R 1 is CH3, and each of R 2 and R 3 is independently CD3 or CH3. In one aspect of these embodiments, R1 is CH3, and each of
R 2 and R 3 is CH3. In one aspect of these embodiments, R 1 is CH3, R2 is CD3 and R 3 is
CH3. In one aspect of these embodiments, R 1 is CH3, R2 is CH3 and R 3 is CD3. In one aspect of these embodiments, R 1 is CH3, and each of R 2 and R 3 is CD3.
[00035] In some embodiments of Formula A, Yl and Ylb are the same; Y2 and
Y2b are the same; Y3 and Y3b are the same; each Y4 is the same; Y5 and Y5b are the same; Y7a and Y7b are the same; Y13a and Y13b are the same; Y14a and Y14b are the same; Y17a and Y17b are the same; Y18a and Y18b are the same; and Y19a and Y19b are the same.
[00036] In some embodiments, the present invention provides a compound of
Formula I:
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein:
each of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and Y10 is independently selected from hydrogen and deuterium; and [00037] at least one of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8,
Y9, or Y10 is deuterium.
[00038] In some embodiments of Formula A and Formula I, Yl and Ylb are the same; Y2 and Y2b are the same; Y3 and Y3b are the same; each Y4 is the same; Y5 and Y5b are the same; and Y7 and Y7b are the same.
[00039] In some embodiments of Formula A and Formula I, Yl and Ylb are deuterium. In one aspect of these embodiments, Yl and Ylb are deuterium; and Y2 and Y2b are hydrogen. In an alternate aspect of these embodiments, Yl and Ylb are deuterium; and Y2 and Y2b are deuterium.
[00040] In some embodiments of Formula A and Formula I, Yl and Ylb are hydrogen. In one aspect of these embodiments, Yl and Ylb are hydrogen; and Y2 and Y2b are hydrogen. In an alternate aspect of these embodiments, Yl and Ylb are hydrogen; and Y2 and Y2b are deuterium.
[00041] In some embodiments of Formula A and Formula I, Y3 and Y3b are deuterium. In one aspect of these embodiments, Y3 and Y3b are deuterium; and each Y4 is hydrogen. In an alternate aspect of these embodiments, Y3 and Y3b are deuterium; and each Y4 is deuterium.
[00042] In some embodiments of Formula A and Formula I, each Y4 is deuterium. In one aspect of these embodiments, Y3 and Y3b are hydrogen.
[00043] In some embodiments of Formula A and Formula I, Y5 and Y5b are hydrogen. In one aspect of these embodiments, Y5 and Y5b are hydrogen; Y6 is deuterium; and each of Y7 and Y7b is deuterium. In an alternate aspect of these embodiments, Y5 and Y5b are hydrogen; Y6 is hydrogen; and each of Y7 and Y7b is deuterium. In another alternate aspect of these embodiments, Y5 and Y5b are hydrogen; Y6 is deuterium; and each of Y7 and Y7b is hydrogen. In still another alternate aspect of these embodiments, Y5 and Y5b are hydrogen; Y6 is hydrogen; and each of Y7 and Y7b is hydrogen.
[00044] In some embodiments of Formula A and Formula I, Y5 and Y5b are deuterium. In one aspect of these embodiments, Y5 and Y5b are deuterium; Y6 is deuterium; and each of Y7 and Y7b is deuterium. In an alternate aspect of these embodiments, Y5 and Y5b are deuterium; Y6 is hydrogen; and each of Y7 and Y7b is deuterium. In another alternate aspect of these embodiments, Y5 and Y5b are deuterium; Y6 is deuterium; and each of Y7 and Y7b is hydrogen. In still another alternate aspect of these embodiments, Y5 and Y5b are deuterium; Y6 is hydrogen; and each of Y7 and Y7b is hydrogen.
[00045] In some embodiments of Formula A and Formula I, Y6 is deuterium.
[00046] In some embodiments of Formula A and Formula I, Y7 and Y7b are hydrogen. In one aspect of these embodiments, Y5 , Y5b, Y7 and Y7b are hydrogen.
[00047] In some embodiments of Formula A and Formula I, Y is hydrogen.
[00048] In some embodiments of Formula A and Formula I, Y9 is deuterium. In one aspect of these embodiments, Y6 and Y9 are deuterium.
[00049] In some embodiments of Formula A and Formula I, Y10 is hydrogen.
[00050] In certain embodiments of Formula A and Formula I, at least one of Yl and Ylb; Y2a and Y2b; Y3a and Y3b; or each Y4 are deuterium. In one aspect of these embodiments, at least one of Yla and Ylb; Y2a and Y2b; Y3a and Y3b; or each Y4 are deuterium; and each of Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and Y10 is hydrogen. In one aspect of these embodiments, Yl , Ylb, Y2 and Y2b are deuterium; and each of Y5 , Y5b, Y6, Y7 , Y7b, Y8, Y9, and Y10 is hydrogen. In another aspect of these embodiments, Y3 , Y3b, and each Y4 are deuterium; and each of Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and Y10 is hydrogen.
[00051] In some embodiments of Formula A and Formula I, Y5a, Y5b, Y7a, Y7b,
Y8, and Y10 are hydrogen. In one aspect of these embodiments, Y5a, Y5b, Y7a, Y7b, Y8, and Y10 are hydrogen and Y6 is deuterium. In another aspect of these embodiments, Y5a, Y5b, Y7a, Y7b, Y8, and Y10 are hydrogen and Y9 is deuterium. In still another aspect of these embodiments, Y5a, Y5b, Y7a, Y7b, Y8, and Y10 are hydrogen; and Yla, Ylb, Y2a and Y2b are deuterium. In still another aspect of these embodiments, Y5 , Y5b, Y7 , Y7b, Y8, and Y10 are hydrogen; and Y3 , Y3b, and each Y4 are deuterium.
[00052] In one embodiment of Formula I, Y5a, Y5b, Y7a, Y7b, Y8, and Y10 are hydrogen; Yl and Ylb are the same; Y2 and Y2b are the same; Y3 and Y3b are the same; each Y4 is the same; and the compound is selected from any one of the compounds set forth in Table 1 (below):
Table 1 : Exemplary Embodiments of Formula I
Figure imgf000012_0001
Compound # yla^ylb
Figure imgf000013_0001
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
[00053] In one embodiment of a compound of Formula A, the invention does not include a compound wherein each of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, y7b γ8 γ9 γΐθ γΐ ΐ γ12 yl3a yl3b yl4a yl4b γ15 γ16 yl7a yl7b yl8a yl8b yl9a
Y19b and Y20 is deuterium; and each of R1, R2 and R3 is CD3.
[00054] In some embodiments of a compound of this invention, when Yl or Y ri1b is deuterium, the level of deuterium incorporation at each Yl or Ylb is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
[00055] In some embodiments of a compound of this invention, when Y2 or Y2b is deuterium, the level of deuterium incorporation at each Y2 or Y2b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, or at least 99%.
[00056] In some embodiments of a compound of this invention, when Y3 or Y3b is deuterium, the level of deuterium incorporation at each Y3 or Y3b is at least 52.5%, is at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
[00057] In some embodiments of a compound of this invention, when Y4 is deuterium the level of deuterium incorporation at each Y4 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, is at least 97%, or at least 99%.
[00058] In some embodiments of a compound of this invention, when Y5 or Y5b is deuterium, the level of deuterium incorporation at each Y5 or Y5b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, is at least 95%, at least 97%, or at least 99%. [00059] In some embodiments of a compound of this invention, when Y6 is deuterium, the level of deuterium incorporation at Y6 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, at least 99%.
[00060] In some embodiments of a compound of this invention, when Y7 or Y7b is deuterium, the level of deuterium incorporation at each Y7 or Y7b is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
[00061] In some embodiments of a compound of this invention, when Y is deuterium, the level of deuterium incorporation at Y is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
[00062] In some embodiments of a compound of this invention, when Y9 is deuterium, the level of deuterium incorporation at Y9 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
[00063] In some embodiments of a compound of this invention, when Y10 is deuterium, the level of deuterium incorporation at Y10 is at least 52.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, or at least 99%.
[00064] The present invention also provides deuterated intermediates useful, e.g., in the preparation of the compounds of Formula I, and as provided in the Exemplary
Schemes.
[00065] In another set of embodiments of a compound of this invention, any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
[00066] The synthesis of compounds of Formula A, Formula I and novel intermediates may be readily achieved by synthetic chemists of ordinary skill by reference to the Exemplary Synthesis and Examples disclosed herein. Relevant procedures analogous to those of use for the preparation of compounds of Formula A, Formula I and intermediates thereof are disclosed, for instance in United States patent numbers 7,138,390, 7,786,102, and 7,994,352, and United States patent publication number US20130345188.
[00067] Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure. Exemplary Synthesis
[00068] A convenient method for synthesizing compounds of Formula I is depicted in Scheme 1 below.
00069] Scheme 1: General Synthesis of Compounds of Formula I
Figure imgf000016_0001
(8) Formula I
Reagents and conditions: (a) MeOH; (b) TMS-C1, TEA; (c) TMS-C1, LDA; (d) BF3; (e) NaOH; (f) NaOD, D20, H2 or D2, Pd/C; (g) NaBH4 or NaBD4, NaOD.
[00070] In a manner analogous to a procedure described in US2013345188, esterification of the carboxylic acid moiety of 3a, 7-keto cholanic acid intermediate (1) using methanol in the presence of acid catalysis and at elevated temperature, produces appropriately deuterated methyl ester intermediate (2), 3a-hydroxy moiety of which is subsequently treated with a chlorosilane such as TMS-C1 in the presence of a base such as TEA providing appropriately deuterated silyl protected intermediate (3). Further treatment of intermediate (3) with a strong base such as LDA followed by treatment with a chlorosilane such as TMS-C1 produces appropriately deuterated silylenolether intermediate (4). Aldol condensation reaction of enol ether intermediate (4) with appropriately deuterated aldehyde intermediate (5) at low reaction temperature produces appropriately deuterated alkylidine intermediate (6) which is comprised of a mixture of E and Z isomers. Saponification of the methyl ester moiety of intermediate (6) at elevated temperature furnishes appropriately deuterated alkylidine carboxylic acid intermediate (7), which is subsequently treated with Pd/C under standard hydrogenation conditions using hydrogen or deuterium gas, followed by isomerization at elevated temperature to produce corresponding and appropriately deuterated alky intermediate (8). Finally, selective reduction of the 7-keto moiety of intermediate (8) using NaBD4 or NaBH4 at reflux furnishes appropriately deuterated compounds of Formula I.
[00071] Using commercially available reagents and deuterated reagents that can be readily prepared by known methods, compounds of Formula I can be prepared with greater than 90% or greater than 95% or greater than 97% or greater than 99% deuterium incorporation at each position designated as D (for example, at positions Yl , ylb Y2^ y2b y3a y3b y4^ y5a y5b γ6^ γ7^ y7b γ8^ γ9^ Qr γ10} ^ pormula j Qr any appropriate intermediate herein, see below for details).
[00072] Appropriately deuterated intermediate (5), for use in the preparation of compounds of Formula I according to Scheme 1, are commercially available (e shown below) or may be prepared from corresponding deuterated reagents .
Intermediate (5)
Figure imgf000017_0001
(5a): Y3= Y4= D
(5b): Y3= D; Y4= H
(5c): Y3= H; Y4= D
The following deuterated intermediates (5) are commercially available: Acetaldehyde-d4 (98 atom %D) (5a), Acetaldehyde-l-di (98 atom %D) (5b), Acetaldehyde-2,2,2-d3 (98 atom %D) (5c).
[00073] Use of appropriately deuterated reagents (for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods) allows deuterium incorporation at the Y 3 and Y 4 positions of intermediate (5a), (5b) or (5c), e.g., 90, 95, 97, or 99% deuterium incorporation at any Y 3 and/or Y 4.
[00074] Appropriately deuterated intermediate (1), for use in the preparation of compounds of Formula I according to Scheme 1 may be prepared from corresponding deuterated reagents exemplified in Scheme 2.
00075] Scheme 2: Preparation of Intermediate (1)
Figure imgf000018_0001
(9) (10)
Figure imgf000018_0002
Figure imgf000018_0003
Figure imgf000019_0001
(1)
Reactions and conditions: (a) (1) KOH, (2) D20, NaOMe; (b) NaBH4, D20; (c) KH2P04, D- Glucose; (d) (1) MeOH (2) Na, MeOD, D20, NaBD4, then HC1, pH 1 (e) TMSCHN2; (f) NaBD4, MeOD; (g) NaOH; (h) NBS, or KH2P04, D-Glucose.
[00076] Appropriately deuterated 7oc-hydroxyl keto ester intermediate (9) is hydro lyzed with a base such as KOH and then deprotonated with sodium methoxide and treated with D20 to afford appropriate deuterated ketone intermediate (10) which is subsequently treated with a reducing agent such as NaBH4 in the presence of D20 to furnish appropriately deuterated 3oc,7oc-dihydroxy cholanoic acid intermediate (11) in a manner analogous to a procedure described by Aragozzini, F., et al., Biochemical J. (1985), 230, 451-455. Intermediate (11) is submitted to regio selective microbial oxidation using D-glucose in a manner analogous to a procedure described by Fantin, G. et al., Tetrahedron, 54(9), 1937-1942; 1998, producing appropriately deuterated dione intermediate (12). Intermediate (12) is treated with methanol to produce 3-dimethyl ketal intermediate which is not isolated but directly treated with sodium and deuterated methanol, followed by reduction with reducing agent such as NaBD4 in the presence D20 to furnish appropriately deuterated 7oc-hydroxyl intermediate (13) in a manner analogous to procedures described by Fantin, G. et al., Steroids, 58(11), 524-6; 1993, and Lai, C. et al. Journal of Labelled Compounds and Radiopharmaceuticals, 21(7), 615-626; 1984. Subsequent treatment of intermediate (13) with trimethyl- silyldiazomethane produces appropriately deuterated 3-ketocholanoate intermediate (14) by analogy to a procedure described by Judkins et al., Angewandte Chemie,
International Edition, 53(8), 2110-2113; 2014, or using appropriate esterification procedures known in the art. Further treatment of intermediate (14) with NaBD4 in MeOD at low temperature produces appropriately deuterated dihydroxy ester intermediate (15) which is hydrolyzed using a base such as sodium hydroxide in methanol to afford appropriately deuterated chenodeoxycholic acid intermediate (16), in a manner analogous to a procedure described by Uekawa, T. et al., Bioscience, Biotechnology, and Biochemistry, 68(6), 1332-1337; 2004. Finally, intermediate (16) is oxidized using suitable oxidizing agent such as NBS to furnish corresponding and appropriately deuterated 3oc-hydroxy-7-keto-5P-cholanic acid intermediate (1) in a manner analogous to a procedure described in CN103319560. Alternatively, microbial oxidation using D-glucose may be employed to furnish intermediate (1) in a manner analogous to a procedure described by Fantin, G. et al., Tetrahedron, 54(9), 1937-1942; 1998.
[00077] Use of appropriately deuterated reagents (for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods) allows deuterium incorporation at the Yla, Ylb, Y2a, Y2b, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and/or Y10 positions of intermediate (1), (9), (10), (11), (12), (13), (14), (15), or (16), e.g., 90, 95, 97, or 99% deuterium incorporation at any Yla, Ylb, Y2a, Y2b, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, or Y10
[00078] Appropriately deuterated intermediate (9), for use in the preparation of compounds of Formula I according to Scheme 1 may be prepared from corresponding deuterated reagents exemplified in Scheme 3.
[00079] Scheme 3: Preparation of Intermediate (9)
Figure imgf000020_0001
(17) (19)
Figure imgf000020_0002
Reactions and conditions: (a) 6 steps: 1) Chloranil, 2) mCPBA, 3) H2, Pd/C, 4) KBH4, (5) NaH, (6) D2 or H2, Pt02; (b) (1), TMSCHN2 (2) Ag2C03-Celite.
[00080] In a manner analogous to a procedure described in CN1308085, appropriately deuterated chenodeoxycholic acid intermediate (19) is prepared in six steps from appropriately deuterated triphenylphosphonium bromide (18) and progesterone (17). Intermediate (19) is subsequently esterified with trimethyl-silyldiazo methane, followed by oxidation with silver carbonate adsorbed on celite to produce appropriately deuterated 3-ketocholanoate intermediate (9) in a manner analogous to a procedure described by Judkins et al., Angewandte Chemie, International Edition, 53(8), 2110- 2113; 2014.
[00081] Use of appropriately deuterated reagents (for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods) allows deuterium incorporation at the Yl , Ylb, Y2 , or Y2b positions of intermediates (9), (17), (18) or (19), e.g., 90, 95, 97, or 99% deuterium incorporation at any Yla, Ylb, Y2a, and/or Y -2b
[00082] Appropriately deuterated intermediate (18), for use in the preparation of compounds of Formula I according to Scheme 1 may be prepared from corresponding deuterated reagents exemplified in Scheme 4.
[00083] Scheme 4: Preparation of Intermediate (18)
Figure imgf000021_0001
(20) (21) (18)
(20a):Y1a=Y1 =Y2a=Y2 = D (18a):Y1a=Y1 =Y2a=Y2 = D
(20b):Y1a=Y1 =D; Y2a=Y2b= H (18b):Y1a=Y1 =D; Y2a=Y2b= H
(20c):Y1a=Y1 =H; Y2a=Y2b=D (18c):Y1a=Y1 =H; Y2a=Y2b=D
Reagents and conditions: (a) 48% aq. HBr; (b) Ph3P.
[00084] Thus, in a manner analogous to a procedure described by Dawadi, P. et al. Journal of Labelled Compounds and Radiopharmaceuticals, 52(9), 341-349; 2009, appropriately deuterated hydroxylnitrile intermediate (20) is refluxed with aqueous HBr to produce corresponding and appropriately deuterated bromo carboxylic acid intermediate (21). Subsequent treatment of intermediate (21) with triphenylphosphine at elevated temperature by analogy to a procedure described by Ahmed, R. et al., Journal of the American Chemical Society, 133(31), 12304-12310;2011, produces corresponding triphenylphosphonium bromide intermediate (18). The following starting materials (20) for use in the preparation of intermediates (18) are commercially available: 3-hydroxypropionitrile-2,2,3,3-d4 (98 atom% D) (20a) (or may be prepared in accordance with a procedure described by Bird, I. et al., Journal of Labelled Compounds and Radiopharmaceuticals, 27(2), 199-216;1989), Propanenitrile-2,2-d2, 3-hydroxy- (90-95 atom% D) (20b) (or prepared as described by Jarman, M. et al., Journal of Labelled Compounds and Radiopharmaceuticals, 18(4), 463-72; 1981). Appropriately deuterated starting material (20c) for use in the preparation of intermediate (18c) is prepared by treating ethyl cyanoacetate with NaBD4 in a manner analogous to a procedure described by Van den Berg, E. et al., Synthetic Communications, 17(10), 1189-98; 1987. Use of appropriately deuterated reagents (for example, commercially available reagents or deuterated reagents that can be readily prepared by known methods) allows deuterium incorporation at the Yl , Ylb, Y2 , or Y2b positions of intermediate (18), (20) or (21), e.g., 90, 95, 97, or 99% deuterium incorporation at any Yla, Ylb, Y2a, and/or Y2b.
[00085] The specific approaches and compounds shown above are not intended to be limiting. The chemical structures in the schemes herein depict variables that are hereby defined commensurately with chemical group definitions (moieties, atoms, etc.) of the corresponding position in the compound formulae herein, whether identified by the same variable name (i.e., Yla, Y4, Y6, etc.) or not. The suitability of a chemical group in a compound structure for use in the synthesis of another compound is within the knowledge of one of ordinary skill in the art.
[00086] Additional methods of synthesizing compounds of Formula A, Formula I and their synthetic precursors, including those within routes not explicitly shown in schemes herein, are within the means of chemists of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the applicable compounds are known in the art and include, for example, those described in Larock R, Comprehensive Organic
Transformations, VCH Publishers (1989); Greene, TW et al., Protective Groups in Organic Synthesis, 3 Ed., John Wiley and Sons (1999); Fieser, L et al., Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette, L, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof.
[00087] Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds.
Pharmaceutical Compositions
[00088] The invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formula A or Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier. The carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
[00089] Pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates (e.g., phosphate buffered saline, etc.), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[00090] If required, the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well- known in the art. One method includes the use of lipid excipients in the formulation. See "Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences)," David J. Hauss, ed. Informa Healthcare, 2007; and "Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley- Interscience, 2006.
[00091] Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROL™ and PLURONIC™ (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
[00092] The pharmaceutical compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. In certain embodiments, the compound of the formulae herein is administered
transdermally (e.g., using a transdermal patch or iontophoretic techniques). Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
[00093] Such preparative methods include the step of bringing into association with the compound to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
[00094] In certain embodiments, the compound is administered orally.
Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc. Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
[00095] In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
[00096] Compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
[00097] Compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit- dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
[00098] Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
[00099] The pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
[000100] The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
[000101] Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For topical application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
[000102] Application of the subject therapeutics may be local, so as to be administered at the site of interest. Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
[000103] In another embodiment, a composition of this invention further comprises a second therapeutic agent. The second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties in the treatment of any of primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia or hyperlipidemia. Such second therapeutic agents are set forth in one or more of the following: United States patent publications US20140186438, US20140148428, US20130345188, US20110257139, and US20060252670; United States Patent Nos. US 7,994,352 and US 7,786,102; and PCT patent publication No. WO/2003080803.
[000104] Preferably, the second therapeutic agent is an agent useful in the treatment of a disease or condition selected from primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, and obesity.
[000105] In one embodiment, the second therapeutic agent is eicosapentanoic acid. In another embodiment, the second therapeutic agent is ursodeoxycholic acid (URSO).
[000106] In another embodiment, the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another. The term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
[000107] In the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term
"effective amount" refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
[000108] The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., Cancer Chemother. Rep, 1966, 50: 219. Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy
Pharmaceuticals, Ardsley, N.Y., 1970, 537.
[000109] In one embodiment, an effective amount of a compound of this invention can range from about 0.1 mg to about 50 mg/day. In more specific aspects of this embodiments, an effective amount of a compound of this invention ranges from about 0.1 mg - 25 mg/day, from about 1 mg - 50 mg/day, from about 1 mg - 25 mg/day, from about 5 mg - 25 mg/day, from about 1 mg - 5 mg/day, from about 1 mg - 10 mg/day, and from about 5 mg - 10 mg/day.
[000110] Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of
administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
[000111] For pharmaceutical compositions that comprise a second therapeutic agent, an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent. Preferably, an effective amount is between about 70% and 100% of the normal monotherapeutic dose. The normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds.,
Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn.
(2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
[000112] It is expected that some of the second therapeutic agents referenced above will act synergistically with the compounds of this invention. When this occurs, it will allow the effective dosage of the second therapeutic agent and/or the compound of this invention to be reduced from that required in a monotherapy. This has the advantage of minimizing toxic side effects of either the second therapeutic agent of a compound of this invention, synergistic improvements in efficacy, improved ease of administration or use and/or reduced overall expense of compound preparation or formulation.
Methods of Treatment
[000113] In another embodiment, the invention provides a method of activating the farnesoid X receptor (FXR) in a cell, comprising contacting a cell with one or more compounds of Formula A or Formula I herein, or a pharmaceutically acceptable salt thereof.
[000114] According to another embodiment, the invention provides a method of treating a disease or condition selected from primary biliary cirrhosis (PBC), nonalcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection, hypercholesterolemia or hyperlipidemia, in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound or a composition of this invention.
[000115] In one particular embodiment, the method of this invention is used to treat a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, non-alcoholic fatty liver disease (NAFLD), chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, and obesity in a subject in need thereof.
[000116] In another particular embodiment, the method of this invention is used to treat a disease or condition selected from primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, and non-alcoholic fatty liver disease (NAFLD) in a subject in need thereof.
[000117] Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
[000118] In another embodiment, any of the above methods of treatment comprises the further step of co- administering to the subject in need thereof one or more second therapeutic agents. The choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for the treatment of any of primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease (NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection,
hypercholesterolemia or hyperlipidemia. The choice of second therapeutic agent is also dependent upon the particular disease or condition to be treated.
[000119] In some embodiments, the combination therapies of this invention include co- administering a compound of Formula A or Formula I and either
eicosapentanoic acid or ursodeoxycholic acid (URSO) to a subject in need thereof. In one aspect of these embodiments, the invention provides a method of treating PBC, wherein the second therapeutic agent is URSO.
[000120] The term "co- administered" as used herein means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be
administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods. The administration of a composition of this invention, comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
[000121] Effective amounts of these second therapeutic agents are well known to those skilled in the art and guidance for dosing may be found in patents and published patent applications referenced herein, as well as in Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR
Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), and other medical texts. However, it is well within the skilled artisan's purview to determine the second therapeutic agent's optimal effective-amount range.
[000122] In one embodiment of the invention, where a second therapeutic agent is administered to a subject, the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
[000123] In yet another aspect, the invention provides the use of a compound of Formula A or Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment in a subject of a disease, disorder or symptom set forth above. Another aspect of the invention is a compound of Formula A or Formula I for use in the treatment in a subject of a disease, disorder or symptom thereof delineated herein. Example 1. Evaluation of Metabolic Stability
[000124] Microsomal Assay : Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). β-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCl2), and dimethyl sulfoxide
(DMSO) are purchased from Sigma- Aldrich.
[000125] Determination of Metabolic Stability: 7.5 mM stock solutions of test compounds are prepared in DMSO. The 7.5 mM stock solutions are diluted to 12.5-50 μΜ in acetonitrile (ACN). The 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl2. The diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate. A 10 aliquot of the 12.5-50 μΜ test compound is added to the
microsomes and the mixture is pre-warmed for 10 minutes. Reactions are initiated by addition of pre-warmed NADPH solution. The final reaction volume is 0.5 mL and contains 0.5 mg/mL human liver microsomes, 0.25-1.0 μΜ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl2. The reaction mixtures are incubated at 37 °C, and 50 μί aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 μί of ice-cold ACN with internal standard to stop the reactions. The plates are stored at 4 °C for 20 minutes after which 100 μί of water is added to the wells of the plate before centrifugation to pellet precipitated proteins. Supernatants are transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Bio-systems API 4000 mass spectrometer. The same procedure is followed for the non-deuterated counterpart of the compound of Formula I and the positive control, 7-ethoxycoumarin (1 μΜ). Testing is done in triplicate.
[000126] Data analysis: The in vitro ti/2s for test compounds are calculated from the slopes of the linear regression of % parent remaining (In) vs incubation time relationship.
in vitro t ½ = 0.693/k
k = -[slope of linear regression of % parent remaining (In) vs incubation time]
[000127] Data analysis is performed using Microsoft Excel Software. [000128] Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. It should be understood that the foregoing discussion and examples merely present a detailed description of certain preferred embodiments. It will be apparent to those of ordinary skill in the art that various modifications and equivalents can be made without departing from the spirit and scope of the invention.

Claims

A compound of Formula I:
Figure imgf000033_0001
(I), or a pharmaceutically acceptable salt thereof, wherein:
each of Yla, Ylb, Y2a, Y2b, Y3a, Y b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, and Y10 is independently selected from hydrogen and deuterium; and
at least one of Yla, Ylb, Y2a, Y2b, Y3a, Y3b, Y4, Y5a, Y5b, Y6, Y7a, Y7b, Y8, Y9, or Y10 is deuterium.
2. The compound of claim 1, wherein:
Yla and Ylb are the same;
Y2a and Y2b are the same;
Y3 and Y3b are the same;
each Y4 is the same;
Y5a and Y5b are the same; and
Y7a and Y7b are the same.
3. The compound of claim 1 or 2, wherein Yl and Ylb are deuterium; and Y2 and Y2b are hydrogen.
4. The compound of claim 1 or 2, wherein Yla and Ylb are deuterium; and Y2 and Y2b are deuterium.
5. The compound of claim 1 or 2, wherein Yl and Ylb are hydrogen; and Y2 and Y2b are hydrogen.
6. The compound of claim 1 or 2, wherein, Yl and Ylb are hydrogen; and Y2 and Y2b are deuterium.
7. The compound of any one of claims 1-6, wherein Y3 and Y3b are deuterium; and each Y4 is hydrogen.
8. The compound of any one of claims 1-6, wherein Y3 and Y3b are deuterium; and each Y4 is deuterium.
9. The compound of any one of claims 1-6, wherein Y3 and Y3b are hydrogen; and each Y4 is hydrogen.
10. The compound of any one of claims 1-6, wherein Y3 and Y3b are hydrogen; and each Y4 is deuterium.
11. The compound of any one of claims 1-10, wherein Y6 is deuterium.
12. The compound of any one of claims 1-11, wherein Y9 is deuterium.
13. The compound of any one of claims 1-12, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
14. The compound of claim 1, wherein Y5a, Y5b, Y7a, Y7b, Y8, and Y10 are hydrogen; Yl and Ylb are the same; Y2 and Y2b are the same; Y3 and Y3b are the same; each Y4 is the same; and the compound is selected from any one of the compounds set forth in the table below:
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
15. A pharmaceutically acceptable composition comprising a compound of any one of claims 1-14, and a pharmaceutically acceptable carrier.
16. A method of treating a disease or condition selected from the group consisting of primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, diabetic hepatic disease, non-alcoholic fatty liver disease
(NAFLD), hypertension, chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, obesity, hypertriglyceridemia, low HDL-cholesterol, arteriosclerosis, atherosclerosis, cholestasis, fibrosis, hepatic Hepatitis C viral infection,
hypercholesterolemia and hyperlipidemia, comprising the step of administering to a subject in need thereof the composition of claim 15.
17. The method of claim 16, wherein the disease or condition is selected from the group consisting of primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, diabetes, non-alcoholic fatty liver disease (NAFLD), chronic diarrhea, bile acid malabsorption, alcoholic hepatitis (AH), gallstones, and obesity.
18. The method of claim 17, wherein the disease or condition is selected from the group consisting of primary biliary cirrhosis (PBC), non-alcoholic steatohepatitis (NASH), sclerosing cholangitis, and non-alcoholic fatty liver disease (NAFLD).
19. The method of any one of claims 16-18, wherein the subject is suffering from PBC, comprising the additional step of co-administering ursodeoxycholic acid to the subject in need thereof.
PCT/US2016/027688 2015-04-17 2016-04-15 Deuterated obeticholic acid WO2016168553A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562148900P 2015-04-17 2015-04-17
US62/148,900 2015-04-17

Publications (2)

Publication Number Publication Date
WO2016168553A1 true WO2016168553A1 (en) 2016-10-20
WO2016168553A8 WO2016168553A8 (en) 2016-11-24

Family

ID=57127023

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/027688 WO2016168553A1 (en) 2015-04-17 2016-04-15 Deuterated obeticholic acid

Country Status (1)

Country Link
WO (1) WO2016168553A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106645497A (en) * 2017-01-03 2017-05-10 山东省药学科学院 Obeticholic acid and detection method for related substances in preparation of obeticholic acid
WO2019023103A1 (en) * 2017-07-24 2019-01-31 Intercept Pharmaceuticals, Inc. Isotopically labeled bile acid derivatives
WO2020030737A1 (en) 2018-08-10 2020-02-13 Phenex Pharmaceuticals Ag Isolithocholic acid or isoallolithocholic acid and deuterated derivatives thereof for preventing and treating clostridium difficile-associated diseases
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
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

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130345188A1 (en) * 2012-06-19 2013-12-26 Intercept Pharmaceuticals, Inc. Preparation and Uses of Obeticholic Acid
US20140206657A1 (en) * 2013-01-18 2014-07-24 City Of Hope Bile acid analog tgr5 agonists
US20140371190A1 (en) * 2013-05-14 2014-12-18 TES Pharma SrI. Farnesoid X receptor modulators
US20150112089A1 (en) * 2013-10-22 2015-04-23 Metselex, Inc. Deuterated bile acids

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130345188A1 (en) * 2012-06-19 2013-12-26 Intercept Pharmaceuticals, Inc. Preparation and Uses of Obeticholic Acid
US20140206657A1 (en) * 2013-01-18 2014-07-24 City Of Hope Bile acid analog tgr5 agonists
US20140371190A1 (en) * 2013-05-14 2014-12-18 TES Pharma SrI. Farnesoid X receptor modulators
US20150112089A1 (en) * 2013-10-22 2015-04-23 Metselex, Inc. Deuterated bile acids

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106645497A (en) * 2017-01-03 2017-05-10 山东省药学科学院 Obeticholic acid and detection method for related substances in preparation of obeticholic acid
WO2019023103A1 (en) * 2017-07-24 2019-01-31 Intercept Pharmaceuticals, Inc. Isotopically labeled bile acid derivatives
CN111050772A (en) * 2017-07-24 2020-04-21 英特塞普特医药品公司 Isotopically labelled bile acid derivatives
US11472831B2 (en) 2017-07-24 2022-10-18 Intercept Pharmaceuticals, Inc. Isotopically labeled bile acid derivatives
WO2020030737A1 (en) 2018-08-10 2020-02-13 Phenex Pharmaceuticals Ag Isolithocholic acid or isoallolithocholic acid and deuterated derivatives thereof for preventing and treating clostridium difficile-associated diseases
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
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

Also Published As

Publication number Publication date
WO2016168553A8 (en) 2016-11-24

Similar Documents

Publication Publication Date Title
WO2016168553A1 (en) Deuterated obeticholic acid
AU2013296627B2 (en) Deuterated ibrutinib
US8471034B2 (en) Niacin prodrugs and deuterated versions thereof
AU2014235462B2 (en) Deuterated palbociclib
US8575221B2 (en) Derivatives of dimethylcurcumin
WO2012151361A1 (en) Carbamoylpyridone derivatives
EP2872159A2 (en) Deuterated carfilzomib
WO2011140078A1 (en) Synthetic triterpenoid derivatives
EP2935251A1 (en) Deuterated alk inhibitors
WO2014110189A1 (en) Deuterated momelotinib
WO2011017612A1 (en) Substituted diphenylpyrazine derivatives
WO2018005328A1 (en) Deuterated bictegravir
WO2009128947A1 (en) Piperazine derivatives
EP2968268A1 (en) Inhibitors of the enzyme udp-glucose: n-acyl-sphingosine glucosyltransferase
WO2012154728A1 (en) Deuterated n-butyl bumetanide
WO2016105547A1 (en) Deuterated dasabuvir
WO2015009889A1 (en) Deuterated intedanib derivatives and their use for the treatment of proliferative disorders
WO2014159511A1 (en) Deuterated pacritinib
US20190367438A1 (en) Deuterated idebenone
WO2011159920A1 (en) [5,6]-dihydro-2h-pyran-2-one derivatives
WO2014152275A1 (en) Deuterium modified derivatives of the ns5b polymerase inhibitor tmc647055
US20110201678A1 (en) Xanthenone-4-Acetic Acid Derivatives
US9181190B2 (en) Deuterated vercirnon
EP2804857A1 (en) Deuterated alpha-lipoic acid
WO2014150044A1 (en) Amine reuptake inhibitors

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: 16780806

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16780806

Country of ref document: EP

Kind code of ref document: A1