EP3394081A1 - Formes cristallines polymorphes de l'acide obéticholique - Google Patents

Formes cristallines polymorphes de l'acide obéticholique

Info

Publication number
EP3394081A1
EP3394081A1 EP16714049.0A EP16714049A EP3394081A1 EP 3394081 A1 EP3394081 A1 EP 3394081A1 EP 16714049 A EP16714049 A EP 16714049A EP 3394081 A1 EP3394081 A1 EP 3394081A1
Authority
EP
European Patent Office
Prior art keywords
obeticholic acid
acid
crystalline
cholan
hydroxy
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP16714049.0A
Other languages
German (de)
English (en)
Inventor
André STEINER
Heidi Waenerlund Poulsen
Emilie JOLIBOIS
Melissa REWOLINSKI
Ralf Gross
Emma Sharp
Fiona DUBAS-FISHER
Alex Eberlin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Intercept Pharmaceuticals Inc
Original Assignee
Intercept 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=55650649&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP3394081(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US14/979,005 external-priority patent/US9982008B2/en
Application filed by Intercept Pharmaceuticals Inc filed Critical Intercept Pharmaceuticals Inc
Publication of EP3394081A1 publication Critical patent/EP3394081A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J17/00Normal steroids containing carbon, hydrogen, halogen or oxygen, having an oxygen-containing hetero ring not condensed with 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
    • C07B2200/13Crystalline forms, e.g. polymorphs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J51/00Normal steroids with unmodified cyclopenta(a)hydrophenanthrene skeleton not provided for in groups C07J1/00 - C07J43/00

Definitions

  • the present invention relates to obeticholic acid, an agonist for FXR, processes of preparation for obeticholic acid, pharmaceutical formulations comprising obeticholic acid, and the therapeutic use of the same.
  • the present invention relates to a process for the preparation of obeticholic acid using crystalline obeticholic acid as a synthetic intermediate.
  • the crystalline obeticholic acid is selected from the group consisting of Forms A, C, D, F, G, and I.
  • the present invention further relates to a process for the selective preparation of crystalline obeticholic acid.
  • the present invention relates to a crystalline obeticholic acid Form A
  • the crystalline obeticholic acid Form A is characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 41.
  • the present invention relates to a crystalline obeticholic acid Form C
  • the crystalline obeticholic acid Form C is characterized by an X-ray diffraction pattern substantially similar to that set forth in Figure 5 and further characterized by a Differential Scanning Calorimetry (DSC) thermogram having an endotherm value at about 98 ⁇ 2 °C.
  • DSC Differential Scanning Calorimetry
  • the present invention relates to a crystalline obeticholic acid Form D
  • the crystalline obeticholic acid Form D is characterized by an X-ray diffraction pattern substantially similar to that set forth in Figure 45.
  • the present invention relates to a crystalline obeticholic acid Form F
  • the crystalline obeticholic acid Form F is characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 49.
  • the present invention relates to a crystalline obeticholic acid Form G
  • the crystalline obeticholic acid Form G is characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 53.
  • the present invention relates to a crystalline obeticholic acid Form I characterized by an X-ray diffraction partem including a characteristic peak at about 7.2 degrees 2- Theta.
  • the crystalline obeticholic acid Form I is characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 57.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the step of converting crystalline obeticholic acid to obeticholic acid Form 1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid with NaBFU to form crystalline obeticholic acid and converting crystalline obeticholic acid to obeticholic acid Form 1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan- 24-oic acid with Pd/C and hydrogen gas to form 3a-hydroxy-6a-ethyl-7-keto-5p-cholan- 24-oic acid; reacting 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid with NaBFU to form crystalline obeticholic acid; and converting crystalline obeticholic acid to obeticholic acid Form 1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan- 24-oic acid methyl ester with NaOH to form E- or E/Z-3a-hydroxy-6-ethylidene-7-keto- 5p-cholan-24-oic acid; reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24- oic acid with Pd/C and hydrogen gas to form 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24- oic acid; reacting 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid with NaBFU to form crystalline obeticholic acid, and converting crystalline obeticholic acid to obeticholic acid Form 1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting 3a,7-ditrimethylsilyloxy-5p-chol-6-en-24-oic acid methyl ester with CH3CHO to form E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid methyl ester; reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid methyl ester with NaOH to form E- or E/Z-3a-hydroxy-6-ethylidene- 7-keto-5p-cholan-24-oic acid; reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid with Pd/C and hydrogen gas to form 3a-hydroxy-6a-ethyl-7-keto-5p- cholan-24-oic acid; reacting 3a
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester with Li[N(CH(CH 3 ) 2 ) 2 ] and Si(CH 3 ) 3 Cl to form 3a,7-ditrimethylsilyloxy-5p-chol-6-en- 24-oic acid methyl ester; reacting 3a,7-ditrimethylsilyloxy-5p-chol-6-en-24-oic acid methyl ester with CH3CHO to form E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid methyl ester; reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid methyl ester with NaOH to form E- or E/Z-3a-hydroxy-6-ethylidene- 7-keto-5p-cholan
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of reacting 3a-hydroxy-7-keto-5p-cholan-24-oic acid with CH3OH and H2SO4 to form 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester; reacting 3a- hydroxy-7-keto-5p-cholan-24-oic acid methyl ester with Li[N(CH(CH3)2)2] and
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form A to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form A in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form C to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form C in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form D to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form D in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form F to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form F in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form G to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form G in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein converting crystalline obeticholic acid Form I to obeticholic acid Form 1 comprises the step of dissolving crystalline obeticholic acid Form I in aqueous NaOH solution and adding HC1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid with NaBH4 to form crystalline obeticholic acid is carried out at a temperature at about 85 °C to about 110 °C in a basic aqueous solution.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid with Pd/C and hydrogen gas to form 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid is carried out at a temperature at about 20 °C to about 105 °C and at a pressure at about 0.5 to about 5 bars.
  • the hydrogenation is performed at a temperature range from about 20 °C to about 105 °C, e.g.
  • the hydrogenation is performed at a pressure at about 0.5 bar to about 5.5 bar, e.g., 0.6 bar, 0.7 bar, 0.8 bar, 0.9 bar, 1.0 bar, 1.2 bar, 1.4 bar, 1.6 bar, 1.8 bar, 2.0 bar, 2.2 bar, 2.4 bar, 2.6 bar, 2.8 bar, 3.0 bar, 3.2 bar, 3.4 bar, 3.6 bar, 3.8 bar, 4.0 bar, 4.2 bar, 4.4 bar, 5.0 bar, 5.2 bar, 5.3 bar, and 5.4 bar, as well as any increment in between.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester with NaOH to form E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid is carried out at a temperature at about 20 °C to about 60 °C.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting 3a,7-ditrimethylsilyloxy-5p-chol-6-en-24-oic acid methyl ester with CH3CHO to form E- or E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester is carried out in a polar aprotic solvent at a temperature at about -50 °C to about -70 °C in the presence of BF3. In one embodiment, the reaction is carried out at a temperature range of below -60 °C to about -70 °C in the presence of BF3. In one embodiment, the reaction is carried out at is carried out at an upper limit temperature of about -60 °C in the presence of BF3.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester with
  • Li[N(CH(CH 3 ) 2 ) 2 ] and Si(CH 3 )3Cl to form 3a,7-ditrimethylsilyloxy-5p-chol-6-en-24-oic acid methyl ester is carried out in a polar aprotic solvent at a temperature at about -10 °C to about -30 °C.
  • the present invention relates to a process for preparing obeticholic acid Form 1, wherein reacting 3a-hydroxy-7-keto-5p-cholan-24-oic acid with CH3OH and H2SO4 to form 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester is heated for about 3 hours and the pH of the reaction mixture is adjusted with an aqueous basic solution to a pH- range of about 9.5 to about 10.
  • the present invention relates to a obeticholic acid, or a pharmaceutically acceptable salt, solvate or amino acid conjugate thereof, having a potency of greater than about 98%, greater than about 98.5%, greater than about 99.0%, or greater than about 99.5%.
  • the present invention relates to a pharmaceutical composition comprising obeticholic acid Form 1 produced by a process of the invention and a pharmaceutically acceptable carrier.
  • the present invention relates to a method of treating or preventing an FXR mediated disease or condition in a subject comprise of administering an effective amount of obeticholic acid Form 1.
  • the disease or condition is selected from biliary atresia, cholestatic liver disease, chronic liver disease, nonalcoholic steatohepatitis (NASH), hepatitis C infection, alcoholic liver disease, primary biliary cirrhosis (PBC), liver damage due to progressive fibrosis, liver fibrosis, and cardiovascular diseases including atherosclerosis, arteriosclerosis, hypercholesterolemia, and hyperlipidemia.
  • the present invention relates to a method for lowering triglycerides in a subject comprise of administering an effective amount of obeticholic acid Form 1.
  • Example 1 injected at 1 mg/mL, injection volume 3 ⁇ 1.
  • the chromatogram is obtained according to the method described in Example 2.
  • the chromatogram is obtained according to the method described in Example
  • Example 2 is a UV chromatogram of crude compound 5 of step 4 of Example 1 using HPLC method. The chromatogram is obtained according to the method described in Example 2.
  • Figure 5 is an XRPD diffractogram of crystalline obeticholic acid Form C (see
  • Figure 6 shows TGA and DSC Thermograms of crystalline obeticholic acid Form C
  • Figure 7 shows VT-XRPD diffractograms of crystalline obeticholic acid at 25 °C
  • Figure 8A is a GVS isotherm plot of crystalline obeticholic acid Form C (see
  • Figure 8B is a GVS kinetic plot of crystalline obeticholic acid Form C (see Example
  • Figure 8C shows XRPD diffractograms of crystalline obeticholic acid Form C before and after GVS analysis (see Example 3).
  • Figure 9 shows XRPD diffractograms of crystalline obeticholic acid Form C before and after storage at 40 °C/75% RH (see Example 3).
  • Figure 10 is an XRPD diffractogram of batch 1 of obeticholic acid Form l(see
  • Figure 11 shows the XRPD diffractorgraphs for batches 1, 2, 3, 4, 5 and 6 of
  • Figure 12 is a NMR spectrum of batch 1 of obeticholic acid Form 1 in c e-DMSO (see
  • Figure 13 shows the 3 ⁇ 4 NMR spectra for batches 1, 2, 3, 4, 5 and 6 of obeticholic acid Form 1 (see Example 5).
  • Figure 14 is an expansion of 1 C DEPTQ NMR spectrum of obeticholic acid Form 1 from region 10-75 ppm (see Example 5).
  • Figure 15 is an expansion of 1 C DEPT135 NMR spectrum of obeticholic acid Form
  • Figure 16 is a quantitative 1 C NMR of obeticholic acid Form 1 (see Example 5).
  • Figure 17 is an expanded view of peaks at 32.3 ppm of Figure 16 (see Example 5).
  • Figure 18 is a FT-IR spectrum of batch 1 of obeticholic acid Form 1 (see Example 5).
  • Figure 19 shows TGA and DSC thermograms of batch 1 of obeticholic acid Form 1
  • Figure 20 shows modulated DSC thermograms of batch 1 of obeticholic acid Form 1
  • Figure 21 shows the TGA traces of batches 1, 2, 3, 4, 5, and 6 of obeticholic acid
  • Figure 22 shows the DSC traces of batches 1, 2, 3, 4, 5, and 6 of obeticholic acid
  • Figure 23 A is a picture of batch 1 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 23B is a picture of batch 2 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 23C is a picture of batch 3 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 23D is a picture of batch 4 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 23E is a picture of batch 5 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 23F is a picture of batch 6 of obeticholic acid Form 1 under polarized light microscopy.
  • Figure 24 shows GVS isotherm plot of batch 1 of obeticholic acid Form 1 (see
  • Figure 25 shows GVS kinetics plot of batch 1 of obeticholic acid Form 1 (see
  • Figure 26 shows XRPD diffractograms of batch 1 of obeticholic acid Form 1 before and after GVS (see Example 5).
  • Figure 27 is a graph of the measurement of pKa at three different methanol/water ratios for obeticholic acid Form 1 (see Example 5).
  • Figure 28 is a Yasuda-Shedlovsky plot for obeticholic acid Form 1 (see Example 5).
  • Figure 29 is a graph showing the distribution of the species depending on pH for obeticholic acid Form 1 (see Example 5).
  • Figure 30 is a graph showing the difference curve obtained by potentiometry for obeticholic acid Form 1 (see Example 5).
  • Figure 31 shows the lipophilicity profile of obeticholic acid Form 1 (see Example 5).
  • Figure 32 shows the XRPD diffractograms of batch 1 of obeticholic acid Form 1 after storage at 40 °C/75% RH (see Example 5).
  • Figure 33 shows the XRPD diffractograms of batch 1 of obeticholic acid Form 1 after storage at 25 °C/97% RH (see Example 5).
  • Figure 34 shows a view of the molecule of obeticholic acid Form G from the crystal structure showing anisotropic atomic displacement ellipsoids for the non- hydrogen atoms at the 50% probability level (see Example 6).
  • Figure 35 shows a view of the intermolecular hydrogen bonds of the crystal structure of obeticholic acid Form G where hydrogen bondings are shown in dashed lines (See Example 6).
  • Figure 36 shows an XRPD overlay of the simulated powder pattern, experimental patterns of the collected crystal, and obeticholic acid Form G (see Example
  • Figure 37 shows a graph of the plasma obeticholic acid profile vs. time after oral administration of 20 mg/kg of obeticholic acid Form 1 and crystalline Form F (see Example 7).
  • Figure 38 shows a graph of the plasma concentration of tauro conjugate of
  • Figure 39 shows the DSC curve of Form 1 (see Example 7).
  • Figure 40 shows the DSC curve of Form F (see Example 7).
  • Figure 41 is an XRPD diffractogram of crystalline obeticholic acid Form A (see example 3).
  • Figure 42 is an overlay of TGA and DSC thermograms of crystalline obeticholic acid
  • Figure 43 is an isotherm plot for the GVS experiment with Form A (see example 3).
  • Figure 44 is a kinetics plot for the GVS experiment with Form A (see example 3).
  • obeticholic acid a pharmaceutically active ingredient (also known as INT-747) having the chemical structure:
  • substantially pure obeticholic acid including, substantially pure obeticholic acid, a process for the preparation of obeticholic acid comprising crystalline obeticholic acid as a synthetic intermediate, and analytical methods for confirming the presence and purity of obeticholic acid and synthetic intermediates in the process to prepare obeticholic acid.
  • the present application also describes pharmaceutical compositions and formulations of obeticholic acid and uses for such compositions.
  • the present application is directed to a process for preparing highly pure obeticholic acid.
  • the process of the present application is shown in Scheme 1.
  • the process is a 6-step synthesis followed by one purification step to produce highly pure obeticholic acid.
  • the process of the present invention also includes a process according to Scheme 1 where compounds 4 and 5 are each comprised of a mixture of the E and Z isomers as illustrated by the structures of compounds 4A and 5A below:
  • the E/Z isomer ratio of E/Z-3a-hydroxy-6-ethylidene-7-keto- 5p-cholan-24-oic acid methyl ester (4A) is about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 83%, greater than about 85%, greater than about 90%, greater than about 93%, greater than about 95%, or greater than about 99%.
  • the E/Z ratio is determined by HPLC.
  • the ratio is greater than about 80%. In one embodiment, the ratio is greater than about 83%. In one embodiment, the ratio is greater than about 85%. In one embodiment, the ratio is greater than about 90%. In one embodiment, the ratio is greater than about 93%. In one embodiment, the ratio is greater than about 95%. In one embodiment, the ratio is greater than about 99%.
  • the E/Z isomer ratio of E/Z-3a-hydroxy-6-ethylidene-7-keto- 5p-cholan-24-oic acid (5 A) is about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 83%, greater than about 85%, greater than about 90%, greater than about 93%, greater than about 95%, or greater than about 99%.
  • the E/Z ratio is determined by HPLC. In one embodiment, the ratio is greater than about 80%. In one embodiment, the ratio is greater than about 83%. In one embodiment, the ratio is greater than about 85%. In one embodiment, the ratio is greater than about 90%. In one embodiment, the ratio is greater than about 93%. In one embodiment, the ratio is greater than about 95%. In one embodiment, the ratio is greater than about 99%.
  • Step 1 is the esterification of the C-24 carboxylic acid of 7-ketolithocholic acid (KLCA) using methanol in the presence of an acid catalyst and heat to produce the methyl ester compound 1.
  • Step 2 is silylenol ether formation from compound 1 using chlorosilane in the presence of a strong base to produce compound 3.
  • Step 3 is an acid mediated aldol condensation reaction of the silylenol ether compound 3 and acetaldehyde in the presence of an acid to produce compound 4 (or compound 4A).
  • Step 4 is ester hydrolysis i.e., saponification of the C-24 methyl ester of compound 4 (or compound 4A) to produce the carboxylic acid compound 5 (or compound 5A).
  • Step 5 is the hydrogenation of the 6-ethylidene moiety of compound 5 (or compound 5A) followed by isomerization of the initially formed 6- ⁇ ethyl group to produce compound 6.
  • Step 6 is the selective reduction of the 7-keto group of compound 6 to a 7 -hydroxy group to produce crystalline obeticholic acid.
  • Step 7 is the conversion of crystalline obeticholic acid to obeticholic acid Form 1.
  • the process of the present invention relates to a process for preparing obeticholic acid Form 1, where the process utilizes a crystalline form of obeticholic acid as a synthetic intermediate.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the step of converting crystalline obeticholic acid to obeticholic acid Form 1.
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1, comprising the steps of
  • the present invention relates to a process for preparing obeticholic acid Form 1 using crystalline obeticholic acid as a synthetic intermediate.
  • the crystalline obeticholic acid is Form A.
  • the crystalline obeticholic acid Form A is characterized by an X-ray diffraction pattern similar to that set forth in Figure 41.
  • the crystalline obeticholic acid Form A is crystallized and recrystallized from n-butyl acetate.
  • the crystalline obeticholic acid is Form C. In one embodiment, the crystalline obeticholic acid Form C is characterized by an X-ray diffraction pattern similar to that set forth in Figure 5. In another embodiment, the crystalline obeticholic acid is Form D. In one embodiment, the crystalline obeticholic acid Form D is characterized by an X-ray diffraction pattern similar to that set forth in Figure 45. In another embodiment, the crystalline obeticholic acid is Form F. In one embodiment, the crystalline obeticholic acid Form F is characterized by an X-ray diffraction pattern similar to that set forth in Figure 49. In another embodiment, the crystalline obeticholic acid is Form G.
  • the crystalline obeticholic acid Form G is characterized by an X-ray diffraction pattern similar to that set forth in Figure 53.
  • the crystalline obeticholic acid is Form I.
  • the crystalline obeticholic acid Form I is characterized by an X-ray diffraction pattern similar to that set forth in Figure 57.
  • Step 1 is the reaction of 3a-hydroxy-7-keto-5p-cholan-24-oic acid (KLCA) with CFbOH and H2SO4 to form 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester (1).
  • the reaction mixture is heated for about 3 hours and the pH of the reaction mixture is adjusted with an aqueous basic solution to a pH-value of about 9.5 to about 10.
  • the isolation of 3a-hydroxy-7-keto-5p-cholan-24- oic acid methyl ester (1) further comprises treatment with activated carbon.
  • the isolation of 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester (1) does not further comprise treatment with activated carbon.
  • isolation of 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester (1) without the treatment with activated carbon affords a higher yield.
  • reacting 3a-hydroxy-7-keto- 5p-cholan-24-oic acid (1) with CFbOH and H2SO4 is carried out in methanol.
  • the basic solution is an aqueous NaOH solution.
  • the pH-value is about 9.5 to about 10.
  • the methyl alcohol acts as the methylating reagent as well as the reaction solvent.
  • the solution containing the product is treated with activated carbon for about 30 minutes and filtered to remove the carbon solids. In one embodiment, the solution containing the product is not treated with activated carbon.
  • water at about 5 °C to about 20 °C and seeding material are added. In another embodiment, the water is at about 10 °C to about 15 °C.
  • the product is isolated with a centrifuge and washed with a mixture of methanol and water.
  • the water content of the wet material is quantified by Karl Fischer (KF).
  • the material is dried in a tumble dryer before use in the next step. In one embodiment, the material is not dried before use in the next step. In one embodiment, the wet material is washed with heptane to improve drying.
  • Step 2 is the reaction of 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester (1) with Li[N(CH(CH 3 ) 2 ) 2 ] and Si(CH 3 ) 3 Cl to form 3a,7-ditrimethylsilyloxy-5p-chol-6- en-24-oic acid methyl ester (3).
  • step 2 is carried out in a polar aprotic solvent at a temperature at about -10 °C to about -30 °C.
  • the polar aprotic solvent is tetrahydrofuran.
  • the temperature is about -20 °C to about -25 °C.
  • reacting 3a-hydroxy-7-keto-5p-cholan-24-oic acid methyl ester (1) with Li[N(CH(CH 3 ) 2 )2] and Si(CH 3 ) 3 Cl is stirred for about 2 hours.
  • compound 1 is charged into the reactor under inert conditions.
  • residual water and methanol are removed by repeated azeotropic distillation at about 65 °C and ambient pressure to avoid water hydrolysis of chlorotrimethylsilane which is added later in this step.
  • THF is added to the residue as necessary and the distillation is repeated about 4 times.
  • the distillation is repeated about 3 times, about 2 times, or about 1 time.
  • the remaining solution containing the product has a final water content of ⁇ 0.05% (Karl Fischer Titration).
  • the solution of the product is pre-cooled to about -10 °C to about -30 °C and then chlorotrimethylsilane is added.
  • the solution is pre-cooled to about -20 °C to about -25 °C.
  • a strong base and THF are charged to a separate reactor and cooled to about -10 °C to about -30 °C.
  • the strong base is lithium diisopropylamide.
  • the reactor is inert, e.g. , under a nitrogen or argon atmosphere.
  • the solution of base and THF is cooled to about -20 °C to about - 25 °C.
  • the dry, cooled solution of 3a-hydroxy-7-keto-5 -cholan-24- oic acid methyl ester, THF, and chlorotrimethylsilane is charged into the basic solution at about -10 °C to about -30 °C. In another embodiment, the temperature is about -20 °C to about -25 °C. In one embodiment, the reaction mixture is stirred for about 2 hours. In one embodiment, for the workup, the reaction mixture is added to a pre-cooled acidic solution. In another embodiment, the acidic solution is an aqueous citric acid solution. In one embodiment, after the addition, the aqueous phase is separated and discarded. In one embodiment, the solvent is removed from the organic phase, by vacuum distillation at about 50 °C.
  • the isolated residue is 3 ⁇ ,7 ⁇ -ditrimethylsilyloxy-5 - chol-6-en-24-oic acid methyl ester (3) is used 'as is' in the next step.
  • compound 3 can be purified before Step 3.
  • Step 3 is the reaction of 3a,7-ditrimethylsilyloxy-5p-chol-6-en-24-oic acid methyl ester (3) with CH3CHO to form 3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester (4).
  • step 3 is carried out in a polar aprotic solvent at a temperature at about -50 °C to about -70 °C in the presence of BF3.
  • the polar aprotic solvent is dichloromethane.
  • the BF3 is a 16% wt. solution in acetonitrile.
  • the reaction is carried out in the presence of BF3 diethyl etherate.
  • the temperature is about -60 °C to about -65 °C.
  • compound 3 in a polar aprotic solvent is charged into an inert reactor.
  • the polar aprotic solvent is the residual solvent from the previous step (e.g., THF).
  • THF is added to help distill off residual water and diisopropylamine.
  • the water content in the residue containing compound 3 is limited to ⁇ 0.5% (Karl Fischer titration).
  • the residue containing compound 3 is then dissolved in a polar aprotic solvent and pre-cooled to about -50 °C to about -70 °C.
  • the polar aprotic solvent is dichloromethane.
  • residue containing compound 3 in the polar aprotic solvent is pre-cooled to about -60 °C to about -65 °C.
  • Acetaldehyde (CH3CHO) is added.
  • a polar aprotic solvent and boron trifluoride (BF3) solvated complex are charged into a separate reactor and then cooled to about -50 °C to about -70 °C.
  • the polar aprotic solvent is dichloromethane.
  • the boron trifluoride solvated complex is a boron trifluoride acetonitrile complex.
  • the temperature of the BF3 solution is about -60 °C to about -65 °C.
  • the solution containing compound 3 and acetaldehyde is added to the BF3 solution at about -60 °C to about -65 °C. In another embodiment, the solution containing compound 3 and acetaldehyde is dry. In one embodiment, the reaction mixture is stirred for about two hours at about -60 °C to about -65 °C, heated up to about 23 °C to about 28 °C, stirred for another about 2 hours and cooled to about 2 °C to about 10 °C for the hydrolysis/work-up. In one embodiment, the total time for addition and stirring is about 4 hours. In one embodiment, for the workup, the cooled solution from the reactor is added to a pre-cooled aqueous basic solution.
  • the aqueous basic solution is about 50% wt. sodium hydroxide (NaOH; caustic soda).
  • the phases are separated and the (lower) organic layer is transferred to a separate reactor.
  • the solvent is removed by distillation at not more than (NMT) 50 °C as far as possible.
  • the residue comprises 3a-hydroxy-6-ethylidene-7-keto-5 -cholan-24-oic acid methyl ester (4) and some remaining acetonitrile and dichloromethane. It is understood that step 4 may form E/Z-3a-hydroxy-6-ethylidene-7-keto-5 -cholan-24-oic acid methyl ester (4A).
  • the product of Step 3 is taken on directly to Step 4.
  • Step 4 is the reaction of 3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester (4) with NaOH to form E-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid (5).
  • the residue from step 3 is heated to about 45 °C to about 60 °C to remove residual amounts of solvent. In one embodiment, the temperature is about 49 °C to about 55 °C.
  • ester hydrolysis reaction reacting 3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester (4) with NaOH is carried out at about 20 °C to about 25 °C in methanol, water, and a NaOH solution.
  • reacting 3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid methyl ester (4) is charged into a reactor.
  • the reactor is inert, e.g. , under a nitrogen or argon atmosphere.
  • NMT 50 °C residual amounts of solvent are distilled off under vacuum.
  • the residue is heated up to about 45 °C to about 60 °C.
  • the residue is heated up to about 49 °C to about 55 °C.
  • the residue from Step 3 (compound 4) is dissolved in methanol and water and an aqueous basic solution.
  • the aqueous basic solution is about 50% wt. sodium hydroxide
  • the ester hydrolysis reaction of Step 4 is carried out at about 20 °C to about 60 °C and stirred until the hydrolysis reaction is complete. In one embodiment, the ester hydrolysis is carried out at about 20 °C to about 25 °C.
  • the pH of the reaction mixture is checked to verify it is > 12. If the pH is ⁇ 12, then additional NaOH is added.
  • the reaction mixture is diluted with water and the temperature is adjusted to about 20 °C to about 35 °C. In another aspect, the reaction mixture is diluted with water and the temperature is adjusted to about 25 °C to about 35 °C.
  • the phases are separated and the lower product-rich aqueous layer is transferred into a separate reactor and the organic layer is discarded.
  • the pH of the product-rich aqueous phase is adjusted with aqueous acid in the presence of ethyl acetate.
  • the acid is an aqueous citric acid solution.
  • the phases are separated and the upper product-rich organic layer is retained and the lower aqueous layer is discarded.
  • ethyl acetate is distilled off from the organic layer and replaced with ethyl acetate.
  • the distillation is repeated until the water content of the distillate is NMT 1% or until a constant boiling point is reached.
  • the resulting product suspension is cooled to about 10 °C to about 30 °C and isolated and washed with ethyl acetate. In one embodiment, the resulting product is isolated by filtration (e.g. filter nutche, centrifuge, etc). In another embodiment, the resulting product suspension containing compound 5 is cooled to about 20 °C to about 25 °C. In one embodiment, the drying of the resulting product is done under vacuum (e.g, cone dryer, paddle dryer, tray dryer, etc.) at about 60 °C.
  • vacuum e.g, cone dryer, paddle dryer, tray dryer, etc.
  • crude E-3a-hydroxy-6-ethylidene-7-keto-5 -cholan-24-oic acid (5) is crystallized using ethanol.
  • ethanol and crude compound 5 are charged into reactor.
  • the reactor is inert.
  • to dissolve the crude compound 5 the mixture is heated to reflux.
  • mixture is cooled in a controlled cooling ramp to about 15 °C to about 20 °C.
  • the crystalline compound 5 is isolated using a centrifuge and then washed with ethyl acetate.
  • drying of crystalline compound 5 is done under vacuum (e.g, cone dryer, paddle dryer, tray dryer, etc.) and at about 60 °C.
  • purified compound 5 contains both E and Z isomers of 3a-hydroxy-6- ethylidene-7-keto-5 -cholan-24-oic acid.
  • the E to Z ratio is about 99: 1, about 98:2, about 95: 5, about 90: 10, about 85: 15, about 80:20, about 75:25, about 70:30, about 65:35, about 60:40, about 55:45, or about 50:50.
  • Step 4 can also be carried out starting with a compound that is a mixture of E/Z isomer.
  • Step 4 is the reaction of E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid methyl ester (4A) with NaOH to form E/Z-3a-hydroxy-6-ethylidene- 7-keto-5p-cholan-24-oic acid (5 A).
  • the residue from step 3 is heated about 45 °C to about 60 °C to remove residual amounts of solvent.
  • the temperature is about 49 °C to about 55 °C.
  • the ester hydrolysis reaction involving reacting E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan- 24-oic acid methyl ester (4A) with NaOH is carried out at about 20 °C to about 25 °C in methanol, water, and a NaOH solution.
  • the NaOH solution is a 50% wt. aqueous solution.
  • reacting E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24- oic acid methyl ester (4A) is charged into a reactor.
  • the reactor is inert, e.g. , under a nitrogen or argon atmosphere.
  • NMT 50 °C residual amounts of solvent are distilled off under vacuum.
  • the residue is heated up to about 45 °C to about 60 °C.
  • the temperature is about 49 °C to about 55 °C.
  • the residue from step 3 (compound 4A) is dissolved in methanol and water and an aqueous basic solution.
  • the aqueous basic solution is about 50% wt. sodium hydroxide (NaOH; caustic soda).
  • the ester hydrolysis reaction of step 4 is carried out at about 20 °C to about 60 °C and stirred until the hydrolysis reaction is complete. In one embodiment, the ester hydrolysis is carried out at about 20 °C to about 25 °C.
  • the pH of the reaction mixture is checked to verify it is > 12. If the pH is ⁇ 12, then additional NaOH is added.
  • the reaction mixture is diluted with water and the temperature is adjusted to about 25 °C to about 35 °C. In one embodiment, for the workup, the phases are separated and the lower product-rich aqueous layer is transferred into a separate reactor and the organic layer is discarded.
  • the pH of the product-rich aqueous is adjusted with aqueous acid in the presence of ethyl acetate.
  • the acid is an aqueous citric acid solution.
  • the phases are separated and the upper product-rich organic layer is retained and the lower aqueous layer is discarded.
  • ethyl acetate is distilled off from the organic layer and replaced with ethyl acetate.
  • the distillation is repeated until the water content of the distillate is NMT 1% or until a constant boiling point is reached.
  • the resulting product suspension is cooled to about 10 °C to about 30 °C and isolated and washed with ethyl acetate.
  • the resulting product is isolated by filtration (e.g. filter nutche, centrifuge, etc.).
  • the resulting product suspension containing compound 5A is cooled to about 20 °C to about 25 °C.
  • drying of the resulting product is done under vacuum (e.g, cone dryer, paddle dryer, tray dryer, etc.) at about 60 °C.
  • Compound 5A can be carried on without purification to Step 5.
  • crude E/Z-3a-hydroxy-6-ethylidene-7-keto-5 -cholan-24-oic acid (5 A) is crystallized using ethanol.
  • ethanol and crude compound 5A are charged into reactor.
  • the reactor is inert.
  • to dissolve the crude compound 5A the mixture is heated to reflux.
  • mixture is cooled in a controlled cooling ramp to about 15 °C to about 20 °C.
  • the crystalline compound 5A is isolated using a centrifuge and then washed with ethyl acetate.
  • drying of crystalline compound 5A is done under vacuum (e.g, cone dryer, paddle dryer, tray dryer, etc.) and at about 60 °C.
  • the isolated crystalline product of step 4 is compound 5.
  • Compound 5 can be prepared according to an alternative method.
  • compound 4 is charged into the inert reactor.
  • solvent e.g., acetonitrile, dichloromethane
  • solvent e.g., acetonitrile, dichloromethane
  • the residue is dissolved in methanol and cooled.
  • Water and caustic soda 50 % weight NaOH
  • the reaction mixture is stirred for about four hours at about 20 °C to about 25 °C.
  • the solution is diluted with water and toluene is added. After stirring, the phases are separated and the lower product-rich aqueous layer is transferred into the inert reactor. The organic layer is discarded.
  • the pH of the product rich aqueous layer is adjusted with aqueous citric acid in the presence of ethyl acetate.
  • the phases are separated and the lower aqueous layer is discarded.
  • the organic layer is transferred into the inert reactor. From the organic layer ethyl acetate is distilled off and replaced with ethyl acetate. In one embodiment, this operation is repeated until the water content of the distillate is not more than about 1 % or until a constant boiling point is reached.
  • the resulting product suspension is cooled to about 20 °C to about 25 °C, and compound 5 is isolated and washed with ethyl acetate in the inert centrifuge. Drying is done in the tumble dryer under vacuum and approximately 60 °C.
  • Step 4 can also be carried out starting with a compound that is a mixture of E/Z isomer.
  • compound 4A is charged into the inert reactor.
  • solvent e.g., acetonitrile, dichloromethane
  • the residue is dissolved in methanol and cooled.
  • Water and caustic soda (50%wt, NaOH) are added.
  • the reaction mixture is stirred for approximately four hours at about 20 °C to about 25 °C.
  • the solution is diluted with water and toluene is added. After stirring, the phases are separated and the lower product-rich aqueous layer is transferred into the inert reactor. The organic layer is discarded.
  • the pH of the product rich aqueous layer is adjusted with aqueous citric acid in the presence of ethyl acetate.
  • the phases are separated and the lower aqueous layer is discarded.
  • the product-rich organic layer is transferred into the inert reactor. From the organic layer ethyl acetate is distilled off and replaced with ethyl acetate. In one embodiment, this operation is repeated until the water content of the distillate is not more than about 1 % or until a constant boiling point is reached.
  • the resulting product suspension is cooled to 20 °C to 25 °C, and compound 5A is isolated and washed with ethyl acetate in the inert centrifuge. Drying is done in the tumble dryer under vacuum and approximately 60 °C.
  • Step 5 is the reaction of E-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid
  • Step 5 can be carried out in one phase (hydrogenation and isomerization together) or in two discrete phases (hydrogenation followed by isomerization). In one embodiment, Step 5 is carried out at a temperature at about 90 °C to about 110 °C and at a pressure at about 0.5 to about 5 bars. In one embodiment, during workup, the organic phase of the reaction mixture is treated with activated carbon.
  • the pressure is about 0.5 to about 5.5 bars. In another embodiment, the pressure is about 5 bars. In another embodiment, the hydrogenation is performed at a pressure of about 0.5 bar to about 5.5 bar, e.g 0.6 bar, 0.7 bar, 0.8 bar, 0.9 bar, 1.0 bar, 1.2 bar, 1.4 bar, 1.6 bar, 1.8 bar, 2.0 bar, 2.2 bar, 2.4 bar, 2.6 bar, 2.8 bar, 3.0 bar, 3.2 bar, 3.4 bar, 3.6 bar, 3.8 bar, 4.0 bar, 4.2 bar, 4.4 bar, 5.0 bar, 5.2 bar, as well as any increment in between. In one embodiment, the hydrogenation reaction mixture is allowed to stir for about 1 hour.
  • E-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid (5) in the presence of PdVC and hydrogen gas is heated to about 100 °C and stirred for about 2 hours to about 5 hours. In one embodiment, E-3a-hydroxy-6-ethylidene-7- keto-5p-cholan-24-oic acid (5) in the presence of Pd/C and hydrogen gas is heated to about 100 °C and stirred for about 3 hours.
  • E-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid (5) in the presence of Pd/C and hydrogen gas is carried out in the presence of a basic solution.
  • the basic solution is water containing a 50% wt. sodium hydroxide (NaOH; caustic soda) solution.
  • NaOH sodium hydroxide
  • the reaction mixture is heated up to about 100 °C (to carry out the isomerisation of the C-6 position from beta configuration to alpha configuration) and then cooled to about 40 °C to about 50 °C.
  • the Pd/C is filtered off.
  • n- butyl acetate and an acid are added to the filtrate.
  • the acid is hydrochloric acid (HC1).
  • HC1 hydrochloric acid
  • the aqueous phase is separated and discarded after checking the pH-value to make sure that it was acidic.
  • the product-rich organic phase is treated with activated carbon.
  • the activated carbon is filtered off and the resulting product- rich filtrate is concentrated by distillation and the resulting product suspension is cooled to about 10 °C to about 30 °C.
  • the product suspension is cooled to about 15 °C to about 20 °C.
  • the suspension containing compound 6 is isolated and washed with n-butyl acetate.
  • Compound 6 is filtered using a pressure filter. In one embodiment, drying is done in the pressure filter under vacuum at about 80 °C.
  • E-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24- oic acid (5), water, NaOH solution (e.g. 50% wt.), and Pd/C are mixed at about 5 bar of 3 ⁇ 4 gas and at a temperature at about 100 °C to about 105 °C until 3 ⁇ 4 uptake has ceased.
  • the reaction mixture is cooled to about 40 °C to about 50 °C and Pd/C is filtered off.
  • n-butyl acetate and HC1 are added to the solution containing compound 6.
  • the aqueous phase is separated and discarded.
  • the organic phase containing compound 6 is treated with activated carbon.
  • the carbon is filtered off and the filtrate is moved to another reactor where it is concentrated by distillation, and then the suspension is cooled to about 5 °C to about 20 °C.
  • compound 6 is isolated via filtration and the filtrate is dried on the pressure filter under vacuum at about 80 °C.
  • Step 5 can also be carried out starting with a compound that is a mixture of E/Z isomer.
  • Step 5 is the reaction of E/Z-3a-hydroxy-6-ethylidene-7-keto-5p- cholan-24-oic acid (5 A) with Pd/C and hydrogen gas and heat to form 3a-hydroxy-6a- ethyl-7-keto-5p-cholan-24-oic acid (6).
  • Step 5 can be carried out in one phase
  • step 5 is carried out at a temperature at about 90 °C to about 110 °C and at a pressure at about 4 to about 5 bars.
  • the organic phase of the reaction mixture is treated with activated carbon.
  • the pressure is about 4.5 to about 5.5 bars. In another embodiment, the pressure is about 5 bars.
  • the hydrogenation reaction mixture is allowed to stir for about 1 hour.
  • reacting E/Z-3a-hydroxy-6- ethylidene-7-keto-5p-cholan-24-oic acid (5 A) with Pd/C and hydrogen gas is heated to about 100 °C and stirred for about 2 hour to about 5 hours. In one embodiment, reacting E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24-oic acid (5A) with Pd/C and hydrogen gas is heated to about 100 °C and stirred for about 3 hours.
  • reacting E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24- oic acid (5 A) with Pd/C and hydrogen gas is carried out in the presence of a basic solution.
  • the basic solution is water and a 50% wt. sodium hydroxide (NaOH; caustic soda) solution.
  • NaOH sodium hydroxide
  • the reaction mixture is heated up to about 100 °C (to carry out the isomerisation of the C-6 position from beta configuration to alpha configuration) and then cooled to about 40 °C to about 50 °C.
  • the Pd/C is filtered off.
  • n-butyl acetate and an acid are added.
  • the acid is hydrochloric acid (HC1).
  • HC1 hydrochloric acid
  • the aqueous phase is separated and discarded after checking the pH-value to make sure that it was acidic.
  • the product-rich organic phase is treated with activated carbon.
  • the activated carbon is filtered off and the resulting product-rich filtrate is concentrated by distillation and the resulting product suspension is cooled to about 10 °C to about 30 °C.
  • the product suspension is cooled to about 15 °C to about 20 °C.
  • the suspension containing compound 6 is isolated and washed with n- butyl acetate.
  • Compound 6 is filtered using a pressure filter. In one embodiment, drying is done in the pressure filter under vacuum at about 80 °C.
  • E/Z-3a-hydroxy-6-ethylidene-7-keto-5p-cholan-24- oic acid (5 A), water, NaOH solution (e.g. 50% wt.), and Pd/C are mixed at about 5 bar of H2 gas and at a temperature at about 100 °C to about 105 °C until H2 uptake has ceased.
  • the reaction mixture is cooled to about 40 °C to about 50 °C and Pd/C is filtered off.
  • n-butyl acetate and HC1 are added to the solution containing compound 6.
  • the aqueous phase is separated and discarded.
  • the product rich organic phase is treated with activated carbon.
  • the carbon is filtered off and the filtrate is moved to another reactor where it is concentrated by distillation, and then the product suspension is cooled to about 5 °C to about 20 °C.
  • compound 6 is isolated via filtration and the filtrate is dried on the pressure filter under vacuum at about 80 °C.
  • the hydrogenation/isomerization reactions described above to prepare compound 6 are carried out in two phases (starting from compound 5 or compound 5A). First, the hydrogenation is carried out at about 4 to 5 bars and at about 20 °C to about 40 °C and then second, the reaction mixture is heated to about 95 °C to about 105 °C. Heating the reaction mixture isomerizes the ethyl group at the 6-position from the initially formed 6-beta configuration to the desired 6-alpha configuration. The reaction mixture is heated until the isomerization is complete.
  • Step 6 is the reaction of 3a-hydroxy-6a-ethyl-7-keto-5p-cholan-24-oic acid (6) with NaBFU to form crystalline obeticholic acid.
  • Step 6 is carried out at a temperature at about 85 °C to about 110 °C in a basic aqueous solution. In one embodiment, the temperature is about 90 °C to about 95 °C.
  • the basic aqueous solution is an aqueous NaOH solution. In one embodiment, the basic aqueous solution is a mixture of 50% wt. NaOH solution and water.
  • the reaction mixture of compound 6 and NaBH4 was stirred for about 3 hours to about 5 hours. In another embodiment, the reaction mixture was stirred for about 4 hours.
  • the mixture is cooled to about 80 °C and transferred to a cooled reactor.
  • n-butyl acetate and an acid are added.
  • the temperature is about 40 °C to about 45 °C.
  • the acid is citric acid.
  • the aqueous phase is separated and discarded after checking the pH-value to make sure that it was acidic.
  • the product-rich organic phase is concentrated by distillation.
  • n-butyl acetate is added to the residue and distilled off again.
  • n-butyl acetate is added again to the residue and then is slowly cooled down.
  • the residue is seeded at about 50 °C.
  • the mixture is heated to 52 °C and then slowly cooled down to about 15 °C to about 20 °C.
  • the residue is cooled to about 15 °C to about 20 °C.
  • the resulting obeticholic acid is washed with n-butyl acetate.
  • the obeticholic acid is isolated and washed with n-butyl acetate (e.g, in a pressure filter).
  • the pressure filter is inert.
  • the crystalline product is dried under vacuum at about 60 °C and is identified as Form A characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 41 including characteristic peaks at about 5.0 and 5.3 degrees 2-Theta.
  • the resulting crystalline obeticholic acid is isolated from organic solvent (e.g., heptane) and is characterized by an X-ray diffraction partem including characteristic peaks at about 5.0 and 5.3 degrees 2-Theta.
  • the crystalline obeticholic acid Form A is characterized by an X-ray diffraction pattern substantially similar to that set forth in Figure 41. See example 3 for full details regarding the identification and characterization of crystalline obeticholic acid Form A.
  • Step 7 is the conversion of crystalline obeticholic acid Form A to obeticholic acid Form 1.
  • Step 7 comprises the step of dissolving crystalline obeticholic acid Form A in aqueous NaOH solution and adding HC1.
  • crystalline obeticholic acid is dissolved in water and caustic soda solution (50% wt.) at about 20 °C to about 50 °C.
  • the temperature is about 30 °C to about 40 °C.
  • the crystalline obeticholic acid is Form A.
  • the resulting solution of crystalline obeticholic acid Form A is added to diluted acid at about 20 °C to about 50 °C.
  • the temperature is about 30 °C to about 40 °C.
  • the acid is hydrochloric acid (e.g., 37%).
  • the 37% hydrochloric acid solution is diluted with water to less than about 1% by volume.
  • the 37% hydrochloric acid solution is diluted with water to about 0.7% by volume.
  • the suspension of product in the diluted acid is stirred for about 30 minutes at about 20 °C to about 50 °C.
  • the temperature is about 30 °C to about 40 °C.
  • obeticholic acid Form 1 is isolated and washed with water (e.g., in the pressure filter) at NMT about 20 °C.
  • obeticholic acid Form 1 is isolated and washed with water (e.g., in the pressure filter) at NMT about 20 °C.
  • the pressure filter is inert. The product is dried on the pressure filter under vacuum at a temperature of NMT about 50 °C.
  • Step 6 of the synthesis produces a novel crystalline form of obeticholic acid.
  • the production of this crystalline form leads to substantially pure obeticholic acid Form 1.
  • the '977 process to prepare obeticholic acid is an 8-step synthetic process which includes one purification step (step 7) followed by 2 additional purification steps.
  • step 7 There are a significant number of differences between the '977 process and the process of the present application. Table A below describes at least some of the differences between the two processes:
  • Step 3 Safety concerns of (Process of application Boron trifluoride diethyl Boron trifluoride handling etherate (T4 step 3 same as '977 etherate acetonitrile complex emission controls not Step 4) needed)
  • Step 7 Ammonia solution NaOH solution Scale-up (Process of application
  • step 7 same as '977 Phosphoric acid Hydrochloric acid
  • the differences in the process of the present application as compared to the '977 process result in significant improvements to the process, including improvements related to scale-up optimization, safety, as well as purity and improvements in the overall process.
  • the purity of obeticholic acid produced by the processes of the present application is substantially pure.
  • obeticholic acid produced by the processes of the present application is substantially more pure than obeticholic acid produced by processes in the prior art, including the '390 process and the '977 process.
  • Table B The percentages of impurities were determined using HPLC methods. Table B: Comparison of Impurities of Obeticholic Acid Generated from Process of the Application and '977 Process
  • Impurity 1 is 6-ethylursodeoxycholic acid.
  • Impurity 2 is 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid.
  • Impurity 3 is 6 -ethylchenodeoxycholic acid.
  • Impurity 4 is 3a,7a-dihydroxy-6-ethyliden-5 -cholan-24-oic acid.
  • Impurity 5 is chenodeoxycholic acid.
  • Impurity 6 is 3a(3a a-chhydroxy-6a-ethyl-5 -cholan-24-oyloxy)-7a-hydroxy-6a-ethyl-5 -cholan-24-oic acid (6ECDCA dimer).
  • NMT refers to "not more than”.
  • Obeticholic acid is currently being developed as an active pharmaceutical ingredient as a non-crystalline solid.
  • an initial crystallization and polymorphism study was carried out in order to determine if crystalline forms were accessible and if so, if they were suitable for development. After a preliminary solubility screen designed to give a better
  • Crystalline obeticholic acid could readily be isolated on large scale using the process of the application. This crystalline obeticholic acid was determined to be consistent with Form A from the initial crystallization and polymorph study. The formation, ease of isolation, and highly pure crystalline obeticholic acid produced as a synthetic intermediate in step 7 in the process of the present application is indeed critical to the preparation of substantially pure obeticholic acid.
  • the present invention relates to a crystalline obeticholic acid Form A characterized by an X-ray diffraction partem including characteristic peaks at about 5.0 and 5.3 degrees 2-Theta.
  • the X-ray diffraction pattern includes characteristic peaks at about 5.0, 5.3 and 7.7 degrees 2-Theta.
  • the X-ray diffraction pattern includes characteristic peaks at about 5.0, 5.3, 7.7, and 10.0 degrees 2-Theta.
  • the X-ray diffraction pattern includes characteristic peaks at about 5.0, 5.3, 7.7, 10.0, and 11.0 degrees 2-Theta.
  • the X-ray diffraction pattem includes characteristic peaks at about 5.0, 5.3, 7.7, 10.0, 11.0, and 12.4 degrees 2-Theta. In one embodiment, the X-ray diffraction pattem includes characteristic peaks at about 5.0, 5.3, 7.7, 10.0, 11.0, 12.4, and 14.9 degrees 2-Theta. In one embodiment, the present invention relates to a crystalline obeticholic acid Form A characterized by an X-ray diffraction pattern substantially similar to that set forth in Figure 41. In one embodiment, the X-ray diffraction pattern is collected on a diffractometer using Cu K radiation (40 kV, 40 mA).
  • the present invention relates to a crystalline obeticholic acid, wherein said crystalline obeticholic acid is Form A and has a purity greater than about 90%.
  • the purity of said crystalline obeticholic acid Form A is determined by HPLC.
  • the present invention relates to a crystalline obeticholic acid Form A, or a pharmaceutically acceptable salt, solvate or amino acid conjugate thereof.
  • the solvate is a n-butyl solvate.
  • the purity is greater than about 92%.
  • the purity is greater than about 94%.
  • the purity is greater than about 96%.
  • the purity is greater than about 98%.
  • the purity is greater than about 99%.
  • the present invention relates to a crystalline obeticholic acid, wherein said crystalline obeticholic acid is Form A and has a potency greater than about 90%.
  • the purity of said crystalline obeticholic acid Form A is determined by HPLC and/or other analytical procedures known in the art.
  • the present invention relates to a crystalline obeticholic acid Form A, or a pharmaceutically acceptable salt, solvate or amino acid conjugate thereof.
  • the solvate is a hydrate.
  • the potency is greater than about 92%.
  • the potency is greater than about 94%.
  • the potency is greater than about 96%.
  • the potency is greater than about 98%.
  • the potency is greater than about 99%.
  • the present invention relates to a crystalline obeticholic acid Form A that contains a total of less than about 4% of one or more impurities selected from 6-ethylursodeoxycholic acid, 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid, 6p-ethylchenodeoxycholic acid, 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid, chenodeoxycholic acid, and 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a- hydroxy-6a-ethyl-5p-cholan-24-oic acid.
  • the total impurities is less than about 3.8%. In one embodiment, the total impurities is less than about 3.6%.
  • the present invention relates to a crystalline obeticholic acid Form C characterized by an X-ray diffraction pattern including characteristic peaks at about 4.2, 6.4, 9.5, 12.5, and 16.7 degrees 2-Theta.
  • the X-ray diffraction pattern includes characteristic peaks at about 4.2, 6.4, 9.5, 12.5, 12.6, 15.5, 15.8, 16.0, 16.7 and 19.0 degrees 2-Theta.
  • the X-ray diffraction partem includes characteristic peaks at about 4.2, 6.4, 8.3, 9.5, 11.1, 12.2, 12.5, 12.6, 15.5, 15.8, 16.0, 16.3, 16.7, 18.6 and 19.0 degrees 2-Theta.
  • the X-ray diffraction pattern includes characteristic peaks at about 4.2, 6.4, 8.3, 9.5, 11.1, 12.2, 12.5, 12.6, 15.5, 15.8, 16.0, 16.3, 16.7, 17.0, 17.8, 18.6, 18.8, 19.0, 20.5 and 20.9 degrees 2-Theta.
  • the present invention relates to a crystalline obeticholic acid Form C characterized by an X-ray diffraction pattern substantially similar to that set forth in Figure 5.
  • the X-ray diffraction pattern is collected on a diffractometer using Cu K radiation (40 kV, 40 mA).
  • the X-ray diffraction pattern includes characteristic peaks at about 12.0 to about 12.8 and about 15.4 to about 21.0.
  • the present invention relates to a crystalline obeticholic acid Form C characterized by a Differential Scanning Calorimetry (DSC) thermogram having an endotherm value at about 98 ⁇ 2 °C, as measured by a Mettler DSC 823e instrument.
  • the Differential Scanning Calorimetry (DSC) thermogram has an endotherm value at about 98 ⁇ 2 °C, as measured by a Mettler DSC 823e instrument.
  • the present invention relates to a crystalline obeticholic acid, wherein said crystalline obeticholic acid is Form C and has a purity greater than about 90%.
  • the purity of said crystalline obeticholic acid Form C is determined by HPLC.
  • the present invention relates to a crystalline obeticholic acid Form C, or a pharmaceutically acceptable salt, solvate or amino acid conjugate thereof.
  • the solvate is a hydrate.
  • the purity is greater than about 92%.
  • the purity is greater than about 94%.
  • the purity is greater than about 96%.
  • the purity is greater than about 98%.
  • the purity is greater than about 99%.
  • the present invention relates to a crystalline obeticholic acid, wherein said crystalline obeticholic acid is Form C and has a potency greater than about 90%.
  • the purity of said crystalline obeticholic acid Form C is determined by HPLC and/or other analytical procedures known in the art.
  • the present invention relates to a crystalline obeticholic acid Form C, or a pharmaceutically acceptable salt, solvate or amino acid conjugate thereof.
  • the solvate is a hydrate.
  • the potency is greater than about 92%.
  • the potency is greater than about 94%.
  • the potency is greater than about 96%.
  • the potency is greater than about 98%.
  • the potency is greater than about 99%.
  • the present invention relates to a crystalline obeticholic acid Form C that contains a total of less than about 4% of one or more impurities selected from 6-ethylursodeoxycholic acid, 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid, 6p-ethylchenodeoxycholic acid, 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid, chenodeoxycholic acid, and 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a- hydroxy-6a-ethyl-5p-cholan-24-oic acid.
  • the total impurities is less than about 3.8%. In one embodiment, the total impurities is less than about 3.6%.
  • Example 3 of the application provides full characterization of these crystalline forms of obeticholic acid.
  • the single crystal X-ray structure of obeticholic acid was obtained and the absolute stereochemistry assigned.
  • the single crystal X-ray structure of crystalline obeticholic acid Form G was determined from a crystal obtained from the recrystallization of obeticholic acid from an acetonitrile solution after cooling to 5°C at 0.1°C/min followed by maturation at RT/50°C 8 h cycles for 1 week.
  • the structure is orthorhombic, space group P2i2i2i, and contains one molecule of obeticholic acid in the asymmetric unit.
  • Final Rl [ ⁇ >2 ⁇ ( ⁇ )] 3.22 %.
  • a bioavailability study of obeticholic acid Form 1 (non-crystalline) vs. crystalline obeticholic acid Form F was carried out (Example 7).
  • the results of the study show that that physical state of a solid obeticholic acid can play a role in the bioavailability of the molecule when administered orally to a subject.
  • the plasma kinetics after oral administration and the efficiency of the intestinal absorption and the pharmacokinetics of solid obeticholic acid Form 1 (non-crystalline) and crystalline Form F were evaluated according to methods known in the art.
  • Example 8 of the present invention shows the profiles of obeticholic acid plasma concentration vs time, the tmax, Cmax and AUC after administration of Form 1 or Form F of obeticholic acid (see Figures 37-38).
  • Crystalline Form F has a higher bioavailability than obeticholic acid Form 1 (non-crystalline).
  • the plasma profiles show that the Form F is absorbed more efficiently (higher AUC) and even the kinetics is more regular, reflecting an optimal distribution of the drug in the intestinal content.
  • the present application provides substantially pure obeticholic acid and pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof:
  • obeticholic acid names for the pharmaceutically active ingredient obeticholic acid are INT-747, 3a,7a-dihydroxy-6a-ethyl-5 -cholan-24-oic acid, 6a-ethyl-chenodeoxycholic acid, 6- ethyl-CDCA, 6ECDCA, and cholan-24-oic acid,6-ethyl-3,7-dihydroxy-,(3a,5 ,6a,7a)-
  • compositions comprising obeticholic acid
  • obeticholic acid Form 1 and processes for the synthesis of highly pure obeticholic acid Form 1 which are safe and which produce obeticholic acid on a large scale.
  • obeticholic acid Form 1 is produced on a commercial scale process.
  • the term "commercial scale process" refers to a process which is run as a single batch of at least about 100 grams.
  • the process of the present application produces obeticholic acid Form 1 in high yield (>80%) and with limited impurities.
  • purity refers to the amount of obeticholic acid based on HPLC. Purity is based on the "organic" purity of the compound. Purity does not include a measure of any amount of water, solvent, metal, inorganic salt, etc. In one aspect, the purity of obeticholic acid is compared to the purity of the reference standard by comparing the area under the peak. In another aspect, the known standard for purity is an obeticholic acid reference standard. In one aspect, obeticholic acid has a purity of greater than about 96%. In one aspect, obeticholic acid has a purity of greater than about 98%.
  • the purity of obeticholic acid Form 1 is 96.0%, 96.1%, 96.2%, 96.3%, 96.4%, 96.5%, 96.6%, 96.7%, 96.8%, 96.9%, 97.0%, 97.1%, 97.2%, 97.3%, 97.4%, 97.5%, 97.6%, 97.7%, 97.8%, 97.9 %, 98.0%, 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the purity of obeticholic acid Form 1 is 98.0%, 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the purity of obeticholic acid is 98.0%, 98.5%, 99.0%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the purity of obeticholic acid is 98.5%, 99.0%, or 99.5%.
  • the obeticholic acid is obeticholic acid Form 1. In one embodiment, the present invention relates to obeticholic acid having a purity greater than about 98%. In one embodiment, the purity is determined by HPLC. In another embodiment, the present invention relates to obeticholic acid, or a
  • the purity is greater than about 98.5%. In one embodiment, the purity is greater than about 99.0%. In one embodiment, the purity is greater than about 99.5%. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • potency is a measure of the amount of obeticholic acid based on that of a known standard (e.g., acceptance criteria of about 95% to about 102%). Potency takes into account all possible impurities including water, solvents, organic, and inorganic impurities.
  • the known standard is obeticholic acid.
  • obeticholic acid has a potency of greater than about 96%.
  • obeticholic acid has a potency of greater than about 98%.
  • the known standard is obeticholic acid.
  • potency is 100% minus the amounts of water, sulphated ash, residual solvents, and other impurity contents such as 6- ethylursodeoxycholic acid, 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid, 6 ⁇ - ethylchenodeoxycholic acid, 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid, chenodeoxycholic acid, and 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a- hydroxy-6a-ethyl-5p-cholan-24-oic acid.
  • potency accounts for impurities due to water, solvent, metals, inorganic salts, and other inorganic or organic impurities.
  • the potency of obeticholic acid Form 1 is 96.0%, 96.1%, 96.2%, 96.3%, 96.4%, 96.5%, 96.6%, 96.7%, 96.8%, 96.9%, 97.0%, 97.1%, 97.2%, 97.3%, 97.4%, 97.5%, 97.6%, 97.7%, 97.8%, 97.9 %, 98.0%, 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the potency of obeticholic acid Form 1 is 98.0%, 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the potency of obeticholic acid is 98.0%, 98.5%, 99.0%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%.
  • the potency of obeticholic acid is 98.5%, 99.0%, or 99.5%.
  • the obeticholic acid is obeticholic acid Form 1.
  • the present invention relates to obeticholic acid containing a total of less than about 2% of one or more impurities selected from 6- ethylursodeoxycholic acid, 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid, 6 ⁇ - ethylchenodeoxycholic acid, 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid, chenodeoxycholic acid, and 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a- hydroxy-6a-ethyl-5p-cholan-24-oic acid.
  • the total of impurities is less than about 1.5%.
  • the total of impurities is less than about 1.4%.
  • the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains less than about 10% of water, less than about 9% of water, less than 8% of water, less than 7% of water, less than 6% of water, less than 5% of water, less than 4% of water, less than 3% of water, less than 2% of water, or less than 1% of water. In one embodiment, obeticholic acid contains less than about 1.2% of water. In one embodiment, obeticholic acid contains less than about 1.0% of water. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains not more than (NMT) 0.15 % of 6-ethylursodeoxycholic acid and 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid. In another embodiment, obeticholic acid contains a total of less than about 0.07% of 6- ethylursodeoxycholic acid and 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid. In one embodiment, obeticholic acid contains a total of less than about 0.06% of 6- ethylursodeoxycholic acid and 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid.
  • obeticholic acid contains a total of less than about 0.05% of 6- ethylursodeoxycholic acid and 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24-oic acid. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains not more than (NMT) 0.15 % of 3a- hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid. In one embodiment, obeticholic acid contains less than about 0.07% of 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid. In one embodiment, obeticholic acid contains less than about 0.06% of 3a-hydroxy-6a- ethyl-7-cheto-5p-cholan-24-oic acid. In one embodiment, obeticholic acid contains less than about 0.05% of 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan-24-oic acid. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains not more than (NMT) 0.15% of 6 ⁇ - ethylchenodeoxycholic acid. In one embodiment, obeticholic acid contains less than about 0.07% of 6p-ethylchenodeoxycholic acid. In one embodiment, obeticholic acid contains less than about 0.06% of 6p-ethylchenodeoxycholic acid. In one embodiment, obeticholic acid contains less than about 0.05% of 6p-ethylchenodeoxycholic acid. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains no more than (NMT) 3% of chenodeoxycholic acid (CDCA). In one embodiment, obeticholic acid contains less than about 1% of CDCA. In one embodiment, obeticholic acid contains less than about 0.5% of CDCA. In one embodiment, obeticholic acid contains less than about 0.3% of CDCA. In one embodiment, obeticholic acid contains less than about 0.2% of CDCA. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • obeticholic acid contains no more than (NMT) 4% of CDCA and 6-ethylursodeoxycholic acid.
  • obeticholic acid contains no more than (NMT) 1.5 % of 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a-hydroxy-6a-ethyl-5p-cholan- 24-oic acid. In one embodiment, obeticholic acid contains less than about 1% of 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a-hydroxy-6a-ethyl-5p-cholan- 24-oic acid. In one embodiment, obeticholic acid contains less than about 0.07% of
  • obeticholic acid contains less than about 0.06% of 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a-hydroxy-6a-ethyl-5p-cholan- 24-oic acid.
  • obeticholic acid contains less than about 0.05% of 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24-oyloxy)-7a-hydroxy-6a-ethyl-5p-cholan- 24-oic acid. In one embodiment, the obeticholic acid is obeticholic acid Form 1.
  • Obeticholic acid is for oral administration.
  • the formulation is oral administration for the prevention and treatment of FXR mediated diseases and conditions.
  • the formulation comprises of obeticholic acid Form 1.
  • the formulation comprises of substantially pure obeticholic acid.
  • Formulations suitable for oral administration may be provided as discrete units, such as tablets, capsules, cachets (wafer capsule used by pharmacists for presenting a drug), lozenges, each containing a predetermined amount of obeticholic acid; as powders or granules; as solutions or suspensions in aqueous or non-aqueous liquids; or as oil-in- water or water-in-oil emulsions.
  • Formulations of the invention may be prepared by any suitable method, typically by uniformly and intimately admixing obeticholic acid with liquids or finely divided solid carriers or both, in the required proportions and then, if necessary, shaping the resulting mixture into the desired shape.
  • a tablet may be prepared by compressing an intimate mixture comprising a powder or granules of obeticholic acid and one or more optional ingredients, such as a binder, lubricant, inert diluent, or surface active dispersing agent, or by moulding an intimate mixture of powdered active ingredient and inert liquid diluent.
  • optional ingredients such as a binder, lubricant, inert diluent, or surface active dispersing agent, or by moulding an intimate mixture of powdered active ingredient and inert liquid diluent.
  • one or more tablets may be administered to get to a target dose level based on the subject's weight, e.g., a human between about 30 kg to about 70 kg.
  • the subject is a child and the formulation is used to treat biliary atresia.
  • Biliary atresia also known as "extrahepatic ductopenia” and “progressive obliterative cholangiopathy” is a congenital or acquired disease of the liver and one of the principal forms of chronic rejection of a transplanted liver allograft.
  • the congenital form the common bile duct between the liver and the small intestine is blocked or absent.
  • the acquired type most often occurs in the setting of autoimmune disease, and is one of the principal forms of chronic rejection of a transplanted liver allograft.
  • the human child has had a Kasai procedure, where the Kasai procedure effectively gives them a functional bile duct when they born either without a bile duct or one that is completely blocked at birth.
  • oral formulations of the present invention may include other agents known to those skilled in the art of pharmacy, having regard for the type of formulation in issue.
  • Oral formulations suitable may include flavoring agents.
  • the present invention relates to a pharmaceutical formulation of obeticholic acid or a pharmaceutically acceptable salt, solvate, or amino acid conjugate thereof, wherein obeticholic acid is produced by a process of the invention (obeticholic acid Form 1).
  • the formulation is administered orally.
  • the formulation is in tablet form.
  • the formulation comprises obeticholic acid and one or more components selected from microcrystalline cellulose, sodium starch glycolate, magnesium stearate, coating material, or colloidal silicon dioxide.
  • the coating material is an Opadry® coating material.
  • the formulation comprises about 0.1 mg to about 1500 mg of obeticholic acid per tablet. In another embodiment, the formulation comprises about 1 mg to about 100 mg. In another embodiment, the formulation comprises about 1 mg to about 50 mg. In another embodiment, the formulation comprises about 1 mg to about 30 mg. In another embodiment, the formulation comprises about 4 mg to about 26 mg. In another embodiment, the formulation comprises about 5 mg to about 25 mg. In one embodiment, the formulation comprises about 1 mg to about 2 mg. In one embodiment, the formulation comprises about 1.2 mg to about 1.8 mg. In one embodiment, the formulation comprises about 1.3 mg to about 1.7 mg. In one embodiment, the formulation comprises about 1.5 mg.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 1 mg of obeticholic acid, about 180 to about 190 mg of microcrystalline cellulose, about 10 to about 15 mg of sodium starch glycolate, about 1 to about 3 mg of magnesium stearate, and about 5 mg to about 10 mg of coating material. In one embodiment, the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 1 mg of obeticholic acid, about 185.0 mg of microcrystalline cellulose, about 12.0 mg of sodium starch glycolate, about 2.0 mg of magnesium stearate, and about 8.0 mg of coating material. In one embodiment, the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 5 mg of obeticholic acid, about 175 to about 190 mg of microcrystalline cellulose, about 10 to about 15 mg of sodium starch glycolate, about 1 to about 3 mg of magnesium stearate, and about 5 mg to about 10 mg of coating material. In one embodiment, the coating material is an Opadry® coating material. In one embodiment, the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet.
  • the formulation comprises about 5 mg of obeticholic acid, about 181.0 mg of microcrystalline cellulose, about 12.0 mg of sodium starch glycolate, about 2.0 mg of magnesium stearate, and about 8.0 mg of coating material.
  • the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 10 mg of obeticholic acid, about 170 mg to about 180 mg of microcrystalline cellulose, about 10 mg to about 15 mg of sodium starch glycolate, about 1 mg to about 3 mg of magnesium stearate, and about 5 mg to about 10 mg of coating material. In one embodiment, the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 10 mg of obeticholic acid, about 176.0 mg of microcrystalline cellulose, about 12.0 mg of sodium starch glycolate, about 2.0 mg of magnesium stearate, and about 8.0 mg of coating material. In one embodiment, the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 25 mg of obeticholic acid, about 150 mg to about 160 mg of microcrystalline cellulose, about 10 mg to about 15 mg of sodium starch glycolate, about 1 mg to about 3 mg of magnesium stearate, about 5 to about 10 mg of coating material, and about 1 to about 10 mg of colloidal silicon dioxide. In one embodiment, the coating material is an Opadry® coating material.
  • the formulation comprises of about 1 mg to about 25 mg of obeticholic acid per tablet. In one embodiment, the formulation comprises about 25 mg of obeticholic acid, about 157.0 mg of microcrystalline cellulose, about 12.0 mg of sodium starch glycolate, about 2.0 mg of magnesium stearate, about 8.0 mg of coating material, and about 4.0 mg of colloidal silicon dioxide. In one embodiment, the coating material is an Opadry® coating material.
  • the percent dimeric impurity is on an area percent basis, typically as quantified by analytical HPLC.
  • compositions are described as having, including, or comprising specific components, it is contemplated that compositions also consist essentially of, or consist of, the recited components.
  • methods or processes are described as having, including, or comprising specific process steps, the processes also consist essentially of, or consist of, the recited processing steps.
  • steps or order for performing certain actions is immaterial so long as the invention remains operable.
  • two or more steps or actions can be conducted simultaneously.
  • API Active pharmaceutical ingredient
  • JP Japanese Pharmacopeia
  • the tablet comprises yellow Opadry®. In another embodiment, the tablet comprises white Opadry®. In another embodiment, the tablet comprises green Opadry®.
  • Obeticholic acid including obeticholic acid Form 1, substantially pure forms of obeticholic acid and crystalline forms of obeticholic acid, or a pharmaceutically acceptable salt, solvate, or amino acid conjugate thereof is useful for a variety of medicinal purposes.
  • Obeticholic acid may be used in methods for the prevention or treatment of FXR mediated diseases and conditions.
  • the disease or condition is selected from biliary atresia, cholestatic liver disease, chronic liver disease, nonalcoholic steatohepatitis (NASH), hepatitis C infection, alcoholic liver disease, primary biliary cirrhosis (PBC), liver damage due to progressive fibrosis, liver fibrosis, and cardiovascular diseases including atherosclerosis, arteriosclerosis,
  • obeticholic acid Form 1 may be used in methods for lowering triglycerides.
  • crystalline obeticholic acid may be used in methods for lowering triglycerides.
  • Obeticholic acid Form 1 or crystalline obeticholic acid may increase HDL.
  • Other effects of obeticholic acid Form 1 or crystalline obeticholic acid include lowering of alkaline phosphatase (ALP), bilirubin, ALT, AST, and GGT.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising obeticholic acid and a pharmaceutically acceptable carrier, wherein the obeticholic acid is produced by a process of the invention, e.g. , obeticholic acid Form 1.
  • the pharmaceutical composition comprises of substantially pure obeticholic acid and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises of crystalline obeticholic acid and a pharmaceutically acceptable carrier.
  • the crystalline obeticholic acid is the Form A.
  • the crystalline obeticholic acid is the Form C.
  • the crystalline obeticholic acid is Form D.
  • the crystalline obeticholic acid is Form F.
  • the crystalline obeticholic acid is Form G.
  • the crystalline obeticholic acid is Form I.
  • the present invention relates to a method of treating or preventing an FXR mediated disease or condition in a subject comprising administering an effective amount of obeticholic acid Form 1 produced by a process of the invention or a pharmaceutical composition thereof. In one embodiment, the present invention relates to a method of treating or preventing an FXR mediated disease or condition in a subject comprising administering an effective amount of substantially pure obeticholic acid produced by a process of the invention or a pharmaceutical composition thereof. In one embodiment, the present invention relates to a method of treating or preventing an FXR mediated disease or condition in a subject comprising administering an effective amount of crystalline obeticholic acid or a pharmaceutical composition thereof.
  • the crystalline obeticholic acid is Form C. In one embodiment, the crystalline obeticholic acid is Form A. In one embodiment, the crystalline obeticholic acid is Form C. In one embodiment, the crystalline obeticholic acid is Form D. In one embodiment, the crystalline obeticholic acid is Form F. In one embodiment, the crystalline obeticholic acid is Form G. In one embodiment, the crystalline obeticholic acid is Form I.
  • the disease or condition is cardiovascular disease or cholestatic liver disease and for lowering triglycerides.
  • the cardiovascular disease is atherosclerosis or hypercholesterolemia.
  • the subject is a mammal. In another embodiment, the mammal is human.
  • the compound or pharmaceutical composition is administered orally, parenterally, or topically. In another embodiment, the compound or pharmaceutical composition is administered orally.
  • the present invention relates to a method for inhibiting fibrosis in a subject who is suffering from a cholestatic condition, the method comprising the step of administering to the subject an effective amount of obeticholic acid or a pharmaceutical composition thereof, wherein obeticholic acid is produced by the process of the invention.
  • the present invention relates to a method for inhibiting fibrosis in a subject who is not suffering from a cholestatic condition, the method comprising the step of administering to the subject an effective amount of obeticholic acid or a pharmaceutical composition thereof, wherein obeticholic acid is produced by the process of the invention.
  • the fibrosis to be inhibited occurs in an organ where FXR is expressed.
  • the cholestatic condition is defined as having abnormally elevated serum levels of alkaline phosphatase, 7-glutamyl transpeptidase (GGT), and 5' nucleotidase.
  • the cholestatic condition is further defined as presenting with at least one clinical symptom.
  • the symptom is itching (pruritus).
  • the fibrosis is selected from the group consisting of liver fibrosis, kidney fibrosis, and intestinal fibrosis.
  • the cholestatic condition is selected from the group consisting of primary biliary cirrhosis, primary sclerosing cholangitis, drug-induced cholestasis, hereditary cholestasis, and intrahepatic cholestasis of pregnancy.
  • the subject is not suffering from a cholestatic condition associated with a disease or condition selected from the group consisting of primary liver and biliary cancer, metastatic cancer, sepsis, chronic total parenteral nutrition, cystic fibrosis, and granulomatous liver disease.
  • the subject has liver fibrosis associated with a disease selected from the group consisting of hepatitis B; hepatitis C; parasitic liver diseases; post-transplant bacterial, viral and fungal infections; alcoholic liver disease (ALD); non- alcoholic fatty liver disease (NAFLD); non-alcoholic steatohepatitis (NASH); liver diseases induced by methotrexate, isoniazid, oxyphenistatin, methyldopa,
  • a disease selected from the group consisting of hepatitis B; hepatitis C; parasitic liver diseases; post-transplant bacterial, viral and fungal infections; alcoholic liver disease (ALD); non- alcoholic fatty liver disease (NAFLD); non-alcoholic steatohepatitis (NASH); liver diseases induced by methotrexate, isoniazid, oxyphenistatin, methyldopa,
  • chlorpromazine, tolbutamide, or amiodarone autoimmune hepatitis; sarcoidosis; Wilson's disease; hemochromatosis; Gaucher's disease; types III, IV, VI, IX and X glycogen storage diseases; ai-antitrypsin deficiency; Zellweger syndrome; tyrosinemia;
  • fructosemia galactosemia
  • vascular derangement associated with Budd-Chiari syndrome, veno-occlusive disease, or portal vein thrombosis and congenital hepatic fibrosis.
  • the subject has intestinal fibrosis associated with a disease selected from the group consisting of Crohn's disease, ulcerative colitis, post-radiation colitis, and microscopic colitis.
  • the subject has renal fibrosis associated with a disease selected from the group consisting of diabetic nephropathy, hypertensive nephrosclerosis, chronic glomerulonephritis, chronic transplant glomerulopathy, chronic interstitial nephritis, and polycystic kidney disease.
  • a disease selected from the group consisting of diabetic nephropathy, hypertensive nephrosclerosis, chronic glomerulonephritis, chronic transplant glomerulopathy, chronic interstitial nephritis, and polycystic kidney disease.
  • OCA obeticholic acid
  • obeticholic acid examples include: 3a,7a-dihydroxy-6a-ethyl-5 -cholan-24-oic acid, 6a-ethyl-chenodeoxycholic acid, 6-ethyl-CDCA, 6ECDCA, cholan-24-oic acid,6-ethyl-3,7-dihydroxy-,(3a,5 , 6a,7a)- and INT-747.
  • the CAS registry number for obeticholic acid is 459789-99-2. This term refers to all forms of obeticholic acid, e.g., non-crystalline, crystalline and substantially pure.
  • crystalline obeticholic acid refers to any crystalline form of a compound having the chemical structure:
  • stalline obeticholic acid means that the compound is crystallized into a specific crystal packing arrangement in three spatial dimensions or the compound having external face planes.
  • the crystalline form of obeticholic acid (or a pharmaceutically acceptable salt, amino acid conjugate, solvate thereof) can crystallize into different crystal packing arrangements, all of which have the same elemental composition of obeticholic acid.
  • Different crystal forms usually have different X-ray diffraction patterns, infrared spectral, melting points, density hardness, crystal shape, optical and electrical properties, stability and solubility. Recrystallization solvent, rate of crystallization, storage temperature, and other factors may cause one crystal form to dominate. Crystals of obeticholic acid can be prepared by crystallization under different conditions, e.g., different solvents, temperatures, etc.
  • crystalline obeticholic acid Form A refers to a crystalline form of obeticholic acid with an X-ray diffraction pattern that is substantially similar to that set forth in Figure 41, e.g., one of the crystalline forms as characterized in Example 3.
  • crystalline obeticholic acid Form C refers to a crystalline form of obeticholic acid with an X-ray diffraction pattern that is substantially similar to that set forth in Figure 5, e.g., one of the crystalline forms as characterized in Example 3.
  • the term "substantially pure obeticholic acid” refers to obeticholic acid that has a potency of greater than about 95%.
  • the potency of the obeticholic acid takes into account impurities including e.g., water, solvents, and other organic and inorganic impurities that are in a sample of obeticholic acid.
  • the known standard for potency is 100% obeticholic acid, and the potency is determined by subtracting percentages of impurities such as solvent, water, and other organic and inorganic impurities from 100% of the known standard.
  • the inorganic impurities include e.g., inorganic salts and sulphated ash.
  • the organic impurities include 6-ethylursodeoxycholic acid, 3a-hydroxy-6a-ethyl-7-cheto-5p-cholan- 24-oic acid, 6p-ethylchenodeoxycholic acid, 3a,7a-dihydroxy-6-ethyliden-5p-cholan-24- oic acid, chenodeoxycholic acid, and 3a(3a,7a-dihydroxy-6a-ethyl-5p-cholan-24- oyloxy)-7a-hydroxy-6a-ethyl-5p-cholan-24-oic acid.
  • the amounts of the impurities can be determined by procedures known in the art, e.g., HPLC, NMR, or methods from US Pharmacopeia, or European Pharmacopeia, or a combination of two or more of these methods.
  • the term "purity" refers to a chemical analysis of a compound obtained from e.g., HPLC.
  • the purity of a compound is compared to the purity of the reference standard, e.g., obeticholic acid, via the area under their respective peak for comparisons.
  • purity accounts for the organic impurities in a sample.
  • reaction mixture refers a mixture of one or more substances combined together.
  • the mixing or combining of the substances causes a chemical transformation or change in one or more of the original substances.
  • obeticholic acid Form 1 refers to non-crystalline obeticholic acid.
  • this form of obeticholic acid is produced via a crystalline obeticholic acid as a synthetic intermediate.
  • this form of obeticholic acid is produced by the process of the application via crystalline obeticholic acid Form A as the synthetic intermediate.
  • obeticholic acid Form 1 is the form that it used as the pharmaceutically active ingredient. See Example 5 for more details.
  • Treating includes any effect, e.g. , lessening, reducing, modulating, or eliminating, that results in the improvement of the condition, disease, disorder, etc.
  • Treating" or “treatment” of a disease state includes: inhibiting the disease state, i.e. , arresting the development of the disease state or its clinical symptoms; or relieving the disease state, i.e. , causing temporary or permanent regression of the disease state or its clinical symptoms.
  • Preventing the disease state includes causing the clinical symptoms of the disease state not to develop in a subject that may be exposed to or predisposed to the disease state, but does not yet experience or display symptoms of the disease state.
  • Disease state means any disease, disorder, condition, symptom, or indication.
  • effective amount refers to an amount of obeticholic acid (e.g. , an FXR-activating ligand) that produces an acute or chronic therapeutic effect upon appropriate dose administration. The effect includes the prevention, correction, inhibition, or reversal of the symptoms, signs and underlying pathology of a
  • disease/condition e.g., fibrosis of the liver, kidney, or intestine
  • a therapeutically effective amount means the amount of obeticholic acid that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on obeticholic acid, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • a therapeutically effective amount of obeticholic acid can be formulated with a pharmaceutically acceptable carrier for administration to a human or an animal.
  • obeticholic acid or its formulations can be administered, for example, via oral, parenteral, or topical routes, to provide an effective amount of the compound.
  • obeticholic acid prepared in accordance with the present invention can be used to coat or impregnate a medical device, e.g., a stent.
  • pharmacological effect means that primary indications of the subject being treated are prevented, alleviated, or reduced.
  • a pharmacological effect would be one that results in the prevention, alleviation or reduction of primary indications in a treated subject.
  • a pharmacological effect means that disorders or symptoms of the primary indications of the subject being treated are prevented, alleviated, or reduced.
  • a pharmacological effect would be one that results in the prevention or reduction of primary indications in a treated subject.
  • the invention also comprehends isotopically-labeled obeticholic acid, or pharmaceutically acceptable salts, solvate, or amino acid conjugates thereof, which are identical to those recited in formulae of the invention and following, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature.
  • isotopes that can be incorporated into obeticholic acid, or pharmaceutically acceptable salts, solvate, or amino acid conjugates thereof include isotopes of hydrogen, carbon, nitrogen, fluorine, such as H, n C, 14 C and 18 F.
  • Obeticholic acid, or pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of the present invention.
  • Isotopically-labeled obeticholic acid, or pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof, for example those into which radioactive isotopes such as 3 ⁇ 4, 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e. , H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • isotopically labeled obeticholic acid, or pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples of the invention, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • obeticholic acid, or pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof are not isotopically labelled.
  • deuterated obeticholic acid is useful for bioanalytical assays.
  • obeticholic acid, or pharmaceutically acceptable salts, solvates, or amino acid conjugates thereof are radiolabelled.
  • “Geometric Isomers” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules.
  • Solidvates means solvent addition forms that contain either stoichiometric or non stoichiometric amounts of solvent.
  • Obeticholic acid may have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H2O, such combination being able to form one or more hydrate.
  • the compounds of the present invention can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
  • hydrates include monohydrates, dihydrates, etc.
  • solvates include ethanol solvates, acetone solvates, etc.
  • obeticholic acid may be depicted as different tautomers. It should also be understood that when obeticholic acid and synthetic intermediates of the invention have tautomeric forms, all tautomeric forms are intended to be within the scope of the invention, and the naming of obeticholic acid does not exclude any tautomer form.
  • Obeticholic acid and synthetic intermediates of the invention can exist in several tautomeric forms, including the keto-enol.
  • keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs.
  • Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present invention includes all tautomers of the present compounds.
  • isomers arising from asymmetric carbon atoms are included within the scope of the invention, unless indicated otherwise.
  • Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically controlled synthesis.
  • the structures and other compounds and moieties discussed in this application also include all tautomers thereof.
  • Alkenes can include either the E- or Z- geometry, where appropriate.
  • Obeticholic acid and synthetic intermediates may exist in stereoisomeric form, and therefore can be produced as individual stereoisomers or as mixtures.
  • a “pharmaceutical composition” is a formulation containing obeticholic acid in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form. It is can be advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active reagent calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active reagent and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active agent for the treatment of individuals.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler, or a vial.
  • the quantity obeticholic acid (e.g. , a formulation of obeticholic acid, or a pharmaceutically acceptable salt, solvate, or amino acid conjugate thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved.
  • obeticholic acid e.g. , a formulation of obeticholic acid, or a pharmaceutically acceptable salt, solvate, or amino acid conjugate thereof
  • the dosage will also depend on the route of administration.
  • routes including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • obeticholic acid is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
  • flash dose refers to obeticholic acid formulations that are rapidly dispersing dosage forms.
  • immediate release is defined as a release of obeticholic acid from a dosage form in a relatively brief period of time, generally up to about 60 minutes.
  • modified release is defined to include delayed release, extended release, and pulsed release.
  • pulsed release is defined as a series of releases of drug from a dosage form.
  • a "subject” includes mammals, e.g. , humans, companion animals (e.g. , dogs, cats, birds, and the like), farm animals (e.g. , cows, sheep, pigs, horses, fowl, and the like) and laboratory animals (e.g., rats, mice, guinea pigs, birds, and the like).
  • the subject is human.
  • the subject is human child (e.g., between about 30 kg to about 70 kg).
  • the human child has had a Kasai procedure, where the Kasai procedure effectively gives them a functional bile duct when they bom either without a bile duct or one that is completely blocked at birth.
  • the phrase "pharmaceutically acceptable” refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • a "pharmaceutically acceptable excipient” as used in the specification and claims includes both one and more than one such excipient.
  • obeticholic acid is usually administered in the form of pharmaceutical formulations comprising a pharmaceutically acceptable excipient and obeticholic acid. These formulations can be administered by a variety of routes including oral, buccal, rectal, intranasal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Oral formulation of obeticholic acid are described further herein under the section entitled "Oral Formulation and Administration".
  • obeticholic acid can be administered transdermally.
  • a transdermal delivery device (“patch") is needed.
  • Such transdermal patches may be used to provide continuous or discontinuous infusion of a compound of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g. , U.S. Patent No. 5,023,252. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • a pharmaceutical formulation comprising at least obeticholic acid as described above in a formulation adapted for buccal and/or sublingual, or nasal administration.
  • This embodiment provides administration of obeticholic acid in a manner that avoids gastric complications, such as first pass metabolism by the gastric system and/or through the liver. This administration route may also reduce adsorption times, providing more rapid onset of therapeutic benefit.
  • the compounds of the present invention may provide particularly favorable solubility profiles to facilitate sublingual/buccal formulations.
  • Such formulations typically require relatively high concentrations of active ingredients to deliver sufficient amounts of active ingredients to the limited surface area of the sublingual/buccal mucosa for the relatively short durations the formulation is in contact with the surface area, to allow the absorption of the active ingredient.
  • the very high activity of obeticholic acid combined with its high solubility, facilitates its suitability for sublingual/buccal formulation.
  • Obeticholic acid is preferably formulated in a unit dosage form, each dosage containing from about 0.1 mg to about 1500 mg.
  • the formulation comprises about 1 mg to about 100 mg.
  • the formulation comprises about 1 mg to about 50 mg.
  • the formulation comprises about 1 mg to about 30 mg.
  • the formulation comprises about 4 mg to about 26 mg.
  • the formulation comprises about 5 mg to about 25 mg.
  • the formulation comprises about 1 mg to about 2 mg.
  • the formulation comprises about 1.2 mg to about 1.8 mg.
  • the formulation comprises about 1.3 mg to about 1.7 mg. In one embodiment, the formulation comprises about 1.5 mg.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient as described above.
  • Obeticholic acid is generally effective over a wide dosage range. For examples, dosages per day normally fall within the range of about 0.0001 to about 30 mg/kg of body weight. In the treatment of adult humans, the range of about 0.1 to about 15 mg/kg/day, in single or divided dose, is especially preferred.
  • the formulation comprises about 0.1 mg to about 1500 mg. In another embodiment, the formulation comprises about 1 mg to about 100 mg. In another embodiment, the formulation comprises about 1 mg to about 50 mg.
  • the formulation comprises about 1 mg to about 30 mg. In another embodiment, the formulation comprises about 4 mg to about 26 mg. In another embodiment, the formulation comprises about 5 mg to about 25 mg. In one embodiment, the formulation comprises about 1 mg to about 2 mg. In one embodiment, the formulation comprises about 1.2 mg to about 1.8 mg. In one embodiment, the formulation comprises about 1.3 mg to about 1.7 mg. In one embodiment, the formulation comprises about 1.5 mg.
  • the amount of obeticholic acid actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the form of obeticholic acid administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several smaller doses for administration throughout the day.
  • Process of the invention refers to a method for preparing obeticholic acid as described herein, wherein the method comprises of crystalline obeticholic acid.
  • Fibrosis refers to a condition involving the development of excessive fibrous connective tissue, e.g. , scar tissue, in a tissue or organ. Such generation of scar tissue may occur in response to infection, inflammation, or injury of the organ due to a disease, trauma, chemical toxicity, and so on. Fibrosis may develop in a variety of different tissues and organs, including the liver, kidney, intestine, lung, heart, etc.
  • inhibiting refers to any detectable positive effect on the development or progression of a disease or condition. Such a positive effect may include the delay or prevention of the onset of at least one symptom or sign of the disease or condition, alleviation or reversal of the symptom(s) or sign(s), and slowing or prevention of the further worsening of the symptom(s) or sign(s).
  • a “cholestatic condition” refers to any disease or condition in which bile excretion from the liver is impaired or blocked, which can occur either in the liver or in the bile ducts.
  • Intrahepatic cholestasis and extrahepatic cholestasis are the two types of cholestatic conditions. Intrahepatic cholestasis (which occurs inside the liver) is most commonly seen in primary biliary cirrhosis, primary sclerosing cholangitis, sepsis (generalized infection), acute alcoholic hepatitis, drug toxicity, total parenteral nutrition (being fed intravenously), malignancy, cystic fibrosis, and pregnancy.
  • Extrahepatic cholestasis (which occurs outside the liver) can be caused by bile duct tumors, strictures, cysts, diverticula, stone formation in the common bile duct, pancreatitis, pancreatic tumor or pseudocyst, and compression due to a mass or tumor in a nearby organ.
  • Clinical symptoms and signs of a cholestatic condition include: itching (pruritus), fatigue, jaundiced skin or eyes, inability to digest certain foods, nausea, vomiting, pale stools, dark urine, and right upper quadrant abdominal pain.
  • a patient with a cholestatic condition can be diagnosed and followed clinically based on a set of standard clinical laboratory tests, including measurement of levels of alkaline phosphatase, ⁇ -glutamyl transpeptidase (GGT), 5' nucleotidase, bilirubin, bile acids, and cholesterol in a patient's blood serum.
  • GTT ⁇ -glutamyl transpeptidase
  • a patient is diagnosed as having a cholestatic condition if serum levels of all three of the diagnostic markers alkaline phosphatase, GGT, and 5' nucleotidase, are considered abnormally elevated.
  • the normal serum level of these markers may vary to some degree from laboratory to laboratory and from procedure to procedure, depending on the testing protocol.
  • a physician will be able to determine, based on the specific laboratory and test procedure, what is an abnormally elevated blood level for each of the markers.
  • a patient suffering from a cholestatic condition generally has greater than about 125 IU/L alkaline phosphatase, greater than about 65 IU/L GGT, and greater than about 17 NIL 5' nucleotidase in the blood.
  • a cholestatic condition may be diagnosed on the basis of abnormal levels of these three markers in addition to at least one of the symptoms mentioned above, such as itching (pruritus).
  • organ refers to a differentiated structure (as in a heart, lung, kidney, liver, etc.) consisting of cells and tissues and performing some specific function in an organism. This term also encompasses bodily parts performing a function or cooperating in an activity (e.g. , an eye and related structures that make up the visual organs). The term “organ” further encompasses any partial structure of differentiated cells and tissues that is potentially capable of developing into a complete structure (e.g. , a lobe or a section of a liver).
  • Step 1 Preparation of 3a-hydroxy-7-keto-5B-cholan-24-oic acid methyl ester (1):
  • Reaction 1 Esterification of C-24 carboxylic acid of 7-keto lithocholic acid (KLCA) 3a-hydroxy-7-keto-5 -cholan-24-oic acid (KLCA; 500.0 g, 1.28 mol) was esterified using methyl alcohol (2500 mL), in the presence of acidic catalysis (sulfuric acid, 1.0 mL) and was heated up to 62 °C to 64 °C for approximately 3 hours, to yield 3a- hydroxy-7-keto-5 -cholan-24-oic acid methyl ester (1). In this reaction, the methyl alcohol acts as the methylating reagent as well as the reaction solvent.
  • KLCA 7-keto lithocholic acid
  • KLCA 3a-hydroxy-7-keto-5 -cholan-24-oic acid
  • the pH-value was adjusted with sodium hydroxide solution (2N) to pH 9.5 to 10.
  • the solution was treated with activated carbon (25 g) for approximately 30 minutes and filtered to remove the carbon solids. Alternatively, the solution was not treated with activated carbon.
  • water (625 mL) at 10 °C to 15 °C was added over 15 minutes and seeding material was added. The reaction mixture is stirred for 1 hour at 10 °C to 15 °C. Another portion of water (1875 mL) was added over about 20 to 25 minutes. The product suspension was stirred for 30 minutes at 10 °C to 15 °C.
  • the product was isolated with a centrifuge and washed with a mixture of methanol and water (1 : 1, 350 mL).
  • the water content of the wet material was quantified by Karl Fischer (KF).
  • the material was dried in a tumble dryer under vacuum at NMT 70 °C. The material can also be used in the next step without drying.
  • the yield (calculated on dried product) is 501.4 g (1.24 mol, 96.8%).
  • chlorotrimethylsilane was charged into the LDA solution at -20 °C to
  • reaction mixture was stirred for approximately 2 hours.
  • the reaction mixture was added to a pre-cooled aqueous solution of citric acid (34.6 g in 300 mL) at 2 °C to 8 °C. After the addition, the aqueous phase was separated and discarded. From the organic phase, the liquid was removed by vacuum distillation at maximum 50 °C. The isolated residue contained compound 3 and some residual solvents and was used 'as is' in the next step.
  • Reaction 3 Aldol condensation of the silylenol ether and acetaldehyde Compound 3 (164.68 g, 300 mmol, calculated as dried substance) solution in THF was charged into an inert reactor. At a maximum temperature of 50 °C, residual amounts of THF were distilled off under vacuum. The water content in the residue was limited to ⁇ 0.5% (Karl Fischer titration) in order to proceed. The residue was then dissolved in dichloromethane (200 mL) and pre-cooled to -60 °C to -65 °C. Acetaldehyde (33.8 mL, 600 mmol) was then added.
  • dichloromethane (700 mL) and boron trifluoride (16 wt% solution in acetonitrile, 318 g, 750 mmol) acetonitrile complex were charged into a separate reactor and then cooled to -60 °C to -65 °C. At -60 °C to -65 °C, the dry compound 3 solution was added. The reaction mixture was stirred for approximately two hours at -60 °C to -65 °C, heated up to 23 °C to 28 °C, stirred for another approximately 3 hours and cooled to approximately 2 °C to 10 °C for the hydrolysis/work-up.
  • the cooled solution from the reactor was added to a pre-cooled aqueous solution of 50% wt. caustic soda (40 mL) and 660 mL of water. After about 10 minutes of intensive stirring, the phases were separated and the (lower) organic layer was transferred to a separate reactor. From the organic layer, the solvent was removed by distillation at NMT 50 °C as far as possible. The residue, consisting of compound 4 and some remaining acetonitrile and dichloromethane, was discharged into drums.
  • Compound 4A a mixture of E/Z-isomers can also be prepared by the procedure described above for Step 3.
  • reaction mixture was allowed to rest for at least 30 minutes. The phases were separated and the lower aqueous layer was transferred into a separate reactor and the organic layer was discarded. Ethyl acetate (1400 mL) and aqueous citric acid (244 g in 480 mL) were added with intensive stirring to the aqueous layer. The reaction mixture was stirred at 25 °C to 35 °C for 10 minutes. The phases were separated and the lower aqueous layer was discarded. Ethyl acetate was distilled off from the organic layer and replaced with ethyl acetate (800 mL). This operation was repeated until the water content of the distillate was NMT 1% or until a constant boiling point was reached.
  • Crude compound 5 was then crystallized using ethanol.
  • the crude compound for crystallization can also be a mixture of E/Z isomers, compound 5 A.
  • Ethanol (390 to 520 mL) and crude compound 5 (130 g) were charged into an inert reactor. To dissolve the crude compound 5, the reaction mixture was heated to reflux. Then, the reaction mixture was cooled in a controlled cooling ramp to 15 °C to 20 °C within 3 to 5 hours by a linear profile.
  • the crystalline compound 5A was isolated using a centrifuge and then washed with ethyl acetate (50-100 mL, 2 times). Drying was done in the tumble dryer under vacuum and at approximately 60 °C. This leads to 85.8 g (66%) yield.
  • Purified compound 5 is the E isomer of 3a-hydroxy-6-ethylidene-7-keto-5 -cholan-24-oic acid. See example 2 for full details regarding the identification and characterization of purified compound 5. Isolation of the purified compound 5, the E isomer, can be optional. The E isomer and Z isomers have different solubilities. The E isomer is less soluable and crystallizes such that the Z isomer can be washed away.
  • a mixture of purified compound 5 (110 g, 264 mmol), water (1100 mL), caustic soda solution (35.8 mL, 682 mmol) at 50% and palladium catalyst (Pd/C, 11 g) were charged to a hydrogenation reactor.
  • a hydrogen pressure of 5 bar was applied and the reaction mixture was heated up to 100 °C (for isomerisation to the alpha position) over a period of 1.5 hours and then stirred for 3 hours while maintaining the hydrogen pressure at 4.5 to 5 bar.
  • the reaction mixture is then cooled to 40 °C to 50 °C.
  • the Pd/C is filtered off.
  • n- butyl acetate (1320 mL) and hydrochloric acid (67.8 mL, 815 mmol, 37%) were added.
  • the aqueous phase was separated and discarded.
  • the organic phase was treated with activated carbon (5.5 g) for about 10 minutes at 40 to 50 °C.
  • the activated carbon was filtered off and the filtrate was condensed by distillation and the resulting suspension was cooled to 15 °C to 20 °C within 2 to 3 hours.
  • the precipitated compound 6 was isolated and washed with n-butyl acetate (160 mL).
  • n-butyl acetate 860 mL
  • citric acid 320.2 g, anhydrous
  • the organic phase was transferred and distilled.
  • the residue is diluted with n-butyl acetate and was slowly cooled to 15 °C to 20 °C and the crude obeticholic acid was filtered using a centrifuge.
  • the wet product was crystallized from n-butyl acetate.
  • the product obeticholic acid was isolated and washed with n-butyl acetate (43 mL, 4 times) in an inert pressure filter. Drying was done in the pressure filter under vacuum at approximately 80 °C. This led to 67.34 g (77.9%) of crystalline obeticholic acid characterized by an X-ray diffraction partem including characteristic peaks at about 5.0 and 5.3 degrees 2-Theta.
  • the crystalline obeticholic acid Form A is characterized by an X-ray diffraction partem substantially similar to that set forth in Figure 41. See example 3 for full details regarding the identification and characterization of crystalline obeticholic acid.
  • Step 7 Preparation of obeticholic acid Form 1 :
  • Reaction 7 Preparation of obeticholic acid Form 1 from crystalline obeticholic acid Form A
  • Crystalline obeticholic acid Form A characterized by an X-ray diffraction pattern including characteristic peaks at about 5.0 and 5.3 degrees
  • 2-Theta (58 g) was dissolved in water (870 mL) and caustic soda solution (50%, 8.7 mL, 166 mmol) at 30 °C to 40 °C. The mixture was stirred until all solid has dissolved. The product was precipitated using the following workup.
  • the obeticholic acid solution was slowly added via a filter to diluted hydrochloric acid (37%, 16.05 mL, 193 mmol) in water (870 mL) at 30 °C to 40 °C.
  • Compound 5 is the key intermediate for the process of the application.
  • the compound was isolated from ethyl acetate and was then crystallized from ethanol.
  • the highly pure compound 5 allows for efficient and high yielding production of compound 6 and subsequently crystalline obeticholic acid Form A characterized by an X-ray diffraction partem including characteristic peaks at about 5.0 and 5.3 degrees 2-Theta and obeticholic acid Form 1, including substantially pure obeticholic acid.
  • Figures 1 and 2 are UPLC UV/MS chromatograms for "crude compound 5" ( Figure 1) and compound 5 "purified reference" ( Figure 2) obtained on a high performance UPLC column.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C. n-Butyl acetate (60 uL) was added and observations were made. The sample was stirred at 50 °C for 1 hour and was cooled to 5 °C using a linear cooling rate of 0.1 °C/min. The sample was stirred at 5 °C for 2 days. Solids were filtered, air dried and analyzed by XRPD. The solution was stored in the freezer for -48 hours, and then left to evaporate at ambient conditions to afford Form A. Additional methods of preparing Form A are provided in Example 9.
  • Amorphous obeticholic acid (approximately 1 g) was weighed into a large glass vial and warmed to 50 °C. Ethyl acetate (1.5 ml, 1.5 volumes) was added at 50 °C, forming a clear solution. Anti-solvent (heptane, 2.0 ml, 2 volumes) was added, initially forming a cloudy precipitate, which converted to an oil on stirring. After stirring at 50 °C for 1 hour the oil remained so an additional 0.5 ml (0.5 volumes) ethyl acetate was added, which dissolved the oil, forming a cloudy liquid. The sample was seeded with approximately 1 mg of sample from the small scale experiment directly above.
  • the instrument was calibrated for energy and temperature using certified indium. Typically 0.5-1 mg of each sample, in a pin-holed aluminium pan, was heated at 10 -min "1 from 25 °C to 350 °C. A nitrogen purge at 50 ml min "1 was maintained over the sample.
  • the instrument control and data analysis software was STARe v 9.20.
  • TGA/SDTA 85 le equipped with a 34 position auto-sampler The instrument was temperature calibrated using certified indium. Typically 5-10 mg of each sample was loaded onto a pre-weighed aluminium crucible and was heated at 10 "C min "1 from ambient temperature to 300 °C. A nitrogen purge at 50 ml-min "1 was maintained over the sample.
  • the instrument control and data analysis software was STARe v 9.20.
  • Crystalline obeticholic acid Form C contained between 0.15 and 0.2 moles solvent (heptane) and ca. 1.5 % w/w (0.3 moles).
  • the DSC thermogram of crystalline obeticholic acid Form C contained one endotherm. This was fairly sharp and had an onset of around 98 °C. See Figure 6. Different solvents would have different boiling points and therefore would evaporate at different temperatures within the DSC and TGA experiments.
  • X-Ray Powder Diffraction patterns were collected on a Bruker AXS C2 GADDS diffractometer using Cu Ka radiation (40 kV, 40 mA), automated XYZ stage, laser video microscope for auto sample positioning and a HiStar 2-dimensional area detector.
  • X-ray optics consisted of a single Gobel multilayer mirror coupled with a pinhole collimator of
  • the beam divergence i.e. the effective size of the X-ray beam on the sample, was approximately 4 mm.
  • a ⁇ - ⁇ continuous scan mode was employed with a sample - detector distance of 20 cm which gives an effective 2 ⁇ range of 3.2 ° - 29.7 °.
  • the sample was exposed to the X-ray beam for 120 seconds.
  • the software used for data collection was GADDS for WNT 4.1.16 and the data were analyzed and presented using
  • Non-ambient conditions Samples run under non-ambient conditions were mounted on a silicon wafer with heat-conducting compound. The sample was then heated to the appropriate temperature at ca. 10 "C min "1 and subsequently held isothermally for ca. 1 minute before data collection was initiated.
  • X-Ray Powder Diffraction patterns were collected on a Siemens D5000 diffractometer using Cu Ka radiation (40 kV, 40 mA), ⁇ - ⁇ goniometer, divergence of V20 and receiving slits, a graphite secondary monochromator and a scintillation counter.
  • the instrument is performance checked using a certified Corundum standard (NIST 1976).
  • the software used for data collection was Diffrac Plus XRD Commander v2.3.1 and the data were analyzed and presented using Diffrac Plus EVA v 11,0.0.2 or v 13.0.0.2.
  • X-Ray Powder Diffraction patterns were collected on a Bruker D8 diffractometer using Cu Ka radiation (40 kV, 40 mA), ⁇ -2 ⁇ goniometer, and divergence of V4 and receiving slits, a Ge monochromator and a Lynxeye detector.
  • the instrument is performance checked using a certified Corundum standard (NIST 1976).
  • the software used for data collection was Diffrac Plus XRD Commander v 2.5.0 and the data were analyzed and presented using Diffrac Plus EVA v 11.0.0.2 or v 13.0.0.2.
  • VT-XRPD Very Temperature-X-ray Diffraction revealed that the endotherm seen in the DSC thermogram corresponded to the desolvation of the sample as no form changes were observed on heating.
  • VT-XRPD shows that drying of the solvent from the material resulted in loss of crystallinity which is consistent with the material being in a solvated form. See Figure 7.
  • Sorption isotherms were obtained using a SMS DVS Intrinsic moisture sorption analyzer, controlled by DVS Intrinsic Control software v 1.0.0.30.
  • the sample temperature was maintained at 25 °C by the instrument controls.
  • the humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 200 ml-min "1 .
  • the relative humidity was measured by a calibrated Rotronic probe (dynamic range of 1.0-100% RH), located near the sample.
  • the weight change, (mass relaxation) of the sample as a function of % RH (relative humidity) was constantly monitored by the microbalance (accuracy ⁇ 0.005 mg).
  • the water content of each sample was measured on a Mettler Toledo DL39 Coulometer using Hydranal Coulomat AG reagent and an argon purge. Weighed solid samples were introduced into the vessel on a platinum TGA pan which was connected to a subaseal to avoid water ingress. Approximately 10 mg of sample was used per titration and duplicate determinations were made.
  • the stability of obeticholic acid at 40 °C and 75% RH was determined as follows. A sample of obeticholic acid was stored in a humidity chamber for one week at 40 °C/75 % RH. The sample was re-analyzed by XRPD and was found to have been unchanged.
  • the table below shows the quantitative composition of obeticholic acid tablets.
  • the 5 mg, 10 mg, and 25 mg formulations have been used as phase 3 clinical trial material.
  • API Active pharmaceutical ingredient
  • JP Japanese Pharmacopeia
  • API is anhydrous and 100% pure; actual amount is adjusted based on the potency of the drug substance Lot used, and amount of microcrystalline cellulose is correspondingly decreased.
  • Obeticholic acid Form 1 refers to the non-crystalline form of obeticholic acid. This form of obeticholic acid can be produced via a crystalline obeticholic acid as a synthetic intermediate. Obeticholic acid Form 1 can be used as the pharmaceutically active ingredient. Obeticholic acid Form 1 was characterized and analyzed as follows.
  • Batch 1 of obeticholic acid form 1 was characterized using the following techniques: assessment by X-ray powder diffraction (XPRD) for crystallinity, 3 ⁇ 4 and 1 C nuclear magnetic resonance (NMR), Fourier transform infrared spectroscopy (FT-IR), optical assessment (e.g., particle shape/size), thermal properties (e.g., differential scanning calorimetry (DSC) and thermo-gravimetric analysis (TGA)), water
  • XPRD X-ray powder diffraction
  • NMR nuclear magnetic resonance
  • FT-IR Fourier transform infrared spectroscopy
  • optical assessment e.g., particle shape/size
  • thermal properties e.g., differential scanning calorimetry (DSC) and thermo-gravimetric analysis (TGA)
  • X-Ray Powder Diffraction patterns were collected on a Bruker AXS C2 GADDS diffractometer using Cu K radiation (40 kV, 40 mA), automated XYZ stage, laser video microscope for auto-sample positioning and a HiStar 2-dimensional area detector.
  • X-ray optics consists of a single Gobel multilayer mirror coupled with a pinhole collimator of 0.3 mm. The beam divergence, i.e. the effective size of the X-ray beam on the sample, was approximately 4 mm.
  • a ⁇ - ⁇ continuous scan mode was employed with a sample - detector distance of 20 cm which gives an effective 2 ⁇ range of 3.2 ° - 29.7 °. Typically the sample was exposed to the X-ray beam for 120 seconds.
  • the software used for data collection was GADDS for WNT 4.1.16 and the data were analyzed and presented using Diffrac Plus EVA v 9.0.0.2 or v 13.0.0.2.
  • NMR Characterization NMR spectra were collected on a Bruker 400 MHz instrument equipped with an auto-sampler and controlled by a DRX400 console. Automated experiments were acquired using ICON MR v4.0.4 (build 1) running with Topspin v 1.3 (patch level 8) using the standard Bruker loaded experiments. For non-routine spectroscopy, data were acquired through the use of Topspin alone. Samples were prepared in d6-OMSO, unless otherwise stated. Off-line analysis was carried out using ACD SpecManager v 9.09 (build 7703).
  • Figure 12 shows the 3 ⁇ 4 NMR spectrum for batch 1.
  • 3 ⁇ 4 NMR spectra of batches 2- 6 were also recorded and compared with the spectrum of batch 1. See Figure 13.
  • the spectra are all similar, but with varying amounts of water. Some differences are noted in the integration of the large group of protons between 0.75 ppm and 2 ppm, where peaks overlap and cannot be integrated separately.
  • Table J shows the total number of protons integrated in the spectra of batches 1-6, taking into account the variation in the 0.75 - 2 ppm region.
  • the carboxylic acid proton has been excluded, so the number of protons should be 43, but it actually varies from 40 to 43 between the 6 spectra. However, the area where the variation comes from (0.75-2 ppm) is quite wide, and due to the quality of the baseline, this integration cannot be relied upon.
  • Figure 14 shows the DEPTQ spectrum, where CH2 and quaternary carbons peaks point up, while CH3 and CH groups point down. There are thirteen peaks pointing down, which correspond to nine CHs and four CH3 groups. This is consistent with the structure. The peak of the carbon of the carboxylic acid was seen at
  • DSC data were collected on a TA Instruments Q2000 equipped with a 50 position autosampler. The instrument was calibrated for energy and temperature calibration using certified indium. Typically 0.5-3 mg of each sample, in a pin-holed aluminum pan, was heated at 10 ⁇ .min "1 from 25 °C to 300 °C. A nitrogen purge at 50 ml.min "1 was maintained over the sample.
  • the instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3 and the data were analyzed using Universal Analysis v4.3A.
  • modulated DSC the sample was prepared as before, and the pan was heated at 2 ⁇ .min "1 from 25 °C to 200 °C. Modulator conditions were an amplitude of 0.20 °C and a periodicity of 40 s. The sampling interval was 1 sec/pt.
  • TGA data were collected on a TA Instruments Q500 TGA, equipped with a 16 position autosampler.
  • the instrument was temperature calibrated using certified Alumel. Typically 5-10 mg of each sample was loaded onto a pre-tared platinum crucible and aluminum DSC pan, and was heated at 10 ⁇ .min "1 from ambient temperature to 350 °C. A nitrogen purge at 60 ml.min "1 was maintained over the sample.
  • the instrument control software was Advantage for Q Series v2.8.0.392 and Thermal Advantage v4.8.3.
  • Thermal analysis of batch 1 was performed by DSC and TGA.
  • the TGA trace shows a weight loss of 1.7% between ambient temperature and 121 °C, which is likely to be loss of water.
  • the DSC trace shows a broad low temperature endotherm, probably corresponding to the loss of water, followed by a small endotherm with onset at 94 °C.
  • This second endotherm might indicate a glass transition and was further investigated by modulated DSC (see Figure 20).
  • modulated DSC modulated DSC
  • This technique enables reversible events, such as a glass transition, to be separated from irreversible ones, such as loss of solvent or a melt of a crystalline form.
  • the reversible heat flow trace in modulated DSC shows the glass transition as a step with an inflexion point (Tg) at 95 °C. This is high for a glass transition and suggests that Form 1 is stable.
  • Tg inflexion point
  • the small endotherm with onset at 89 °C on the non-reversible heat flow trace corresponds to molecular relaxation of the bulk material at the glass transition temperature.
  • the DSC trace shows decomposition starting around 220 °C, which also corresponds to the TGA trace curving down.
  • Figure 22 shows the DSC traces of the six batches for comparison.
  • the traces are similar, with a broad low temperature endotherm of varying size, consistent with varying amounts of water, followed by a small endotherm around the glass transition temperature as seen in section DSC and TGA.
  • Table L The results are summarized in Table L.
  • Table L Summary of DSC results of received samples Batch 1 st endotherm, broad 2 nd endotherm, small Start of number decomposition
  • Samples were studied on a Leica LM/DM polarized light microscope with a digital video camera for image capture. A small amount of each sample was placed on a glass slide, mounted in silicone oil and covered with a glass slip, the individual particles being separated as well as possible. The sample was viewed with appropriate magnification and partially polarized light, coupled to a ⁇ false-color filter.
  • FIGS 23A-23F show that batches 1, 2, 3, 4, 5, and 6 are material made up of large hard agglomerates of small irregular particles. Batches 1, 2, 3, 4, 5, and 6 all look similar. No birefringence was observed under plane polarized light, which is consistent with the material being non-crystalline. Particle size ranges from less than ⁇ to 3 ⁇ . The small size of these particles suggests that they have been precipitated out very quickly. Gravimetric Vapor Sorption (GVS)
  • Sorption isotherms were obtained using a SMS DVS Intrinsic moisture sorption analyzer, controlled by SMS Analysis Suite software.
  • the sample temperature was maintained at 25 °C by the instrument controls.
  • the humidity was controlled by mixing streams of dry and wet nitrogen, with a total flow rate of 200 ml.min "1 .
  • the relative humidity was measured by a calibrated Rotronic probe (dynamic range of 1.0-100 % RH), located near the sample.
  • the weight change, (mass relaxation) of the sample as a function of % RH was constantly monitored by the microbalance (accuracy ⁇ 0.005 mg).
  • sample typically 5-20 mg was placed in a tared mesh stainless steel basket under ambient conditions. The sample was loaded and unloaded at 40% RH and 25 °C (typical room conditions). A moisture sorption isotherm was performed as outlined below (2 scans giving 1 complete cycle). The standard isotherm was performed at 25 °C at 10% RH intervals over a 0.5-90 % RH range.
  • the water content of each sample was measured on a Mettler Toledo DL39 Coulometer using Hydranal Coulomat AG reagent and an argon purge. Weighed solid samples were introduced into the vessel on a platinum TGA pan, which was connected to a subaseal to avoid water ingress. Approximately 10 mg of sample was used per titration and duplicate determinations were made.
  • pKa determination data were collected on a Sirius GlpKa instrument with a D- PAS attachment. Measurements were made at 25 °C in aqueous solution by UV and in methanol water mixtures by potentiometry. The titration media was ionic-strength adjusted (ISA) with 0.15 M KC1 (aq). The values found in the methanol water mixtures were corrected to 0 % co-solvent via a Yasuda-Shedlovsky extrapolation. The data were refined using Refinement Pro software vl.O. Prediction of pKa values was made using ACD pKa prediction software v9.
  • the pKa of obeticholic acid was measured by potentiometry using methanol as a cosolvent (Figure 27) and extrapolated to 0% co-solvent using a Yasuda-Shedlovsky extrapolation (Figure 28).
  • the pKa enables determination of the proportion of the neutral and the ionized form of the compound at a given pH.
  • Figure 29 shows the distribution of the species depending on pH.
  • LogP was predicted using ACD software then measured by potentiometry. Three titrations were performed at three different octanol / ISA water ratios, giving the difference curve plotted in Figure 30.
  • the black curve is the pure aqueous pKa titration and the 3 colored curves correspond to the three octanol / ISA water ratios.
  • the shifts in pKa enable determination of LogP.
  • the lipophilicity curve (logD as a function of pH) is shown in Figure 31.
  • Log D is the distribution coefficient, representing the combined lipophilicity of all species present at a specific pH.
  • LogP is a compound constant, which corresponds to the partition coefficient of the pure neutral species, while LogPion is that of the pure ionized species.
  • LogP and LogPion can be determined from the lipophilicity curve, as the intersection of the Y axis with respectively the tangent at the start of the pH scale (when the molecule is purely in its neutral form) and the tangent at the end of the pH scale (when the molecule is completely ionized).
  • a sample of batch 1 was stored at 40°C and 75% relative humidity (RH) in an accelerated stability testing of the solid form. Another sample was stored at 25°C and 97% relative humidity to check the effect of very high humidity. Both samples were reanalyzed by XRPD after five days and after two weeks. Both samples remained non- crystalline under the two storage conditions for up to two weeks, showing that Form 1 is stable to these conditions. See Figure 32 and Figure 33.
  • the six batches analyzed were all non-crystalline. The glass transition temperature was measured at 95°C with a modulated DSC experiment. The six batches appeared very similar with all analytical techniques used, the only difference between them being their water content, which varied from 1.9% to 2.8% by Karl Fischer titration. Thermal analysis showed the varying amount of water and indicated decomposition starting around 175-220°C. Measured pKa was 4.82 and LogP is 5.54. Microscopic evaluation showed large hard agglomerates of very small irregular particles.
  • the single crystal X-ray structure of obeticholic acid was determined from a crystal obtained from the recrystallization of obeticholic acid from an acetonitrile solution after cooling to 5°C at 0.1°C/min followed by maturation at RT/50°C 8 h cycles for 1 week (see Figure 34).
  • the structure is consistent with Form G and a simulated XRPD partem has been generated as a reference pattern for this material.
  • Form G can be prepared by cooling a solution of obeticholic acid in e.g., acetonitrile.
  • the structure is orthorhombic, space group P2i2i2i, and contains one molecule of obeticholic acid in the asymmetric unit.
  • Final Rl [ ⁇ >2 ⁇ ( ⁇ )] 3.22 %.
  • the crystal exhibited prism morphology of approximate dimensions 0.4 x 0.4 x 0.3mm.
  • the Flack parameter 1.01(13), confirming the assignation mentioned above.
  • the software used to assign the stereochemistry determines the chiral center (C8) as an R stereocenter, whereas ACD software (and the Cahn-Ingold-Prelog) assignment for (C8) is S.
  • the assignment of the trans ring junction for B/C ring system is absolutely defined from the crystal structure.
  • the structure of obeticholic acid contains one 5 membered ring and 3 six membered rings which are fused together.
  • Conformational analysis on the 5 membered ring (CI 3, CI 4, CI 5, CI 6 and CI 7)) reveals that the closest puckering descriptor for this ring is a half-chair.
  • Conformational analysis on the three 6 membered rings (CI, C2, C3, C4, C5 and CIO); (C5, C6, C7, C8, C9 and CIO) and (C 8, C9, Cl l, C12 C13 and C14) reveals that the closest puckering descriptor for these rings is a chair.
  • the physical state of a solid obeticholic acid can play a role in the bioavailability of the molecule when administered orally to a subject (e.g., rats).
  • a subject e.g., rats.
  • the study described below was carried out to evaluate the plasma kinetics after a single oral administration and the efficiency of the intestinal absorption and the pharmacokinetics of solid noncrystalline and crystalline forms of obeticholic acid.
  • the profiles of obeticholic acid plasma concentration vs time, the tmax, Cmax and AUC after administration of obeticholic acid Form 1 (non-crystalline) or Form F were compared (see Figures 37-38)
  • Obeticholic acid Form 1 non-crystalline
  • Form F Obeticholic acid Form 1 (non-crystalline) and Form F were administered to rats and in each animal blood was collected at different period of times for at least 3 hours. Six animals were studied for each form of obeticholic acid.
  • the test substance used was obeticholic acid Form 1 (non-crystalline) and crystalline Form F.
  • Form F can be prepared by maturation from acetonitrile or nitromethane.
  • the formulation was prepared as a suspension in water at pH 4.
  • the study model is adult male Sprague Dawley rats about 225 to about 250 g (Harlan Laboratories).
  • Six animals were used per dosage route. The dosage is PO 20 mg/kg/5 mL.
  • the animals were fasted overnight before treatment with the formulation of obeticholic acid.
  • Oral administration was performed by gastric gavage.
  • the Cmax After administration of the crystalline form the Cmax is achieved after 1.5 hours and the plasma obeticholic acid concentration follows a regular kinetics with one maximum value and after 3 hours the dose is almost half of the Cmax.
  • the kinetics profile after the administration of obeticholic acid Form 1 (noncrystalline) Form 1 is different from that of the crystalline Form F.
  • An early plasma concentration peak is obtained after 30 minutes and a second one after 2 hours.
  • the variability of the data in the 6 rats is very low and this behaviour is statistically different from that of the crystalline form.
  • the AUC for the three hours studied is higher for the crystalline form.
  • the kinetics suggest that the obeticholic acid is still present in plasma after 3 hours. It has previously been demonstrated that the passage of obeticholic acid through the liver produce the hepatic metabolite tauro conjugate, which is secreted into bile and accumulate in the enterohepatic circulation.
  • the measurement of the tauro conjugate can be used to determine the passage of the amount of obeticholic acid through the liver.
  • the rate of tauro conjugate formation is reported in Figure 38, which shows that the tauro conjugate formation is faster and a higher concentration is achieved after administration of the crystalline form.
  • the melting point of obeticholic acid Form 1 (non-crystalline) Form 1 and crystalline Form F were measured using a conventional method.
  • the melting point of Chenodeoxycholic acid and Ursodeoxycholic acid were measured as reference compounds. Measurements have been performed in triplicate.
  • Tm melting temperature
  • T glass temperature transition
  • Table 3 Melting points of obeticholic acid (Form 1 and Form F) and CDCA and UDCA
  • DSC Differential scanning calorimetry
  • Figure 39 shows the DSC curve obtained for obeticholic acid crystalline Form F.
  • Form 1 present a higher solubility 17.9 ⁇ /L vs. 9.1 ⁇ /L for Form F.
  • crystalline Form F is higher than the obeticholic acid Form 1 (non-crystalline).
  • the plasma profiles show that the Form F is absorbed more efficiently (higher AUC) and even the kinetics is more regular, reflecting an optimal distribution of the drug in the intestinal content.
  • Form 1 shows this early peak then a later second one with a Cmax lower that than of Form F.
  • the water solubility of the Form 1 is higher than that of Form F.
  • Form F appears to be stable as the thermo gravimetric analysis (TGA) did not show any weight loss in the temperature range studied.
  • Radiolabeled obeticholic acid was prepared according to the scheme below.
  • [Ethyl-l- 14 C]obeticholic acid has a molecular formula of 14 CiC25H4404 and a molecular weight of 421.46 at the specific activity of 29 mCi/mmol by LSC.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C. 2 volume (60 ⁇ ) portions of acetonitrile were added at 50 °C. Initially the sample formed a gum, which dissolved after the addition of 14 volumes of solvent. On stirring flakes of solid precipitate were observed. The sample was cooled to 5 °C using a linear cooling rate of 0.25 °C/min. The resulting suspension was analyzed by XRPD and was then matured (shaken in 8 hour cycles at 50 °C/RT). After 5 days of maturation the sample was equilibrated to room temperature and was re-analyzed by XRPD.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into glass vials and warmed to 50 °C. Solvent was added (as per Table 5 and observations were made. Samples were stirred at 50 °C for 1 hour then were cooled to 5 °C using a linear cooling rate of 0.1 °C/min. Samples were stirred at 5 °C for 2 days. Any solids were filtered, air dried and analyzed by XRPD. Solutions were stored in the freezer for -48 hours, and then were left to evaporate at ambient conditions. Solids were analyzed by XRPD.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into glass vials and warmed to 50 °C. Solvent was added (as per Table 5) to dissolve the amorphous material and then anti-solvent (heptane, volumes as per Table 5) was added and observations were made. Samples were stirred at 50 °C for 1 hour then were cooled to 5 °C using a linear cooling rate of 0.1 °C/min. Samples were stirred at 5 °C for 2 days. Solids were filtered, air dried and analyzed by XRPD. Procedure 4: Maturation
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into glass vials and warmed to 50 °C. Solvent was added (as per Table 5) to dissolve the amorphous material. Samples were stirred at 50 °C for 1 hour then were matured (shaken in 8 hour cycles at 50 °C/RT). After 3 days samples containing solids were filtered, air dried and analyzed by XRPD. Samples that were solutions were matured with a cooler upper temperature (stirred in 4 hour cycles at 40 °C/RT, Procedure 4b) and any that formed solids were analyzed by XRPD after 1 and 2 days.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C.
  • MTBE (90 ⁇ , 3 volumes) was added to dissolve the amorphous material and then anti-solvent (heptane, 90 ⁇ , 3 volumes) was added and observations were made.
  • the sample was stirred at 50 °C for 1 hour then was cooled to 25 °C using a linear cooling rate of 0.2 °C/min. After ⁇ 30 minutes at 25 °C the resulting solid was filtered, air dried and analyzed by XRPD.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C. Anisole (90 ⁇ , 3 volumes) was added to dissolve the amorphous material. The sample was matured (shaken in 4 hour cycles at 40 °C/RT) for a total of 17 days.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and cooled to 5 °C. Acetonitrile/water mixtures (as per, pre-cooled to 5 °C) were added and samples were stirred at 5 °C overnight. Observations were made and solids were filtered, air dried and analyzed by XRPD. Some solids were large lumps which were crushed prior to XRPD analysis. One sample was a sticky solid so was left to stir at 5 °C for a total of 17 days, forming a hard lump of solid, which was crushed and analyzed by XRPD.
  • Amorphous obeticholic acid (approximately 30 mg) was weighed into glass vials and warmed to 50 °C. Solvent (see Table 5) was added to dissolve the amorphous material the samples were stirred at 50 °C for 1 hour. The samples were cooled to -20°C using a linear cooling rate of 0.1 °C/min. After stirring at -20 °C overnight the samples were equilibrated to room temperature and resulting solids were filtered, air dried and analyzed by XRPD.
  • amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C. Methyl isobutyl ketone (90 ⁇ ,) was added and observations were made. The sample was stirred at 50 °C for 1 hour then was cooled to 5 °C using a linear cooling rate of 0.1 °C/min. The sample was stirred at 5 °C for 2 days. Any solids were filtered, air dried and analyzed by XRPD. Solutions were stored in the freezer for -48 hours, and then were left to evaporate at ambient conditions. Solids were analyzed by XRPD and the peaks are provided in Table 6.
  • amorphous obeticholic acid (approximately 1 g) was weighed into a large glass vial and warmed to 50 °C. MIBK (2.0 ml, 2 volumes) was added at 50 °C, forming a clear solution on stirring. The sample was stirred at 50 °C for 2 hours then was cooled to -20 °C using a linear cooling rate of 0.1 °C/min. The sample was stirred at -20 °C overnight, after which a small amount of the solid was filtered, air dried and analyzed by XRPD. The remainder of the sample was filtered through a 0.45 ⁇ PTFE filter. The sample was air dried for approximately 30 minutes then was dried in a vacuum oven overnight at RT.
  • Figure 45 is an XRPD diffractogram of crystalline obeticholic acid Form D.
  • Figure 46 is an overlay of TGA and DSC thermograms of crystalline obeticholic acid Form D.
  • Figure 47 is an isotherm plot for the GVS experiment with crystalline obeticholic acid Form D.
  • Figure 48 is a kinetics plot for the GVS experiment with crystalline obeticholic acid Form D.
  • amorphous obeticholic acid (approximately 30 mg) was weighed into glass vials and warmed to 50 °C. Nitromethane was added to dissolve the amorphous material. Samples were stirred at 50 °C for 1 hour then were matured (shaken in 8 hour cycles at 50 °C/RT). After 3 days samples containing solids were filtered, air dried and analyzed by XRPD. Solids were analyzed by XRPD and the peaks are provided in Table 7.
  • obeticholic acid (approximately 1 g) was weighed into a large glass vial and warmed to 50 °C. Nitromethane (15.0 ml, 15 volumes) was added at 50 °C, resulting in the sample forming balls of gummy solid. The sample was matured (shaken in 8 hour cycles at 50 °C/RT) overnight after which the sample was a suspension of large hard lumps of solid and needle shaped particles. Some of the solid was crushed into a white powder and was analyzed by XRPD. The lumps of solid were broken up manually and the sample was matured for a further 24 hours. A small amount of the resulting suspension was filtered, air dried and analyzed by XRPD.
  • FIG. 49 is an XRPD diffractogram of crystalline obeticholic acid Form F.
  • Figure 50 is an overlay of TGA and DSC thermograms of crystalline obeticholic acid Form F.
  • Figure 51 is an isotherm plot for the GVS experiment with Form F.
  • Figure 52 is a kinetics plot for the GVS experiment with Form F.
  • amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and warmed to 50 °C. 2 volume (60 ⁇ ) portions of acetonitrile were added at 50 °C. Initially the sample formed a gum, which dissolved after the addition of 14 volumes of solvent. On stirring flakes of solid precipitate were observed. The sample was cooled to 5 °C using a linear cooling rate of 0.25 °C/min. The resulting suspension was analyzed by XRPD and was then matured (shaken in 8 hour cycles at 50 °C/RT). After 5 days of maturation the sample was equilibrated to room temperature and was reanalyzed by XRPD and the peaks are provided in Table 8.
  • Amorphous obeticholic acid (approximately 100 mg) was weighed into glass vials and warmed to 50 °C. Ten 100 mg scale experiments were carried out in parallel.
  • Acetonitrile (0.5 ml, 5 volumes, pre-warmed to 50 °C) was added to each sample forming a colorless gum which mostly dissolved on standing at 50 °C.
  • Each sample was seeded with ⁇ 1 mg of the product in the small scale experiment and the samples were matured (shaken in 8 hour cycles at 50 °C/RT) for 3 days. After maturation the samples contained crystals on the walls of the vial and a hard solid on the base of the vial which formed a white powder on scratching.
  • Each sample was analyzed by XRPD and the samples were combined by filtering them all through a 0.45 ⁇ PTFE filter. The sample was air dried for approximately 30 minutes then was dried in a vacuum oven overnight at RT.
  • Figure 53 is an XRPD diffractogram of crystalline obeticholic acid Form G.
  • Figure 54 is an overlay of TGA and DSC thermograms of crystalline obeticholic acid Form G.
  • Figure 55 is an isotherm plot for the GVS experiment with Form G.
  • Figure 56 is a kinetics plot for the GVS experiment with Form G.
  • amorphous obeticholic acid (approximately 30 mg) was weighed into a glass vial and cooled to 5 °C. Acetonitrile/water mixtures (50/50, 60 uL) were added and samples were stirred at 5 °C overnight. Observations were made and solids were filtered, air dried and analyzed by XRPD. Some solids were large lumps which were crushed prior to XRPD analysis. One sample was a sticky solid so was left to stir at 5 °C for a total of 17 days, forming a hard lump of solid, which was crushed and analyzed by XRPD and the peaks are provided in Table 9.
  • Amorphous obeticholic acid (approximately 1 g) was weighed into a large glass vial and cooled to 5 °C.
  • Acetonitrile/water mixture (2 ml, 2 volumes of a 50/50 v/v pre- mixed mixture, pre-cooled to 5 °C) was added and the sample formed a large lump of gummy off-white solid.
  • the sample was stirred at 5 °C overnight after which it consisted of a large lump of hard white solid.
  • the solid lump was broken up into a white powder and a small amount was filtered, air dried and analyzed by XRPD.
  • FIG. 57 is an XRPD diffractogram of crystalline obeticholic acid Form I.
  • Figure 58 is an overlay of TGA and DSC thermograms of crystalline obeticholic acid Form I.
  • Figure 59 is an isotherm plot for the GVS experiment with Form I.
  • Figure 60 is a kinetics plot for the GVS experiment with Form I.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Vascular Medicine (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Steroid Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des formes cristallines A, D, F, G et I de l'acide obéticholique. Ces formes cristallines sont utiles dans la production d'acide obéticholique (en particulier dans sa purification), qui est un médicament utile pour le traitement ou la prévention d'une maladie ou d'une affection, d'une maladie cardiovasculaire ou une maladie hépatique cholestatique médiées par le FXR et pour la réduction du cholestérol HDL, pour la diminution des triglycérides chez un mammifère ou pour l'inhibition de la fibrose.
EP16714049.0A 2015-12-22 2016-01-08 Formes cristallines polymorphes de l'acide obéticholique Withdrawn EP3394081A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14/979,005 US9982008B2 (en) 2012-06-19 2015-12-22 Preparation and uses of obeticholic acid
PCT/US2016/012651 WO2017111979A1 (fr) 2015-12-22 2016-01-08 Formes cristallines polymorphes de l'acide obéticholique

Publications (1)

Publication Number Publication Date
EP3394081A1 true EP3394081A1 (fr) 2018-10-31

Family

ID=55650649

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16714049.0A Withdrawn EP3394081A1 (fr) 2015-12-22 2016-01-08 Formes cristallines polymorphes de l'acide obéticholique

Country Status (15)

Country Link
EP (1) EP3394081A1 (fr)
JP (1) JP2018538331A (fr)
KR (1) KR20180095070A (fr)
CN (1) CN108495858A (fr)
AU (1) AU2016375566A1 (fr)
BR (1) BR112018012590A2 (fr)
CA (1) CA3009149A1 (fr)
CL (1) CL2018001720A1 (fr)
CO (1) CO2018006701A2 (fr)
EA (1) EA201891491A1 (fr)
IL (1) IL259998A (fr)
MX (1) MX2018007776A (fr)
PH (1) PH12018501318A1 (fr)
SG (1) SG11201805235XA (fr)
WO (1) WO2017111979A1 (fr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105801653B (zh) * 2014-12-30 2018-04-17 苏州晶云药物科技有限公司 奥贝胆酸的晶型a及其制备方法
WO2018211413A1 (fr) * 2017-05-15 2018-11-22 Dr. Reddy’S Laboratories Limited Formes solides d'acide obéticholique et procédé de préparation associé
CN109280071A (zh) * 2017-07-19 2019-01-29 东莞东阳光药物研发有限公司 奥贝胆酸的晶型及其制备方法
CN107383139A (zh) * 2017-08-09 2017-11-24 杭州和泽医药科技有限公司 一种3α‑羟基‑7‑氧代‑5β‑胆烷酸新衍生物制备奥贝胆酸的方法
CZ31099U1 (cs) * 2017-09-05 2017-10-17 Zentiva, K.S. Krystalické formy (3a,5B,6a,7a)-6-ethyl-3,7- dihydroxycholan-24-ové kyseliny
CN109485687A (zh) * 2017-09-12 2019-03-19 成都弘达药业有限公司 奥贝胆酸的晶型j及其制备方法
WO2019106043A1 (fr) 2017-11-29 2019-06-06 Hexal Ag Composition pharmaceutique comprenant de l'acide obéticholique
GB201812382D0 (en) 2018-07-30 2018-09-12 Nzp Uk Ltd Compounds
CN113264972A (zh) * 2020-02-14 2021-08-17 四川科伦药物研究院有限公司 一种制备奥贝胆酸的方法

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITMI20050912A1 (it) * 2005-05-19 2006-11-20 Erregierre Spa Processo di preparazione di acidi 3-a-ya(b)-diidrossi-6-a(b)-alchil-5b-colanici
DK3336097T3 (da) * 2012-06-19 2020-09-28 Intercept Pharmaceuticals Inc Fremstilling af ikke-krystallinsk obeticholsyre
WO2016045480A1 (fr) * 2014-09-28 2016-03-31 上海源力生物技术有限公司 Procédé de préparation d'acide obéticholique
WO2016079520A1 (fr) * 2014-11-19 2016-05-26 Dextra Laboratories Limited Stéroïdes 6.alpha.-alkyl-6,7-dione utilisés en tant qu'intermédiaires pour la production de modulateurs de fxr stéroïdiens
EA033445B1 (ru) * 2014-11-19 2019-10-31 Nzp Uk Ltd 5-бета-6-алкил-7-гидрокси-3-оновые стероиды в качестве промежуточных соединений для получения стероидных модуляторов fxr
CA2968305A1 (fr) * 2014-11-19 2016-05-26 NZP UK Limited Steroides 6.alpha.-alkyl-3,7-dione utilises en tant qu'intermediaires pour la production de modulateurs de fxr steroidiens
DK3221331T3 (da) * 2014-11-19 2019-12-02 Nzp Uk Ltd 6-alkyl-7-hydroxy-4-en-3-on-steroider som mellemprodukter til fremstilling af steroide fxr-modulatorer

Also Published As

Publication number Publication date
CL2018001720A1 (es) 2018-08-10
AU2016375566A1 (en) 2018-07-05
CN108495858A (zh) 2018-09-04
EA201891491A1 (ru) 2018-11-30
KR20180095070A (ko) 2018-08-24
BR112018012590A2 (pt) 2018-12-04
WO2017111979A1 (fr) 2017-06-29
MX2018007776A (es) 2018-08-09
PH12018501318A1 (en) 2019-02-18
SG11201805235XA (en) 2018-07-30
CO2018006701A2 (es) 2018-07-10
JP2018538331A (ja) 2018-12-27
CA3009149A1 (fr) 2017-06-29
IL259998A (en) 2018-07-31

Similar Documents

Publication Publication Date Title
AU2018241172B2 (en) Preparation, uses and solid forms of obeticholic acid
EP3394081A1 (fr) Formes cristallines polymorphes de l'acide obéticholique
US9982008B2 (en) Preparation and uses of obeticholic acid

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180718

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RAV Requested validation state of the european patent: fee paid

Extension state: MD

Effective date: 20180718

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20190221