WO2021044351A1 - Methods of treating liver disease using lta4h inhibitors - Google Patents

Methods of treating liver disease using lta4h inhibitors Download PDF

Info

Publication number
WO2021044351A1
WO2021044351A1 PCT/IB2020/058229 IB2020058229W WO2021044351A1 WO 2021044351 A1 WO2021044351 A1 WO 2021044351A1 IB 2020058229 W IB2020058229 W IB 2020058229W WO 2021044351 A1 WO2021044351 A1 WO 2021044351A1
Authority
WO
WIPO (PCT)
Prior art keywords
liver
measured
patient
fibrosis
lta4h inhibitor
Prior art date
Application number
PCT/IB2020/058229
Other languages
French (fr)
Inventor
Ralf Glatthar
Linda GREENBAUM
Iwona KSIAZEK
Christian Loesche
Till Roehn
Andreas Sailer
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Publication of WO2021044351A1 publication Critical patent/WO2021044351A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/468-Azabicyclo [3.2.1] octane; Derivatives thereof, e.g. atropine, cocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients

Definitions

  • the present disclosure relates to methods for treating or preventing liver disease using leukotriene A4 hydrolase (LTA4H) inhibitors.
  • LTA4H leukotriene A4 hydrolase
  • Nonalcoholic fatty liver disease is the most common cause of chronic liver disease in the Western world (Ratziu et al., J Gastrointestin Liver Dis. 2010 19(4):415-23).
  • the main stages of NAFLD are 1- simple fatty liver (steatosis); 2- non-alcoholic steatohepatitis (NASH), a more serious form of NAFLD; 3- fibrosis, where there is a persistent inflammation in the liver resulting in the generation of fibrous scar tissue around the liver cells and blood vessels; and 4-cirrhosis; this damage is permanent and can lead to liver failure and liver cancer.
  • NASH includes fat accumulation in the liver, as well as inflammation which over time can lead to increased fibrosis, cirrhosis and end stage liver disease.
  • Liver transplantation is the only treatment for advanced cirrhosis with liver failure, and transplantation is increasingly performed in persons suffering from NASH.
  • NAFLD National Air Traffic Deficiency Virus
  • 33% the estimated prevalence of NASH is lower, ranging from 3 to 5% (Younossi et al., 2016, Hepatology, Vol. 64(5):1577-1586).
  • NASH is a worldwide problem with growing prevalence over the last few decades. Over the last decade, NASH has risen from uncommon to the second indication for liver transplantation in the US. It is expected to be the leading cause of transplant by 2020 (Wong, et al, Gastroenterology, 2015, 148(3):547-55).
  • NASH is highly associated with the metabolic syndrome and Type 2 diabetes mellitus. NASH is a cause of progressive fibrosis and of cirrhosis. Cirrhosis due to NASH increases the risk of hepatocellular carcinoma and hepatocellular cancer. Furthermore, cardiovascular mortality is an important cause of death in NASH patients.
  • Chronic cholestasis and liver inflammation are the two main pathophysiological components of the two major classes of disease - primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) - leading to bile duct destruction and ultimately to cirrhosis and liver failure. Liver transplantation appears to be the only life-saving procedure.
  • PBC primary biliary cirrhosis
  • PSC primary sclerosing cholangitis
  • Ursodeoxycholic acid also known as ursodiol
  • UDCA Ursodeoxycholic acid
  • PBC primary biliary cholangitis
  • OCA Obeticholic acid
  • FXR Fem X receptor
  • obeticholic acid When tested in nonalcoholic steatohepatitis patients, obeticholic acid showed efficacy, in particular a significant improvement in NASH, i.e. strong impact on steatosis with additional effects on inflammation and ballooning.
  • OCA long term administration raises safety concerns because it can be associated with pruritus, as well as with increased LDL cholesterol (see “Intercept Announces New FLINT Trial Data Showing OCA Treatment Increases Fibrosis Resolution and Cirrhosis Prevention in High-Risk NASH Patients”, April 23, 2015).
  • concomitant administration of statins may be required for long term treatment of NASH patients.
  • liver diseases such as NAFLD, NASH or PBC, and for late stage liver diseases.
  • a medicament would be particularly efficient if it has an impact on one or more of the different aspects of the liver disease. Therefore, there is a need to provide treatments for fibrotic / cirrhotic diseases or disorders, e.g. liver diseases or disorders, that can address the different aspects of these complex conditions, while demonstrating an acceptable safety and/or tolerability profile and ultimately avoiding the need for liver transplant.
  • the combination of two or more molecules with different Mechanisms of Action (MoA) might provide additional benefits for improving treatment efficacy and response rates.
  • one approach to the treatment of NASH is administering to a patient in need thereof, a compound with anti-steatotic properties, anti-inflammatory properties and/or anti-fibrotic properties; or a compound with mixed effects.
  • the compounds of the invention i.e. leukotriene A4 hydrolase (LTA4H) inhibitors such as compounds of Formula (I), or pharmaceutically acceptable salt thereof, inhibit LTA4H and thereby prevent the biosynthesis of pro-inflammatory leukotriene B4 (LTB4).
  • LTA4H leukotriene A4 hydrolase
  • LTB4 is responsible for driving the three main processes in the pathophysiology of NASH: Inflammation, insulin resistance and white adipose tissue (WAT) generation and lipolysis in adipocytes (Oh et al. Nat Rev Drug Disc; 2016, 15:161-172).
  • LTB4 Leukotriene B4
  • Adipocyte derived LTB4 has also been shown to contribute to “low-grade” inflammatory state leading to obesity associated co-morbidities (Mothe-Satney et al. Diabetes 2012, 61 (9):2311-9).
  • LTA4H inhibitors of the present invention reduce liver inflammation and damage including reduction in alanine aminotransferase (ALT) biomarker, fibrotic gene expression, hepatic neutrophils, macrophages and macrophage crown like structures.
  • ALT alanine aminotransferase
  • LTA4H of the present invention also reduces TIMP-1 and PIII-NP serum fibrosis biomarkers.
  • LTA4H inhibition can shift production of pro-inflammatory lipid mediator LTB4 to anti-inflammatory lipid mediator Lipoxin A4 (LXA4).
  • LXA4 has been shown to attenuate obesity induced liver inflammation, injury & fibrosis (Borgeson et al. Cell Metab; 2015 22(10): 125-37). LXA4 is also known to protect against diet induced obesity and insulin resistance, increases energy expenditure and browning of fat. (Elias et al.; Diabetes 2016, 65(8) :2139-50).
  • the aim of the present invention is to provide novel methods for treating or preventing liver disease in a subject in need thereof, comprising administering to said subject, a therapeutically effective amount of a leukotriene A4 hydrolase (LTA4H) inhibitor. More particularly, the invention pertains to a method of treating or preventing a chronic liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of Formula (I): wherein,
  • R 1 is OH or NH 2 ;
  • Y is O, S or CH 2 ;
  • X 1 , X 2 , X 3 and X 4 are N; or
  • X 1 , X 2 , X 3 and X 4 are selected from N, NH, C, CH and O with the proviso that at least two of X 1 , X 2 , X 3 and X 4 are N or NH;
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; C 3 -C 6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C 1 -C 6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable salt thereof.
  • the invention further provides a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor with one or more therapeutic agents, such as anti- steatotic agents, anti-inflammatory agents or anti-fibrotic agents.
  • a LTA4H inhibitor with one or more therapeutic agents, such as anti- steatotic agents, anti-inflammatory agents or anti-fibrotic agents.
  • the present invention provides a LTA4H inhibitor for use in the treatment and/or prevention of a liver disease, in a patient in need of such treatment and/or prevention. More particularly, the invention pertains to a compound of Formula (I); or a pharmaceutically acceptable salt thereof, as described herein for the use in the treatment or prevention of a chronic liver disease, in a patient in need of such treatment or prevention.
  • the invention further provides pharmaceutical combinations, comprising, separate or together, a LTA4H inhibitor and one or more additional therapeutic agents, for simultaneous, sequentially or separate administration.
  • a medicament comprising such combinations for use in the treatment or prevention of liver diseases.
  • the liver disease is selected from non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis, bile duct obstruction, cholelithiasis and liver fibrosis.
  • Figure 1 provides an illustrative XRPD spectrum for the crystalline form of (S)-3-amino-4-(5-(4- ((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form, designated herein as Form B, showing degrees 2q (2-theta) on the X-axis and relative intensity on the Y-axis.
  • Figure 2 provides an illustrative DSC spectrum for the free form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, designated herein as Form B.
  • Figure 3 provides an illustrative TGA spectrum for the free form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, designated herein as Form B.
  • Figure 4 shows normalized expression of the ALOX5 gene (5-Lipoxygenase enzyme) in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow.
  • Figure 5 shows normalized expression of the ALOX5AP gene (5-Lipoxygenase accessory protein, FLAP) in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow.
  • ALOX5AP gene (5-Lipoxygenase accessory protein, FLAP)
  • FIG. 6 shows normalized expression of the LTA4H gene (Leukotriene A4 hydrolase) versus control gene TBP in all liver biopsies.
  • Figure 7 Liver macrophages of liver biopsies of mice treated with two doses of (S)-3-amino-4- (5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid compared to vehicle treated mice and mice on normal chow by immunohistological analysis using the marker protein IBA1
  • FIG. 8 Liver neutrophils of liver biopsies of mice treated with two doses of (S)-3-amino-4-(5- (4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid compared to vehicle treated mice and mice on normal chow by immunohistological analysis using the marker protein LY-6b.
  • FIG. 9 Liver enzymes ALT and AST in blood of high fat, high fructose diet exposed mice treated with 3 mg/kg and 10 mg/kg of (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid versus vehicle control and mice on normal chow.
  • Figure 10 Inhibition of LTB4 release from ex-vivo stimulated blood in mice treated with 3mg/kg or 10mg/kg of (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid.
  • Figure 11 Mean plasma concentration time profiles following single oral administration of (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different doses.
  • Figure 12 Mean plasma concentration time profile following multiple oral administration of (S)- 3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different doses (Figure 12a: Day 1 ; Figure 12b: Day 12)
  • Figure 13 LTB4 concentration in ex-vivo stimulated blood after oral administration of (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at measured plasma concentration of compound of example 1 in plasma (PK/PD relationship)
  • FIG 14 LTB4 change from baseline (inhibition in blood) after multiple oral administration of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different dose measured at different time (days) since first dose.
  • measured values from XRPD experiments and DSC/TGA experiments can vary as a result of, for example, sample preparation and/or storage and/or environmental conditions, and yet the measured values will still be considered to be representative of a particular solid state form of the crystalline materials described herein.
  • C 1 -C 6 alkyl refers to a fully saturated branched or unbranched hydrocarbon moiety having up to 6 carbon atoms. Unless otherwise provided, it refers to hydrocarbon moieties having 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms.
  • Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso- propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl and the like.
  • C 1 -C 6 alkoxy refers to alkyl-O-, wherein alkyl is defined herein above.
  • Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, hexyloxy, cyclopropyloxy-, cyclohexyloxy- and the like.
  • alkoxy groups have about 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms.
  • C 1 -C 6 alkyl optionally substituted by halogen refers to C 1 -C 6 alkyl as defined above which may be substituted by one or more halogens. Examples include, but are not limited to, trifluoromethyl, difluoro methyl, fluoromethyl, trichloromethyl, 2,2,2- trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl and 1-bromomethyl-2-bromoethyl.
  • di-C 1-6 alkylamino refers to a moiety of the formula -N(R a )-R a where each R a is a C 1-6 alkyl, which may be the same or different, as defined above.
  • C 3 -C 6 cycloalkyl refers to saturated monocyclic hydrocarbon groups of 3-6 carbon atoms. Cycloalkyl may also be referred to as a carbocyclic ring and vice versa additionally referring to the number of carbon atoms present. Unless otherwise provided, cycloalkyl refers to cyclic hydrocarbon groups having between 3 and 6 ring carbon atoms or between 3 and 4 ring carbon atoms. Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. As used herein, the term “halogen” or “halo” refers to fluoro, chloro, bromo, and iodo.
  • heteroaryl refers to a 5-14 membered monocyclic- or bicyclic- or tricyclic-aromatic ring system, having 1 to 8 heteroatoms.
  • the heteroaryl is a 5-10 membered ring system containing 1 to 4 hereroatoms selected from N, S or O ( e.g ., 5-7 membered monocycle or an 8-10 membered bicycle) or a 5-7 membered ring system.
  • heteroaryl is a 5-7 membered monocycle.
  • Typical heteroaryl groups include 2- or 3-thienyl, 2- or 3-furyl, 2- or 3-pyrrolyl, 2-, 4-, or 5-imidazolyl, 3-, 4-, or 5- pyrazolyl, 2-, 4-, or 5-thiazolyl, 3-, 4-, or 5-isothiazolyl, 2-, 4-, or 5-oxazolyl, 3-, 4-, or 5-isoxazolyl, 3- or 5- 1,2,4-triazolyl, 4- or 5-1 , 2, 3-triazolyl, tetrazolyl, 2-, 3-, or 4-pyridyl, 3- or 4-pyridazinyl, 3-, 4-, or 5-pyrazinyl, 2-pyrazinyl, and 2-, 4-, or 5-pyrimidinyl.
  • heteroaryl also refers to a group in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Non limiting examples include indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, quinolizinyl, quinoliyl, isoquinoliyl, benzisoqinolinyl, thieno[2,3-b]furanyl, furo[3,2-b]-pyranyl, pyrido[2,3-d]-o-oxazinyl, pyrazolo[4,3-d]-oxazolyl, imidazo[4,5-d] thiazolyl, 3pyrazino[2,3-d]pyridazinyl, imidazo[2,1-b] thiazolyl, benzo[b]thienyl, benzimidazolyl, and benzothiazolyl.
  • Typical fused heteroaryl groups include, but are not limited to quinolinyl, isoquinolinyl, indolyl, benzo[b]thienyl, benzoxazolyl, benzimidazolyl, and -benzothiazolyl.
  • a substituted heteroaryl is a heteroaryl group containing one or more substituents.
  • heterocyclyl refers to a heterocyclic group that is saturated or partially saturated and is preferably a monocyclic or a polycyclic ring (in case of a polycyclic ring particularly a bicyclic, tricyclic or spirocyclic ring); and has 3 to 24, more preferably 4 to 16, most preferably 5 to 10, and most preferably 5 or 6 ring atoms; wherein one or more, preferably one to four, especially one or two ring atoms are a heteroatom (the remaining ring atoms therefore being carbon).
  • the bonding ring i.e. the ring connecting to the molecule
  • heterocyclyl excludes heteroaryl.
  • the heterocyclic group can be attached at a heteroatom or a carbon atom.
  • the heterocyclyl can include fused or bridged rings as well as spirocyclic rings.
  • Examples of heterocycles include tetrahydrofuran (THF), dihydrofuran, 1,4-dioxane, morpholine, 1,4-dithiane, piperazine, piperidine, 1,3- dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane, dithiolane, 1,3-dioxane, 1 ,3-dithiane, oxathiane, thiomorpholine, and the like.
  • a substituted heterocyclyl is a heterocyclyl group independently substituted by 1-4, such as one, or two, or three, or four substituents.
  • aryl refers to an aromatic hydrocarbon group having 6-20 carbon atoms in the ring portion. Typically, aryl is monocyclic, bicyclic or tricyclic aryl having 6- 20 carbon atoms.
  • aryl refers to an aromatic substituent which can be a single aromatic ring, or multiple aromatic rings that are fused together. Non- limiting examples include phenyl, naphthyl or tetrahydronaphthyl.
  • a substituted aryl is an aryl group substituted by 1-5 (such as one, or two, or three) substituents independently selected from the group consisting of hydroxyl, thiol, cyano, nitro, C 1 -C 4 -alkyl, C 1 - C 4 -alkenyl, C 1 -C 4 -alkynyl, C 1 -C 4 -alkoxy, C 1 -C 4 -thioalkyl, C 1 -C 4 -alkenyloxy, C 1 -C 4 -alkynyloxy, halogen, C 1 -C 4 -alkylcarbonyl, carboxy, C 1 -C 4 -alkoxycarbonyl, amino, C 1 -C 4 -alkylamino, di- C 1 - C 4 -alkylamino, C 1 -C 4 -alkylaminocarbonyl, di- C 1 -C 4 -alkylaminocarbonyl,
  • the terms “about” and “substantially” indicate with respect to features such as endotherms, endothermic peak, exotherms, baseline shifts, etc., that their values can vary.
  • “about” or “substantially” means that typical peak position and intensity variability are taken into account.
  • the peak positions (2q) will show some inter-apparatus variability, typically as much as 0.2°. Occasionally, the variability could be higher than 0.2° depending on apparatus calibration differences.
  • phase pure when used in reference to any crystalline form of the compound of Formula I, means a compound having a phase purity of greater than about 90% by weight, including greater than about 91 , 92, 93, 94, 95, 96, 97, 98, and about 99% by weight, and also including equal to about 100% by weight of the compound of Formula (I), based on the weight of the compound on an anhydrous basis.
  • phase pure or phase purity herein refers to phase homogeneity with respect to a particular solid state form of the compound of Formula (I), and does not necessarily imply a high degree of chemical purity absent an express statement to that effect.
  • Phase purity may be determined according to methods known in the art, for example, using XRPD to do quantitative phase analysis using one or more approaches known in the art, for example, via an external standard method, direct comparisons of line (peak) characteristics which are attributed to different phases in a particular spectra, or via an internal standard method.
  • XRPD quantification of phase purity can be complicated by the presence of amorphous material. Accordingly, other methods that may be useful for determining phase purity include, for example, solid state NMR spectroscopy, Raman and/or infrared spectroscopy.
  • solid state NMR spectroscopy Raman and/or infrared spectroscopy.
  • One of skilled in the art would readily understand these methods and how to employ these additional (or alternative) methods for determining phase purity.
  • salt refers to an acid addition or base addition salt of a compound for use in the method of the invention.
  • Salts include, in particular, “pharmaceutically acceptable salts”.
  • pharmaceutically acceptable salts refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable.
  • the compounds for use in the methods of the invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups, or groups similar thereto.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts for use in the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods.
  • such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid.
  • a stoichiometric amount of the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds (i.e. LTA4H inhibitor as described herein) include isotopes of hydrogen, carbon, sulfur, nitrogen, oxygen, phosphorous, fluorine, iodine and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 CI, 125 l respectively.
  • the invention includes various isotopically labeled compounds as defined herein, for example, those into which radioactive isotopes, such as 3 H and 14 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • isotopically labeled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • SPECT single-photon emission computed tomography
  • SPECT single-photon emission computed tomography
  • Isotopically-labeled compounds of Formula (I), or a pharmaceutically acceptable salt thereof can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • solvates for use in the method of the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, d 6 -acetone, d 6 - DMSO (dimethylsulfoxide).
  • Compounds for use in the method of the invention i.e. compounds of Formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers.
  • These co-crystals may be prepared from compounds of Formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of Formula (I) with the co-crystal former under crystallization conditions and isolating co-crystals thereby formed.
  • Suitable co-crystal formers include those described in WO 2004/078163.
  • the invention further provides co-crystals comprising a compound of Formula (I) for use in the method of the present invention.
  • administering in relation to a compound, e.g., an LTA4H inhibitor (e.g. a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a specific compound described herein), or another agent, is used to refer to delivery of that compound to a patient by any administration route.
  • an LTA4H inhibitor e.g. a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a specific compound described herein
  • the term "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 20th Ed. Mack Printing Company, 1990, pp. 1289-1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
  • a therapeutically effective amount of a compound for use in the method of the invention refers to an amount of said compound that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms of liver disease, slow or delay disease progression of liver disease, or prevent liver disease.
  • the term “a therapeutically effective amount” refers to the amount of the compound that, when administered to a subject, is effective to (1) at least partially alleviating, inhibiting, preventing and/or ameliorating liver disease.
  • a therapeutically effective amount refers to the amount of the compound that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of LTA4H; or reducing or inhibiting the expression of LTA4H partially or completely.
  • the term “subject” refers to an animal. Typically, the animal is a mammal. A subject also refers to, for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. The term “subject” is used interchangeably with “patient” when it refers to human.
  • the phrase “population of patients” is used to mean a group of patients.
  • the LTA4H inhibitor e.g., compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a compound disclosed in WO2015/092740 or any compound described herein
  • the LTA4H inhibitor is used to treat a population of liver disease patients.
  • selecting and “selected” in reference to a patient is used to mean that a particular patient is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria.
  • selecting refers to providing treatment to a patient having a particular disease, where that patient is specifically chosen from a larger group of patients on the basis of the particular patient having a predetermined criterion.
  • selective administering refers to administering a drug to a patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criterion.
  • a patient is delivered a personalized therapy based on the patient’s personal history (e.g., prior therapeutic interventions, e.g., prior treatment with biologies), biology (e.g., particular genetic markers), and/or manifestation (e.g., not fulfilling particular diagnostic criteria), rather than being delivered a standard treatment regimen based solely on the patient’s membership in a larger group.
  • Selecting, in reference to a method of treatment as used herein, does not refer to fortuitous treatment of a patient having a particular criterion, but rather refers to the deliberate choice to administer treatment to a patient based on the patient having a particular criterion.
  • selective treatment/administration differs from standard treatment/administration, which delivers a particular drug to all patients having a particular disease, regardless of their personal history, manifestations of disease, and/or biology.
  • the patient is selected for treatment based on having a liver disease.
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment to ameliorating the disease or disorder (i.e. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms or pathological features thereof).
  • “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter or pathological features of the disease, e.g. including those which may not be discernible by the subject.
  • “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g. stabilization of at least one discernible or non-discernible symptom), physiologically (e.g. stabilization of a physical parameter) or both.
  • “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder, or of at least one symptoms or pathological features associated thereof. In yet another embodiment, “treat”, “treating” or “treatment” refers to preventing or delaying progression of the disease to a more advanced stage or a more serious condition, such as e.g. liver cirrhosis; or to preventing or delaying a need for liver transplantation.
  • treating NASH may refer to ameliorating, alleviating or modulating at least one of the symptoms or pathological features associated with NASH; e.g. hepatosteatosis, hepatocellular ballooning, hepatic inflammation and fibrosis; e.g. may refer to slowing progression, reducing or stopping at least one of the symptoms or pathological features associated with NASH, e.g. hepatosteatosis, hepatocellular ballooning, hepatic inflammation and fibrosis.
  • liver disease or disorder encompasses one, a plurality, or all of non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), primary biliary cirrhosis (PBC), bile duct obstruction, cholelithiasis and liver fibrosis.
  • NAFLD non-alcoholic fatty liver disease
  • NASH non-alcoholic steatohepatitis
  • CFLD cystic fibrosis-associated liver disease
  • PBC primary biliary cirrhosis
  • bile duct obstruction cholelithiasis and liver fibrosis.
  • NAFLD may encompass the different stages of the disease: hepatosteatosis, NASH, fibrosis and cirrhosis.
  • NASH may encompass steatosis, hepatocellular ballooning and lobular inflammation.
  • prevention refers delaying the onset or development or progression of the disease or disorder. More specifically, the term “preventing” the liver disease refers preventing or delaying liver cirrhosis or a need for liver transplantation.
  • a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • any asymmetric atom (e.g., carbon or the like) of the compound(s) for use in the method of the invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration.
  • each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R)- or (S)- configuration.
  • Substituents at atoms with unsaturated double bonds may, if possible, be present in cis- (Z)- or trans- (E)- form.
  • a compound for use in the method of the present invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof.
  • the term “possible isomers” shall not include positional isomers.
  • Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
  • any resulting racemates of final products or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound.
  • a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di- 0,0'-p- toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid.
  • Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral stationary phase.
  • the compounds for use in the method of the invention can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the compounds for use in the method of the invention may by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embraces the use of compounds as described herein, both solvated and unsolvated forms.
  • solvate refers to a molecular complex of a compound for use in the method of the invention (including pharmaceutically acceptable salts thereof) with one or more solvent molecules.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • hydrate refers to the complex where the solvent molecule is water.
  • the compounds for use in the method of the present invention include salts, hydrates, solvates and polymorph thereof.
  • “combination” refers to either a fixed combination in one unit dosage form (e.g., capsule, tablet, or sachet), free (i.e. non-fixed) combination, or a kit of parts for the combined administration where a LTA4H inhibitor of the present invention and one or more “combination partner” (i.e. the additional therapeutic agent, also referred to as or “co-agent”) may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the additional therapeutic agent to a single subject in need thereof (e.g. a patient), and the additional therapeutic agent are intended to include treatment regimens in which the LTA4H inhibitor and the additional therapeutic agent are not necessarily administered by the same route of administration and/or at the same time.
  • Each of the components of the combination of the present invention may be administered simultaneously or sequentially and in any order.
  • Co-administration comprises simultaneous, sequential, overlapping, interval, continuous administrations and any combination thereof.
  • pharmaceutical combination means a pharmaceutical composition that results from the combining (e.g. mixing) of more than one active ingredient and includes both fixed and free combinations of the active ingredients.
  • fixed combination means that the active ingredients, i.e. i) a LTA4H inhibitor, e.g. Compound of any one of Formulae (I) to (V) as defined herein, or a compound according to any one of the embodiments below (in free form or e.g. as a pharmaceutically acceptable salt) and ii) the additional therapeutic agent (as herein defined), are both administered to a patient simultaneously in the form of a single entity or dosage.
  • a LTA4H inhibitor e.g. Compound of any one of Formulae (I) to (V) as defined herein, or a compound according to any one of the embodiments below (in free form or e.g. as a pharmaceutically acceptable salt) and ii) the additional therapeutic agent (as herein defined)
  • free combination means that the active ingredients as herein defined are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, and in any order, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient.
  • free combination is sometimes also referred to as “non fixed combinations”.
  • spontaneous administration it is meant that the LTA4H inhibitor and the additional therapeutic agent (as herein defined), are administered on the same day.
  • the two active ingredients can be administered at the same time (e.g. for fixed or free combinations) or one at a time (e.g. for free combinations).
  • “sequential administration” may mean that during a period of two or more days of continuous co-administration only one of the LTA4H inhibitor and the additional therapeutic agent is administered on any given day.
  • overlapping administration it is meant that during a period of two or more days of continuous co-administration, there is at least one day of simultaneous administration and at least one day when only one of LTA4H inhibitor and the additional therapeutic agent, e.g., is administered.
  • interval administration it is meant a period of co-administration with at least one void day, i.e with at least one day where neither the LTA4H inhibitor nor the additional therapeutic agent is administered.
  • continuous administration it is meant a period of co-administration without any void day.
  • the continuous administration may be simultaneous, sequential, or overlapping, as described above.
  • the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor.
  • a liver disease e.g. NAFLD, NASH, liver fibrosis and PBC
  • the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor as described in WO2014/164658.
  • a liver disease e.g. NAFLD, NASH, liver fibrosis and PBC
  • the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor which is 4-(((1S,4S)-5-(4-(4-(oxazol-2-yl)phenoxy)benzyl)-2,5-diazabicyclo[2.2.1]heptan-2- yl)methyl)benzoic acid, or represented by:
  • the present invention relates to a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor which is Acebilustat, also known as CTX 4430.
  • a LTA4H inhibitor which is Acebilustat, also known as CTX 4430.
  • the present invention relates to a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof: wherein,
  • R 1 is OH or NH 2 ;
  • Y is O, S or CH 2 ;
  • X 1 , X 2 , X 3 and X 4 are N; or
  • X 1 , X 2 , X 3 and X 4 are selected from N, NH, C, CH and O with the proviso that at least two of X 1 , X 2 , X 3 and X 4 are N or NH;
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; C 3 -C 6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C 1 -C 6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable
  • Embodiment 2A relates to a method according to embodiment 2, comprising administering to the subject in need thereof, a therapeutically effective amount of a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein the variables R 1 , R2 and Y have the meaning as defined in embodiment 2.
  • Embodiment 2B relates to a method according to embodiment 2, comprising administering to the subject in need thereof, a therapeutically effective amount of a compound of formula (III) or a pharmaceutically acceptable salt thereof, wherein the variables R 1 , R2 and Y have the meaning as defined in embodiment 2.
  • Embodiment 2C relates to a method according to embodiment 2 comprising administering to the subject a therapeutically effective amount of a compound of formula (IV), or a pharmaceutically acceptable salt thereof,
  • Embodiment 2D relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of formula (V), or a pharmaceutically acceptable salt thereof; wherein the variables R 1 , R2 and Y have the meaning as defined in embodiment 2.
  • Embodiment 2E of the present invention relates to the method according to any one of embodiments 2 and 2A to 2D, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is O; and
  • R2 is phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or
  • R2 is a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C 1 -C 6 alkyl optionally substituted by halogen.
  • the present invention relates to the method according to embodiment 2E, comprising administering to the subject a therapeutically effective amount of a compound of Formula (V), or a pharmaceutically acceptable salt thereof, wherein
  • R 1 is H, Y is O and R2 is phenyl optionally substituted by one or two halogen.
  • Embodiment 2F of the present invention relates to the method according to any one of embodiments 2 and 2A to 2D, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is CH 2 ; and
  • R2 is phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or
  • R2 is a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by cyano, halogen or C 1 -C 6 alkyl optionally substituted by halogen.
  • Embodiment 2G of the present invention relates to a method according to any one of embodiments 2 and 2A to 2D, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein Y is O; and
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; or C 3 -C 6 cycloalkyl.
  • Embodiment 2H of the present invention relates to a method according to any one of embodiment 2 and 2A to 2D, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein Y is CH 2 ; and
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; or C 3 -C 6 cycloalkyl.
  • Embodiment 2I relates to a method according to any one of the embodiments 2 and 2A to 2H, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable salt thereof, wherein Y is attached to the para-position of the phenyl moiety.
  • Embodiment 2J relates to a method according to any one of the embodiments 2 and 2A to 2H, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is attached to the meta-position of the phenyl moiety.
  • Embodiment 2K relates to a method according to any one of the embodiments 2 and 2A to 2J, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein R1 is OH.
  • Embodiment 2L relates to a method in accordance to any one of the embodiments 2 and 2A to 2K, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein the amino group on the beta-amino acid side chain has the (R)-configuration.
  • Embodiment 2M relates to a method in accordance to any one of the embodiments 2 and 2A to 2K, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein the amino group on the beta-amino acid side chain has the (S)-configuration.
  • Embodiment 2N relates to a method according to embodiment 2, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof, wherein the compound is disclosed in WO2015/092740 [attorney docket number PAT056044-WO-PCT]; i.e.
  • the compound is selected from: (R)-3-amino-4-(5-(4-(benzo[d]thiazol-2-yloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-((5-chloropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-(oxazol-2-yl)-phenoxy)phenyl)-2H-tetrazol-2-yl)-butanoic acid; (R)-3-amino-4-(5-(3-(4-chlorophenoxy
  • the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) wherein the compound is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
  • the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (R)-3-amino-4-(5-(4-phenethoxyphenyl)-2H-tetrazol-2- yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
  • the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (R)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
  • the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid (compound 2); or a pharmaceutically acceptable salt thereof.
  • the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form (i.e. non-salt form); or a pharmaceutically acceptable salt thereof.
  • the invention relates to the method according to embodiment 3D, wherein the crystalline form is characterized by at least one of the following characteristics:
  • an x-ray powder diffraction pattern comprising four or more 2q values selected from the group consisting of 11 .3 ⁇ 0.2 °2q, 12.8 ⁇ 0.2 °2q, 15.2 ⁇ 0.2 °2q, 19.7 ⁇ 0.2 °2q, 20.0 ⁇ 0.2 °2q,
  • thermo gravimetric analysis (TGA) diagram substantially the same as that shown in FIG.
  • a compound of any one of Formulae (l)-(V), or a pharmaceutically acceptable salt thereof, or a compound according to any one of embodiments 1 , 2, 2A-2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof can be administered as a monotherapy of in combination with one or more additional therapeutic agent (e.g. an anti- steatotic agent, and anti-inflammatory agent, and anti-fibrotic agent or combination thereof).
  • additional therapeutic agent e.g. an anti- steatotic agent, and anti-inflammatory agent, and anti-fibrotic agent or combination thereof.
  • the combination partner (additional therapeutic agent) of the invention can be a farnesoid X receptor (FXR) agonist, e.g. compound of Formula A described below; or a pharmaceutically acceptable salt thereof.
  • FXR farnesoid X receptor
  • FXR farnesoid X receptor
  • the bile acid receptor, farnesoid X receptor (FXR) is a member of the nuclear hormone receptor superfamily, primarily expressed in liver, intestine and kidney. FXR acts as a sensor of elevated bile acids and initiates homeostatic responses to control bile acid levels and modulate other metabolic processes such as gluconeogenesis and lipogenesis (Pattni et al., clin Trans Gastroenterol, 2012, 3: e18; Walters et al., Aliment Pharmacol Ther. 2015, 41 (1):54-64). FXR agonism modulates bile acid synthesis and detoxifying metabolism.
  • FXR agonism in both the liver and the gut has the potential to provide multimodal benefits which, in brief, include: reduction of fat accumulation in liver via decreased de novo lipogenesis, reduction of hepatic inflammation, anti-fibrotic effects mediated by stellate cells via decreased oxidative stress, and reduction of bacterial and lipopolysaccharide translocation from gut which induces inflammation (Schapp et al., Nat Rev Gastroenterol Hepatol, 2014, 11 (1):55-67).
  • OCA obeticholic acid
  • ALT circulating alanine aminotransferase
  • T ropifexor 90 pg daily for 12 weeks improved liver fat and ALT versus placebo.
  • LTA4H inhibitor provides inflammatory effect in addition to those of a FXR agonist (such as Tropifexor) via inhibition of inflammation including hepatic neutrophil and macrophage infiltration and enhanced expression of the pro-resolving factor Lipoxin A4, which results in additional benefits in the treatment of liver disease.
  • FXR agonist such as Tropifexor
  • Suitable FXR agonists for use in the combination include, but are not limited to, obeticholic acid (so called OCA, Intercept), GS9674, elafibranor (GFT505), GW4064, UPF987, FXR-450, fexaramine, methylcolate, methyl deoxycholate, 5b-cholanic acid, 5b-chloanic acid 7a, 12a diol, NIHS700, marchantin A, marchantin E, MFA-1 INT767 (also called 6a-ethyl-CDCA disclosed in WO2014/085474), MET409 (Metacrine), EDP-305 (Enanta), 2-[(1R,3r,5S)-3-( ⁇ 5- cyclopropyl-3-[2-(trifluoromethoxy)phenyl]-1 ,2-oxazol-4-yl ⁇ methoxy)-8-azabicyclo[3.2.1]octan-8-
  • the FXR agonist for use in combination with a LTA4H inhibitor is a compound disclosed in patent application No. WO2012/087519, or a pharmaceutically acceptable salt thereof. More preferably, the FXR agonist for use in the combination with a LTA4H inhibitor is disclosed in WO2012/087519 and is 2-[3-( ⁇ 5-cyclopropyl-3-[2- (trifluoromethoxy)phenyl]-1,2-oxazol-4-yl ⁇ methoxy)-8-azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3- benzothiazole-6-carboxylic acid, a stereoisomer, an enantiomer, a pharmaceutically acceptable salt, solvate, prodrug, ester thereof and/or an amino acid conjugate thereof.
  • the FXR agonist for use in the combination is 2-[(1R,3r,5S)-3-( ⁇ 5-cyclopropyl-3-[2- (trifluoromethoxy)phenyl]-1,2-oxazol-4-yl ⁇ methoxy)-8-azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3- benzothiazole-6-carboxylic acid which is also represented by Formula (A):
  • the FXR agonist for use in the combination is 2-[(1 R, 3r, 5S)-3-( ⁇ 5-cyclopropyl-3-[2-(trifluoromethoxy)phenyl]-1 ,2-oxazol-4-yl ⁇ methoxy)-8- azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3-benzothiazole-6-carboxylic acid in its free form which is also known as Tropifexor or LJN452.
  • the present invention relates to a pharmaceutical combination comprising a therapeutically effective amount of a compound of any one of Formulae (l)-(V), or a compound according to any one of embodiment 1 , 2, 2A-2N, 3, and 3A-3E, or a pharmaceutically acceptable salt thereof, and a FXR agonist, separately or together.
  • the invention relates to a pharmaceutical combination comprising a therapeutically effective amount of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof and Tropifexor.
  • the pharmaceutical combination is a fixed combination.
  • the pharmaceutical combination is a non-fixed combination.
  • the pharmaceutical composition of the invention can be formulated to be compatible with its intended route of administration (e.g. oral compositions generally include an inert diluent or an edible carrier).
  • routes of administration include parenteral (e.g. intravenous), intradermal, subcutaneous, oral (e.g. inhalation), transdermal (topical), transmucosal, and rectal administration.
  • parenteral e.g. intravenous
  • intradermal subcutaneous
  • oral e.g. inhalation
  • transdermal topical
  • transmucosal and rectal administration.
  • the pharmaceutical compositions compatible with each intended route are well known in the art.
  • the pharmaceutical composition of the invention is formulated to be compatible with an oral administration.
  • the pharmaceutical composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the prevention or the treatment of liver disease, can be in unit dosage of about 1 -500 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1 -250 mg or about 1 -150 mg or about 1 -100 mg, or about 1 -50 mg of active ingredients.
  • the therapeutically effective dosage of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, the pharmaceutical composition is dependent on the species of the subject, the body weight, age and individual condition, the severity of the contrast-induced nephropathy disorder. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
  • Preferred formulation is a capsule or a tablet composition comprising from about 1 mg to about 160mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant.
  • the capsule or tablet composition comprises from about 5 mg to about 80mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant.
  • the capsule or tablet composition comprises about 1 mg, about 5mg, about 10mg, about 20mg, about 30mg, about 40mg and about 50mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant.
  • the fibrotic or cirrhotic disease and/or disorder can be a liver disease or disorder, e.g. as defined below herein, or renal fibrosis.
  • the liver diseases or disorders can be cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction,
  • PBC primary biliary cir
  • liver diseases or disorders can also refer to liver transplantation.
  • the pharmaceutical composition (as herein defined, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof) is for the treatment or prevention of a fibrotic disease or disorder, e.g. a liver disease or disorder, e.g. a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of PBC, NAFLD, NASH, drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis.
  • the pharmaceutical combination (as herein defined) is for the treatment or prevention of fibrosis, e.g. renal fibrosis, autoimmune hepatitis or liver fibrosis.
  • the pharmaceutical combination is for the treatment or prevention of a fibrotic disease or disorder, e.g. a liver disease or disorder, e.g. a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of PBC, NAFLD, NASH, drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis.
  • a fibrotic disease or disorder e.g. a liver disease or disorder, e.g. a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of PBC, NAFLD, NASH, drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithia
  • the liver diseases or disorders refer to NAFLD, e.g. any stages of NAFLD, e.g. any of steatosis, NASH, fibrosis and cirrhosis.
  • NAFLD e.g. any stages of NAFLD, e.g. any of steatosis, NASH, fibrosis and cirrhosis.
  • a pharmaceutical combination of the invention for the improvement of liver fibrosis without worsening of steatohepatitis
  • a pharmaceutical combination of the invention for obtaining a complete resolution of steatohepatitis without worsening, e.g. improving, of liver fibrosis.
  • a pharmaceutical combination of the invention for preventing or treating steatohepatitis and liver fibrosis.
  • a pharmaceutical combination of the invention for reducing at least one of the features of the NAS score (Brunt et al. Hepatology, 2011 , 53(3):810-820), i.e. one of hepatosteatosis, hepatic inflammation and hepatocellular ballooning; e.g. at least two features of the NAS score, e.g. hepatosteatosis and hepatic inflammation, or hepatosteatosis and hepatocellular ballooning, or hepatocellular ballooning and hepatic inflammation.
  • a pharmaceutical combination of the invention for reducing at least one or two features of the NAS score and liver fibrosis, e.g. for reducing hepatic inflammation and liver fibrosis, or hepatosteatosis and liver fibrosis or hepatocellular ballooning and liver fibrosis.
  • stage 3 fibrosis to stage 1 fibrosis, e.g. stage 3 and/or stage 2 and/or stage 1 fibrosis.
  • the dosing regimen i.e. administered doses and/or frequency of each component of the pharmaceutical combination may vary.
  • the frequency of dosing of the the LTA4H inhibitor of the invention e.g. compound of any one of Formulae (I) to (V), or a pharmaceutically acceptable salt thereof, or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the additional therapeutic agent e.g as a fixed dose combination
  • the LTA4H inhibitor e.g. compound of any one of Formulae (I) to (V), or a pharmaceutically acceptable salt thereof, or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the additional therapeutic agent e.g as a fixed dose combination
  • the additional therapeutic agent may not be administered following the same regimen, i.e. may not be administered at the same frequency and/or duration and/or dosage, e.g. at the same frequency and/or dosage. This can be the case e.g. for free combinations.
  • the LTA4H inhibitor can be administered twice a day and the additional therapeutic agent, e.g. Compound of Formula A (in free form or as a pharmaceutically acceptable salt, solvate, prodrug and/or ester thereof) once per day.
  • the co-administration is carried out for at least one week, at least one month, at least 6 weeks, at least 12 weeks, at least three months, at least 6 months, at least one year.
  • the pharmaceutical combination of the invention is administered lifelong to the patient.
  • the frequency of administration, and/or the doses of the LTA4H inhibitor and of the additional therapeutic agent, may vary during the whole period of administration.
  • the treatment there can be one or more periods of time, e.g. days, during which nor the LTA4H inhibitor of the invention neither the additional therapeutic agent, e.g. a Compound of Formula A, or a pharmaceutically acceptable salt thereof, are administered to the patient (i.e. periods, e.g. days, void of combination treatment), or during which only one drug amongst the LTA4H inhibitor or the additional therapeutic agent is administered to the patient.
  • periods of time e.g. days
  • the additional therapeutic agent e.g. a Compound of Formula A, or a pharmaceutically acceptable salt thereof
  • the LTA4H inhibitor may be administered prior the additional therapeutic agent, or reciprocally.
  • the time interval between administration of the LTA4H inhibitor and of the additional therapeutic agent may vary from a few minutes to a few days, e.g. a few minutes, e.g. a few hours, e.g. 1 day to 1 week.
  • LTA4H inhibitor e.g. Compound of any one of Formula (I) to (V) or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof (as hereinabove defined, e.g. in free form, crystalline or amorphous form), is administered at a dose of about 1mg to about 160mg, preferably about 4mg to about 100mg; e.g. about 10mg to about 100mg, e.g. about 5mg to about 80mg, e.g. about 20mg to 60mg delivered orally; e.g. about 20mg, e.g about 30mg, e.g.
  • Such doses may be for oral administration.
  • Such doses may be for daily administration, or twice daily administration or every two days administration, e.g. for daily oral administration, twice daily oral administration, e.g. about 5mgQD to about 40mg BID (twice a day), e.g. about 10mg BID to about 30mg BID, e.g. about 10mg BID, or about 15mg BID, preferably about 20mg BID; or every two days oral administration.
  • Dose selection for LTA4H inhibitor was determined from human safety and tolerability, pharmacokinetic and pharmacodynamic data obtained during first-in human clinical study.
  • the second therapeutic agent i.e. the non-bile acid derived FXR agonist, e.g. Compound of Formula A (as hereinabove defined, e.g. in free form or as a pharmaceutically acceptable salt thereof), is administered at a dose of about 1 mg to about 250mg, e.g. about 5mg to about 150mg, e.g. about 10mg to about 150mg, e.g. about 20mg to 150mg, e.g. about 90mg to about 250mg; preferably about 140mg to about 200mg delivered orally, e.g. about 30mg, e.g. about 60mg, e.g. about 90mg, e.g.
  • Such doses may be for oral administration.
  • Such doses may be for daily administration, or twice daily administration or every two days administration, e.g. for daily oral administration, twice daily oral administration or every two days oral administration.
  • Dose selection for FXR agonist was determined from human safety and tolerability, pharmacokinetic and pharmacodynamic data obtained during first-in human clinical study.
  • Disclosed herein are methods of treating or preventing liver disease comprising orally administering to a patient in need thereof a dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg) of a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), daily.
  • Said doses can be administered to the patient either with a once a day dosing regimen or twice a day dosing regimen.
  • the method comprises orally administering to a patient in need thereof a dose of about 10mg QD to about 40mg BID of a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof).
  • a LTA4H inhibitor i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof.
  • the method comprises orally administering to a patient in need thereof a dose of about 10mg to about 30 mg twice a day (e.g. about 10mg BID, about 15mg BID, preferably about 20mg BID) of a LTA4H inhibitor (i.e.
  • the method comprises orally administering a dose of about 20 mg of LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), to said patient twice a day (BID).
  • LTA4H inhibitor i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
  • the method comprises orally administering a dose of about 10mg QD to about 40mg BID of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2- yl)butanoic acid, or a pharmaceutically acceptable salt thereof, to said patient.
  • the method comprises orally administering a dose of about 10mg to about 30 mg twice a day (e.g.
  • the method comprises orally administering a dose of about 20mg of (S)-3-amino- 4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, to said patient twice a day.
  • LTA4H inhibitor is a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form, as described herein.
  • the method comprises orally administering a dose of about 10mg QD to about 40mg BID of a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, as described herein, to said patient.
  • the method comprises orally administering a dose of about 10mg to about 30 mg twice daily (e.g.
  • the method comprises orally administering a dose of about 20mg of a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2- yl)butanoic acid, as described herein, to said patient twice a day.
  • LTA4H inhibitor i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the LTA4H inhibitor is administered orally to a patient in need thereof in a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg).
  • LTA4H inhibitor e.g.
  • a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof is administered orally to a patient in need thereof in a dose of about 10mg QD to about 40mg BID.
  • LTA4H inhibitor e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day e.g. about 10mg BID, or about 15mg BID, or preferably about 20mg BID).
  • LTA4H inhibitor e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • a patient in need thereof in a dose of about 20 mg twice a day.
  • LTA4H inhibitor which is (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a liver disease or disorder, wherein the LTA4H is administered orally to a patient in need thereof a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g.
  • (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H- tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof is administered orally to a patient in need thereof in a dose of about 10mg QD to about 40mg BID.
  • (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H- tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day (e.g. about 10mg BID, about 15mg BID, about 20mg BID).
  • (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof is administered orally to a patient in need thereof in a dose of about 20mg twice a day.
  • LTA4H inhibitor which is a crystalline form of (S)-3-amino-4-(5-(4- ((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, for use in the treatment or prevention of a liver disease or a disorder wherein the LTA4H is administered orally to a patient in need thereof in a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g.
  • the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, is administered orally to a patient in need thereof at a dose of about 10mg QD to about 40mg BID.
  • the crystalline form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day (e.g about 10mg BID, about 15mg BID, or preferably about 20mg BID).
  • the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, is administered orally to a patient in need thereof at a dose of about 20mg twice a day.
  • the dose of the LTA4H inhibitor is about 10mg to about 30mg BID or about 20 mg BID.
  • kits prior to treatment with a LTA4H inhibitor as disclosed herein, the patient has been previously treated with a systemic agent selected from obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
  • a systemic agent selected from obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
  • kits prior to treatment with a LTA4H as described herein, the patient has not been previously treated with a systemic agent such as obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
  • a systemic agent such as obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
  • the LTA4H inhibitor as described herein e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the LTA4H inhibitor as described herein is administered in combination with a FXR agonist.
  • the FXR agonist is T ropifexor.
  • the dose of the LTA4H inhibitor as described herein e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the dose of the LTA4H inhibitor is about 10mg to about 30mg BID.
  • the dose of the LTA4H inhibitor is about 20 mg.
  • the patient is a NAFLD patient with a biomarker phenotype consistent with ongoing liver inflammation and fibrosis such as a NASH-like phenotype as defined based on the presence of all five of the following:
  • the patient is an adult of 18 years and older. In some embodiments of the disclosed methods, uses and kits, the patient must weigh at least 40kg and no more than 150kg.
  • a LTA4H inhibitor as described herein e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test 38.5 and £10.5.
  • ELF Enhance Liver Fibrosis
  • a LTA4H inhibitor as described herein e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF) 38%.
  • MRI-PDFF Magnetic Resonance Imaging-Proton Density Fat Fraction
  • a LTA4H inhibitor as described herein e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof
  • the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) 343 IU/L for male and 328 IU/L for female.
  • ALT Alanine aminotransferase
  • the ALT does not exceed 5 times the upper limit of normal.
  • the patient achieves a change from baseline as measured by ALT, percent liver fat (MRI-PDFF) or ELT Test by week 12 of treatment.
  • MRI-PDFF percent liver fat
  • the patient achieves at least a 3% decrease of ELF by week 12 of treatment.
  • the patient further achieves at least a 10% reduction in ALT, and preferably at least 35% reduction in ALT at week 12 of treatment.
  • the patient achieves at least a 3% decrease of ELF together with at least 10% reduction of ALT together with at least 10% reduction of percent liver fat by week 12 of treatment. In another embodiment of the disclosed methods, uses and kits, the patient achieves at least a 3% decrease of ELF together with at least 10% reduction of ALT together with at least 15% reduction of percent liver fat by week 12 of treatment. In a non-limiting example, the patient achieves a 3% reduction of ELF, a 10% reduction of percent liver fat and a 30% reduction of ALT by week 12 of treatment.
  • the patient achieves between 2% and ⁇ 3% decrease of ELF together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat at week 12 of treatment. In another preferred embodiment of the disclosed methods, uses and kits, the patient achieves between 2% and ⁇ 3% decrease of ELF together with a reduction of at least 35% ALT by week 12 of treatment. In a non-limiting example, the patient achieves by week 12: a 2.5% reduction of ELF, a 10% reduction of percent liver fat and a 40% reduction of ALT.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 30% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 35% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 35% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment. In a non-limiting example, the patient achieves an ELF reduction of 1 %, together with a 30% fat reduction and a 40% ALT reduction by week 12 of treatment.
  • the patient achieve by week 12 of treatment one of the following: a) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or b) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or c) between 2% and ⁇ 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 30% of percent liver fat by week 12 of treatment.
  • the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 35% of percent liver fat by week 12 of treatment.
  • the patient achieves an ELF reduction of 1 %, together with a 30% fat reduction and a 40% ALT reduction by week 12 of treatment.
  • the patient is treated with a LTA4H inhibitor, as described herein, for at least 12 weeks, at least 16 weeks, at least 24 weeks, at least 48 weeks or at least 52 weeks. Most preferably, the patient is treated for at least 12 weeks.
  • the LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
  • the LTA4H inhibitor is a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form (herein designated as Form B).
  • the LTA4H inhibitor is Form B as described in embodiment 3E, in a substantially pure phase.
  • the invention relates to a pharmaceutical combination comprising a therapeutically effective amount of a compound of any one of Formulae (I)- (V) or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof and a FXR agonist, for simultaneous, sequentially or separate administration.
  • the compound of Formula (I) is (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof
  • the FXR agonist is tropifexor.
  • the combination is a fixed dose combination.
  • the combination is a free combination.
  • the invention relates to a combination as described above for use in the treatment and prevention of a liver disease.
  • the invention provides a combination for use in treating and preventing a liver disease, wherein the LTA4H inhibitor is administered at a dose in a range of about 10mg to about 100mg.
  • the invention provides a combination for use in treating and preventing a liver disease, wherein the FXR agonist is administered at a dose in a range of about 1 mg to about 250mg.
  • Item 1 A method of treating or preventing a liver disease, comprising administering to a subject in need thereof a therapeutically effective amount of a LTA4H inhibitor.
  • R 1 is OH or NH 2 ;
  • Y is O, S or CH 2 ;
  • X 1 , X 2 , X 3 and X 4 are N; or
  • X 1 , X 2 , X 3 and X 4 are selected from N, NH, C, CH and O with the proviso that at least two of X 1 , X 2 , X 3 and X 4 are N or NH;
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; C 3 -C 6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C 1 -C 6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable salt thereof.
  • Item 4 The method according to any of the above items, wherein the LTA4H inhibitor is disposed in a pharmaceutical formulation, wherein said pharmaceutical formulation comprises one or more pharmaceutically acceptable carriers, each of which is independently selected from a filler, a lubricant, a binder, a desintegrant and a glidant.
  • the pharmaceutical formulation is in tablet or capsule form.
  • Item 6 The method according to any of the above items, wherein the LTA4H inhibitor is administered in combination with a FXR agonist, and wherein each of the components of the combination are administered simultaneously, or sequentially, or in any order.
  • Item 7 The method according to item 6, wherein the FXR agonist is Tropifexor.
  • Item 8 The method according to item 6 or 7 wherein the FXR agonist is administered at a dose range of about 1 mg to about 250mg.
  • Item 9 The method according to any of the above items, wherein the LTA4H inhibitor is administered at a dose of about 10 mg to about 30 mg twice a day.
  • Item 10 The method according to any of the above items, wherein the patient is a NAFLD patient selected according to at least one of the following criteria: a) prior to the treatment with LTA4H inhibitor, the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test 38.5 and £10.5; b) prior to treatment with the LTA4H inhibitor, the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI- PDFF) 38%; c) prior to treatment with the LTA4H inhibitor, the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) 343 IU/L for male and 328 IU/L for female.
  • ELF Enhance Liver Fibrosis
  • Item 11 The method according to any of the above items, wherein said patient achieves by week 12 of treatment one of the following: g) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or h) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or i) between 2% and ⁇ 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in
  • L less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF).
  • ELF Enhance Liver Fibrosis
  • ALT Alanine aminotransferase
  • MRI-PDFF Magnetic Resonance Imaging-Proton Density Fat Fraction
  • liver disease or disorder is a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibro
  • a liver disease or disorder selected from the group consisting of
  • a LTA4H inhibitor for use in the treatment and/or the prevention of a liver disease or disorder in a patient in need of such treatment and/or prevention.
  • LTA4H inhibitor for use according to item 13, wherein said LTA4H inhibitor is a compound of Formula (I), or a pharmaceutically acceptable salt thereof: wherein,
  • R 1 is OH or NH 2 ;
  • Y is O, S or CH 2 ;
  • X 1 , X 2 , X 3 and X 4 are N; or
  • X 1 , X 2 , X 3 and X 4 are selected from N, NH, C, CH and O with the proviso that at least two of X 1 , X 2 , X 3 and X 4 are N or NH;
  • R2 is C 1 -C 6 alkyl optionally substituted by phenyl; C 3 -C 6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by halogen, C 1 -C 6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C 1 -C 6 alkyl optionally substituted by halogen.
  • Item 15 is C 1 -C 6 alkyl optionally substituted by phenyl; C 3 -C 6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C 1 -C 6 alkyl optionally substituted by
  • a LTA4H inhibitor for use according to item 13 or 14, wherein said LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition comprising a LTA4H inhibitor which is a compound of Formula (I), or a pharmaceutically acceptable salt thereof in accordance to item 14, together with one or more pharmaceutically acceptable carriers for use in the treatment and/or prevention of liver disease or disorder in a patient in need of such treatment and/or prevention.
  • Item 19 A pharmaceutical combination comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof according to item 14, and a FXR agonist.
  • Item 20 A pharmaceutical combination according to item 19, wherein the compound of Formula (I) is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof and the FXR agonist is Tropifexor.
  • Item 21 A pharmaceutical combination according to item 19 or 20, for use in the treatment and/or prevention of a liver disease or disorder in simultaneous, sequentially or separate administration.
  • Item 22 A LTA4H inhibitor for use according to any one of items 13 to 17 or a pharmaceutical composition for use according to item 18, a pharmaceutical combination for use according to item 19-21 , wherein the LTA4H inhibitor is administered at a dose of about 10 mg to about 30mg twice a day.
  • Item 23 A LTA4H inhibitor for use according to any one of items 13 to 17 or a pharmaceutical composition for use according to item 18, or a pharmaceutical combination for use according to item 19-21 , wherein the FXR agonist is administered at a dose in a range of about 1mg to about 250mg.
  • EMF Enhance Liver Fibrosis
  • Item 25 A LTA4H inhibitor for use according to any one of items 13 to 17 and 22-24, or a pharmaceutical composition for use according to any one of items 18 and 22-24, or a pharmaceutical combination for use according to any one of items 19 to 24, wherein said patient achieves by week 12 of treatment at least one of the following: a) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or b) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or c) between 2% and ⁇ 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver
  • Item 26 A LTA4H inhibitor for use according to any one of items 13 to 17 and 22-25, or a pharmaceutical composition for use according to any one of items 18 and 22-25, or a pharmaceutical combination for use according to any one of items 19 to 25, wherein a liver disease or disorder is selected from chronic liver disease, e.g.
  • a liver disease or disorder selected from the group consisting of cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibrosis, dyslipidemia, atherosclerosis, diabetes, diabetic nephropathy, colitis, newborn jaundice, prevention
  • Example 1 Crystalline form B of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid
  • Differential scanning calorimetry was conducted for each crystalline form using a TA Instruments (DSC 2500). For each analysis, 2-4 mg of sample was placed in an aluminium T- zero crucible that closed with a pin-hole lid. The heating rate was 10°C per minute in the temperature range between 30 and 300°C. Temperatures are reported in degrees Celsius (°C) and enthalpies are reported in Joules per gram (J/g). Plots are showing endothermic peaks as down. The endothermic melt peak (melting point) was evaluated for extrapolated onset temperature. The accuracy of the measured sample temperature with this method is within about ⁇ 1 °C, and the heat of fusion can be measured within a relative error of about ⁇ 5%.
  • TGA curves were obtained using a TA-instrument Q5000. 5-15mg of sample was placed into an aluminum crucible and sealed hermetically. The sealed crucible was pierced by the robotic auto sampler immediately before analysis. The TGA curve was measured at 10°C/min between 30- 300°C. The LoD (Loss of drying) was calculated between 40°C and 150°C. The weight loss is plotted against the measured sample temperature. Temperatures are reported in degrees Celsius (°C) and weight loss in %.
  • Example 3 Liver transcriptomic analysis of mice fed a high fat, high fructose NASH (HF/NASH) diet
  • C57BL/6J mice were fed a high fat, high fructose NASH (HF/NASH) diet up to 24 weeks to induce NASH like pathology in liver including steatosis , lobular inflammation and fibrosis accompanied by elevated liver injury enzymes, ALT and AST.
  • HF/NASH high fat, high fructose NASH
  • RNA extracted was prepared from snap frozen liver tissue a homogenate was prepared and RNA extracted.
  • cDNA of the samples was prepared from the same starting amount of RNA using a high capacity cDNA reverse Transcription Kit (Applied Biosystems). 10 ng/ul of cDNA for each sample and qPCR was conducted using TaqMan technology. TBP (TATA-Binding Protein) was used as control gene. The changes of mRNA levels compared with untreated sample were calculated according to the manufacturer’s instructions (DCt).
  • Figure 4 shows normalized expression of the ALOX5 gene (5-Lipoxygenase enzyme).
  • a significant upregulation of ALOX5 in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow could be observed as of week 12. Expression continued to rise until week 24.
  • Figure 5 shows normalized expression of the ALOX5AP gene (5-Lipoxygenase accessory protein, FLAP).
  • a significant upregulation of ALOX5AP in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow could be observed as of week 12. Expression continued to rise until week 24.
  • Figure 6 shows normalized expression of the LTA4H gene (Leukotriene A4 hydrolase).
  • LTA4H Leukotriene A4 hydrolase
  • a strong expression of LTA4H versus control gene TBP was observed in all liver biopsies. Expression was not different between mice on high fat, high fructose diet versus normal chow. **** P ⁇ 0.0001 ; *** P ⁇ 0.001 ; ** P ⁇ 0.01 ; * P ⁇ 0.05, applying a two-tailed unpaired t-test.
  • Example 3b Treatment with (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2- yl)butanoic acid (compound 2) attenuates the development of high fat, high fructose- induced NASH
  • mice were exposed for 20 weeks to a high fat, high fructose diet. Control mice were left on normal chow. 8 weeks after starting the high fat, high fructose NASH diet, mice were treated with 3 mg/kg/day or 10 mg/kg/day p.o. of the LTA4H inhibitor compound 2 or vehicle control until week 20.
  • mice on high fat, high fructose diet showed substantially increased body weight, accompanied by histomorphological signs of NASH, such as liver steatosis, lobular liver inflammation and liver enzyme aberration.
  • Mice treated with the LTA4H inhibitor compound 2 showed significantly reduced signs of liver inflammation, in particular reduced numbers of liver neutrophils and macrophages. Also elevation of liver enzymes ALT and AST was significantly reduced upon treatment with compound 2, indicating the protective effect of LTA4H inhibition in this diet-induced NASH model.
  • FIG. 7 IBA1 positive liver tissue macrophages of mice treated with two doses of the LTA4H inhibitor compound 2 were compared to vehicle treated mice and mice on normal chow.
  • FIG. 8 LY6B positive liver tissue neutrophils of mice treated with two doses of the LTA4H inhibitor compound 2 were compared to vehicle treated mice and mice on normal chow.
  • the LY- 6b stained area was quantified and showed a 42% and 49% reduction in liver tissue neutrophiles in mice treated with 3mg/kg and 10 mg/kg compound 2 as compared to vehicle control.
  • FIG. 9 Liver enzymes ALT and AST were determined in blood of mice fed a high fat, high fructose NASH diet and treated with 3 mg/kg and 10 mg/kg compound 2 versus vehicle control and mice on normal chow. AST blood levels were reduced by 31% and 45% with 3 mg/kg and 10 mg/kg compound 2, respectively. ALT level were reduced by 30 % with 3 mg/kg compound 2 and 46% with 10 mg/kg compound 2. Depicted are international units of enzyme per liter blood with standard deviation. *p ⁇ 0.05; **p ⁇ 0.01 ; ***p ⁇ 0.001 ; ****p ⁇ 0.0001 compared to vehicle control; one-way ANOVA with a post-hoc Dunnett's test.
  • FIG. 10 Mice were treated with either vehicle, 3 mg/kg or 10 mg/kg compound 2. 22 hours after the last dose, blood was obtained, diluted 1 :3 and stimulated for 15 min ex-vivo with 10 mg/ml calcium ionophore to induce LTB4 production. Release of LTB4 was quantified via LTB4 EIA and demonstrated over 90% inhibition of LTB4 in both treated groups. Depicted are averages of 4 mice per group +/- standard error of the mean.
  • Compound of example 1 has been studied in a FIH study designed to characterize its preliminary safety, tolerability, and PK in adult healthy subjects.
  • the study consisted of a part 1 , single ascending dose (SAD) and a part 2 multiple ascending dose (MAD).
  • Compound of example 1 was administered orally in single either QD or BID, in fed or fasted conditions over a dose range of 5 mg to 2 times 100 mg.
  • Compound of example 1 was also administered orally in multiple-dose administration, in fasted conditions for 12 days over a dose range of 5 mg QD to 80 mg BID.
  • Part 1 was a randomized, subject-blinded and Investigator-partial-blinded, placebo-controlled, single ascending oral dose study.
  • Eight subjects were randomized into each cohort (except in Cohort 7 where seven subjects were randomized and in Cohort 9 where six subjects were randomized) to receive either compound of example 1 or matching placebo in a 6:2 ratio (active: placebo), testing nine dose levels.
  • Cohort 1 Single oral dose of 5 mg or matching placebo
  • Cohort 2 Single oral dose of 10 mg or matching placebo
  • Cohort 3 Single oral dose of 20 mg or matching placebo
  • Cohort 4 Single oral dose of 30 mg or matching placebo
  • Cohort 5 Single oral dose of 45 mg or matching placebo
  • Cohort 6 Single oral dose of 70 mg or matching placebo
  • Cohort 7 total oral dose of 140 mg or matching placebo split into two doses of 70 mg taken 12h apart
  • Cohort 8 total oral dose of 200 mg or matching placebo split into two doses of 100 mg taken 12h apart
  • Cohort 9 total oral dose of 80mg or matching placebo split into two doses of 40 mg taken 12h apart
  • Part 2 was a randomized, subject-blinded and investigator-partial-blinded, placebo-controlled, MAD study in which, eight subjects each were randomized into five cohorts to receive either compound of example 1 or matching placebo in a 6:2 ratio (active: placebo).
  • Cohort 1 multiple once daily (qd) oral dose of 5 mg or matching placebo
  • Cohort 2 multiple once daily (qd) oral dose of 15 mg or matching placebo
  • Cohort 3 multiple oral daily dose of 20mg or matching placebo administered twice daily (bid)
  • Cohort 4 multiple oral daily dose of 40mg or matching placebo administered twice daily (bid)
  • Cohort 5 multiple oral daily dose of 80mg or matching placebo administered twice daily (bid)
  • Part 1 Single Ascending Dose, SAD
  • Part 2 Multiple Ascending dose, MAD
  • the subjects received doses up to 80 mg BID for 12 days. No adverse events led to study discontinuation.
  • the highest doses in both parts were safe and no maximal tolerated dose was established.
  • compound of example 1 was well tolerated in healthy subjects at doses up to 80 mg bid (160 mg daily dose) over 12 days.
  • PK parameters calculated were standard parameters used for measuring drug exposure in the systemic circulation after receiving single or multiple doses of compound of example 1 .
  • the plasma exposure to compound of example 1 increased with dose and a median Tmax ranging from 1 to 1 .5 hours post dose indicated a fast absorption.
  • concentration peak plasma concentrations decreased initially very rapidly; at later timepointsthe rate of concentration decline decreased strongly and the mean apparent elimination half-life (T1/2) ranged from 245 to 513 hours.
  • T1/2 mean apparent elimination half-life
  • Cmax increased by 21 .7-fold
  • AUC0-24h increased by 75 fold, with a 40-fold increase in daily dose (5 mg to 100 mg bid).
  • the percentage coefficient of variation (CV%) ranged from 18.5 to 41 .4 for Cmax and 6.4 to 26.3 for AUC0-24h.
  • the plasma concentrations of compound of example 1 increased in a dose dependent manner with a median T max ranging from 1 to 2.5 hours.
  • Cmax and AUC on Day 12 (AUC0-12h,ss for 20, 40 and 80 mg bid cohorts and AUC0-24h,ss for 5 and 15 mg qd cohort) increased by 17.9-fold and 20.9-fold respectively.
  • the estimated slope and the corresponding 90% Cl for Cmax.ss was 1 .19 (1 .08,1 .30) and for AUCtau.ss the slope was 1 .07 with 90% Cl (1 .01 ; 1.12). Dose proportionality over the whole dose range was not demonstrated for both Cmax.ss and AUCtau.ss.
  • Cmax.ss and AUCtau PK can be considered dose proportional for and up to 2.12 and 6.29 fold increase in dose, respectively.
  • Dose proportionality criteria were met for AUCtau over the dose range 5 mg qd to 40 mg bid.
  • the % CV ranged from 17.1 to 30.2 % for Cmax and 11.7 to 18.1 for AUC0-24h.
  • Cmax.ss was between 1.39 to 1.19 times greater than after single dose (mean Cmax (Day 12)/ Cmax (Day 1)).
  • AUC AUC0-12 and AUC0-24
  • the average ratios were between 2.08 to 1 .27. This indicates minor accumulation of compound of example 1 from day 1 to steady state.
  • morning pre-dose concentrations are higher on Day 3 as compared to Day 2, but the concentration differences are small.
  • LTB4 Leukotriene B4
  • LTB4 concentrations were determined in plasma of ex-vivo stimulated whole blood by LC-MS/MS method.
  • Ex-vivo stimulation was performed at the clinical site according to the following procedure: Whole blood (500mL) were stimulated for 30 min using Calcium lonophore A23187. Then plasma supernatant is harvested, and frozen at -80°C for at least 24 hours prior to use.
  • Human plasma was processed with organic precipitation followed by separation by reverse phase high performance liquid chromatography with tandem mass spectrometric detection.
  • Sample from each subject and time point was prepared and analyzed in three aliquots. Aliquot one and two contained plasma from ex-vivo stimulated whole blood, while aliquot three as negative control contained plasma from non-stimulated whole blood. Samples with CV>25% for the first two aliquots and/or with detected values in aliquot three were excluded from the results.
  • LTB4 concentration data were calculated as mean of concentration of aliquot one and two. Concentration of aliquot three was not be included in the calculation. The change from baseline and percent inhibition (percent of baseline) for each biomarker was calculated. Baseline was the mean of the Baseline and Day 1 , -1 h measurements.
  • Target inhibition of 90-99% was maintained at 15 mg (Q.D.) and 20 mg (B.I.D.) doses, in the MAD part of the study.
  • steady state leukotriene A4 hydrolase target inhibition assessed ex-vivo in blood samples was on average 75% at 5mg QD, 90% at 15mg QD and 99% at 20mg QD bid.
  • Example 5 Efficacy and safety of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Form B) in adult patients with NAFLD who manifest a NASH-like biomarker phenotype
  • the NEXSCOT study uses a platform type design to investigate “multiple targeted therapies in the context of a single disease in a perpetual manner” (Woodcock and LaVange 2017; N Engl J Med, 377(1), 62-70). Each single or combination regimen will be considered a unique treatment arm.
  • One or more treatment arms which are bundled together for review and execution are considered a cohort.
  • the treatment arm(s) within a cohort will be studied in a way that allows for the comparison of the treatment effect.
  • a particular treatment arm or cohort will be independent and will not necessarily provide decision making information for future treatment arms or cohorts.
  • Each cohort will consist of a 33-day screening period (Day -60 to Day -28), a baseline period of 27 days (Day -27 to Day -1), a treatment period of 12 weeks (Day 1 to Day 85), and a study completion evaluation approximately 28 days after the last drug administration (Day 113). Patients will be advised to maintain their recommended diet for NAFLD during the study.
  • NASH Two risk factors for NASH: elevated BMI and history of Type II diabetes mellitus, and • Three phenotypic biomarkers for NASH as shown in Table 2: elevated % liver fat diagnostic of NAFLD as measured by MRI-PDFF, liver inflammation as measured by ALT, and ongoing liver fibrotic process as measured by ELF Test. These phenotypic biomarkers are consistent with the pathobiology of NASH and recent studies have supported these biomarkers being highly predictive of NASH (Harrison et al 2018a; J Hepatol Suppl, 68(1), p. S38; Sanyal et al 2017; J Hepatol Suppl, 66(1), p. S89).
  • ELF Test measurement will be conducted at screening (Inclusion criteria), and patients with an ELF Test result within the range of 8.5-10.5 will be eligible for participation in the study.
  • the goal of using these ELF Test inclusion criteria is to enrich the study population for those subjects who have an ongoing fibrotic process in the liver but do not yet have severe fibrosis or cirrhosis, typically considered to be F1-3.
  • This ELF Test inclusion range is based on a weight- of-evidence analysis of the following reports:
  • the cohort 1 consist of two treatment arms
  • Safety and tolerability will be the paramount and primary objective, assessed by safety endpoints (including vital signs, physical examination, laboratory measurements, ECG) and adverse events.
  • Secondary objectives are to measure efficacy.
  • the secondary endpoints are
  • Cardiometabolic risk factors NAFLD and NASH patients are at high risk for cardiovascular morbidity and mortality. Therefore, it is important to characterize any negative or positive effects of single drugs or drug combinations on cardiometabolic risk factors including body weight, Homeostasis Model Assessment (HOMA), hemoglobin A1C (HbA1c), fasting glucose, fasting insulin, and fasting lipid profile.
  • HOMA Homeostasis Model Assessment
  • HbA1c hemoglobin A1C
  • fasting glucose fasting insulin
  • fasting lipid profile fasting glucose, fasting insulin, and fasting lipid profile.
  • ALT % liver fat
  • ELF Test ELF Test
  • Efficacy endpoint Conversely, a review of multiple, recent studies in patients with NASH or a NASH-like phenotype shows that high efficacy, active treatment is associated with relative percent decreases in % liver fat (MRI-PDFF) and ALT of 3 30% (Sanyal et al 2017; Harrison et al 2018a; Harrison et al 2018b). Similarly, a decrease in ELF Test by 3 0.3 units or 3% can be considered evidence of a substantive, beneficial reversal in the ongoing liver fibrotic process (Harrison et al 2018a).

Abstract

The present disclosure relates to methods for treating or preventing a liver disease, using a LTA4H inhibitor, alone or in combination with another therapeutic agent, e.g. a FXR agonist. Also disclosed herein are LTA4H inhibitors, for treating or preventing a liver disease or disorder, as well as medicaments, dosing regimens, pharmaceutical formulations, combinations, dosage forms, and kits for use in the disclosed uses and methods.

Description

METHODS OF TREATING LIVER DISEASE USING LTA4H INHIBITORS
TECHNICAL FIELD
The present disclosure relates to methods for treating or preventing liver disease using leukotriene A4 hydrolase (LTA4H) inhibitors.
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the Western world (Ratziu et al., J Gastrointestin Liver Dis. 2010 19(4):415-23). The main stages of NAFLD are 1- simple fatty liver (steatosis); 2- non-alcoholic steatohepatitis (NASH), a more serious form of NAFLD; 3- fibrosis, where there is a persistent inflammation in the liver resulting in the generation of fibrous scar tissue around the liver cells and blood vessels; and 4-cirrhosis; this damage is permanent and can lead to liver failure and liver cancer.
NASH includes fat accumulation in the liver, as well as inflammation which over time can lead to increased fibrosis, cirrhosis and end stage liver disease. Liver transplantation is the only treatment for advanced cirrhosis with liver failure, and transplantation is increasingly performed in persons suffering from NASH.
Estimates of the worldwide prevalence of NAFLD range from 6.3% to 33% with a median of 20% in the general population. The estimated prevalence of NASH is lower, ranging from 3 to 5% (Younossi et al., 2016, Hepatology, Vol. 64(5):1577-1586). NASH is a worldwide problem with growing prevalence over the last few decades. Over the last decade, NASH has risen from uncommon to the second indication for liver transplantation in the US. It is expected to be the leading cause of transplant by 2020 (Wong, et al, Gastroenterology, 2015, 148(3):547-55).
NASH is highly associated with the metabolic syndrome and Type 2 diabetes mellitus. NASH is a cause of progressive fibrosis and of cirrhosis. Cirrhosis due to NASH increases the risk of hepatocellular carcinoma and hepatocellular cancer. Furthermore, cardiovascular mortality is an important cause of death in NASH patients.
Chronic cholestasis and liver inflammation are the two main pathophysiological components of the two major classes of disease - primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC) - leading to bile duct destruction and ultimately to cirrhosis and liver failure. Liver transplantation appears to be the only life-saving procedure.
Ursodeoxycholic acid (UDCA), also known as ursodiol, is the main treatment for PBC. UDCA is a secondary bile acid, i.e. it is metabolized from a primary bile acid (produced by the liver) by intestinal bacteria, after the primary acid has been secreted into the intestine. UDCA halts progression in many patients, but about 30-40% of the population do not respond. Since May 2016, another molecule has been approved in the US for the treatment of PBC, when combined with UDCA for primary biliary cholangitis (PBC) in adult patients with an inadequate response to UDCA, or as a single therapy in adults unable to tolerate UDCA. This new molecule is Obeticholic acid (OCA), a bile-acid mimetic. OCA is a FXR (Farnesoid X receptor) agonist. When tested in nonalcoholic steatohepatitis patients, obeticholic acid showed efficacy, in particular a significant improvement in NASH, i.e. strong impact on steatosis with additional effects on inflammation and ballooning. But OCA long term administration raises safety concerns because it can be associated with pruritus, as well as with increased LDL cholesterol (see “Intercept Announces New FLINT Trial Data Showing OCA Treatment Increases Fibrosis Resolution and Cirrhosis Prevention in High-Risk NASH Patients”, April 23, 2015). To avoid the risk of adverse cardiovascular events, concomitant administration of statins may be required for long term treatment of NASH patients.
Currently there is no approved treatment for either NAFLD or NASH. As a result, patients suffering from NAFLD or NASH often end up needing a liver transplant. There remains a need for efficacious treatments and therapies for liver conditions, in particular liver diseases such as NAFLD, NASH or PBC, and for late stage liver diseases.
For preventing or treating such diseases or disorders, a medicament would be particularly efficient if it has an impact on one or more of the different aspects of the liver disease. Therefore, there is a need to provide treatments for fibrotic / cirrhotic diseases or disorders, e.g. liver diseases or disorders, that can address the different aspects of these complex conditions, while demonstrating an acceptable safety and/or tolerability profile and ultimately avoiding the need for liver transplant. The combination of two or more molecules with different Mechanisms of Action (MoA) might provide additional benefits for improving treatment efficacy and response rates.
SUMMARY OF THE INVENTION
As previously mentioned, one approach to the treatment of NASH is administering to a patient in need thereof, a compound with anti-steatotic properties, anti-inflammatory properties and/or anti-fibrotic properties; or a compound with mixed effects. The compounds of the invention, i.e. leukotriene A4 hydrolase (LTA4H) inhibitors such as compounds of Formula (I), or pharmaceutically acceptable salt thereof, inhibit LTA4H and thereby prevent the biosynthesis of pro-inflammatory leukotriene B4 (LTB4).
In obesity, myeloid cells, hepatocytes and adipocytes are the source of LTB4. LTB4 is responsible for driving the three main processes in the pathophysiology of NASH: Inflammation, insulin resistance and white adipose tissue (WAT) generation and lipolysis in adipocytes (Oh et al. Nat Rev Drug Disc; 2016, 15:161-172).
Leukotriene B4 (LTB4), which recruits and activates neutrophils and macrophages, changes macrophage phenotype in adipose tissue to a pro-inflammatory phenotype. (Li et al. Mol Endocrinol. 2014, 28(8): 1316-28; Spite et al. J Immunol; 2011 , 187(4):1942-9).
Adipocyte derived LTB4 has also been shown to contribute to “low-grade” inflammatory state leading to obesity associated co-morbidities (Mothe-Satney et al. Diabetes 2012, 61 (9):2311-9).
Furthermore, we have demonstrated in in-house studies which was performed in a high fat/high fructose (HFHF) NASH model, that LTA4H inhibitors of the present invention reduce liver inflammation and damage including reduction in alanine aminotransferase (ALT) biomarker, fibrotic gene expression, hepatic neutrophils, macrophages and macrophage crown like structures. In this model, we have also demonstrated that LTA4H of the present invention also reduces TIMP-1 and PIII-NP serum fibrosis biomarkers. Additionally we have demonstrated that LTA4H inhibition can shift production of pro-inflammatory lipid mediator LTB4 to anti-inflammatory lipid mediator Lipoxin A4 (LXA4).
LXA4 has been shown to attenuate obesity induced liver inflammation, injury & fibrosis (Borgeson et al. Cell Metab; 2015 22(10): 125-37). LXA4 is also known to protect against diet induced obesity and insulin resistance, increases energy expenditure and browning of fat. (Elias et al.; Diabetes 2016, 65(8) :2139-50).
Finally, we have shown in an in-house study performed in a STZ-HFD (streptozotocin- induced high fat diet) model of NASH, that LTA4H inhibitor reduced liver inflammation including hepatic neutrophils and macrophages and fibrotic gene expression in liver.
Therefore the aim of the present invention is to provide novel methods for treating or preventing liver disease in a subject in need thereof, comprising administering to said subject, a therapeutically effective amount of a leukotriene A4 hydrolase (LTA4H) inhibitor. More particularly, the invention pertains to a method of treating or preventing a chronic liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of Formula (I):
Figure imgf000005_0001
wherein,
R1 is OH or NH2;
Y is O, S or CH2;
X1, X2, X3 and X4 are N; or
X1, X2, X3 and X4 are selected from N, NH, C, CH and O with the proviso that at least two of X1, X2, X3 and X4 are N or NH;
R2 is C1-C6 alkyl optionally substituted by phenyl; C3-C6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C1-C6 alkyl optionally substituted by halogen, C1-C6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable salt thereof.
Furthermore, the invention further provides a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor with one or more therapeutic agents, such as anti- steatotic agents, anti-inflammatory agents or anti-fibrotic agents.
In another aspect of the invention, the present invention provides a LTA4H inhibitor for use in the treatment and/or prevention of a liver disease, in a patient in need of such treatment and/or prevention. More particularly, the invention pertains to a compound of Formula (I); or a pharmaceutically acceptable salt thereof, as described herein for the use in the treatment or prevention of a chronic liver disease, in a patient in need of such treatment or prevention.
The invention further provides pharmaceutical combinations, comprising, separate or together, a LTA4H inhibitor and one or more additional therapeutic agents, for simultaneous, sequentially or separate administration. There is also provided a medicament, comprising such combinations for use in the treatment or prevention of liver diseases. More specifically, the liver disease is selected from non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis, bile duct obstruction, cholelithiasis and liver fibrosis.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 provides an illustrative XRPD spectrum for the crystalline form of (S)-3-amino-4-(5-(4- ((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form, designated herein as Form B, showing degrees 2q (2-theta) on the X-axis and relative intensity on the Y-axis.
Figure 2 provides an illustrative DSC spectrum for the free form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, designated herein as Form B.
Figure 3 provides an illustrative TGA spectrum for the free form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, designated herein as Form B.
Figure 4 shows normalized expression of the ALOX5 gene (5-Lipoxygenase enzyme) in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow.
Figure 5 shows normalized expression of the ALOX5AP gene (5-Lipoxygenase accessory protein, FLAP) in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow.
Figure 6 shows normalized expression of the LTA4H gene (Leukotriene A4 hydrolase) versus control gene TBP in all liver biopsies. Figure 7: Liver macrophages of liver biopsies of mice treated with two doses of (S)-3-amino-4- (5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid compared to vehicle treated mice and mice on normal chow by immunohistological analysis using the marker protein IBA1
Figure 8: Liver neutrophils of liver biopsies of mice treated with two doses of (S)-3-amino-4-(5- (4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid compared to vehicle treated mice and mice on normal chow by immunohistological analysis using the marker protein LY-6b.
Figure 9: Liver enzymes ALT and AST in blood of high fat, high fructose diet exposed mice treated with 3 mg/kg and 10 mg/kg of (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid versus vehicle control and mice on normal chow.
Figure 10: Inhibition of LTB4 release from ex-vivo stimulated blood in mice treated with 3mg/kg or 10mg/kg of (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid.
Figure 11 : Mean plasma concentration time profiles following single oral administration of (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different doses.
Figure 12: Mean plasma concentration time profile following multiple oral administration of (S)- 3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different doses (Figure 12a: Day 1 ; Figure 12b: Day 12)
Figure 13: LTB4 concentration in ex-vivo stimulated blood after oral administration of (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at measured plasma concentration of compound of example 1 in plasma (PK/PD relationship)
Figure 14: LTB4 change from baseline (inhibition in blood) after multiple oral administration of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Compound of example 1) at different dose measured at different time (days) since first dose.
More detailed listings of the XRPD peaks for Form B is set forth in Table 1 below, in which the % relative intensity (l/l0 x 100) is also provided. It should be understood that in the X- ray powder diffraction spectra or pattern that there is inherent variability in the values measured in degrees 2q (°2q) as a result of, for example, instrumental variation (including differences between instruments). As such, it should be understood that there is a variability of up to ± 0.2 °2q in XRPD peak measurements and yet such peak values would still be considered to be representative of a particular solid state form of the crystalline materials described herein. It should also be understood that other measured values from XRPD experiments and DSC/TGA experiments, such as relative intensity and water content, can vary as a result of, for example, sample preparation and/or storage and/or environmental conditions, and yet the measured values will still be considered to be representative of a particular solid state form of the crystalline materials described herein.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term “C1-C6 alkyl” refers to a fully saturated branched or unbranched hydrocarbon moiety having up to 6 carbon atoms. Unless otherwise provided, it refers to hydrocarbon moieties having 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso- propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl and the like.
As used herein, the term “C1-C6 alkoxy” refers to alkyl-O-, wherein alkyl is defined herein above. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, hexyloxy, cyclopropyloxy-, cyclohexyloxy- and the like. Typically, alkoxy groups have about 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms.
As used herein, the term “C1-C6 alkyl optionally substituted by halogen” refers to C1-C6 alkyl as defined above which may be substituted by one or more halogens. Examples include, but are not limited to, trifluoromethyl, difluoro methyl, fluoromethyl, trichloromethyl, 2,2,2- trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl and 1-bromomethyl-2-bromoethyl.
As used herein, the term "di-C1-6alkylamino" refers to a moiety of the formula -N(Ra)-Ra where each Ra is a C1-6alkyl, which may be the same or different, as defined above.
As used herein, the term "C3-C6 cycloalkyl" refers to saturated monocyclic hydrocarbon groups of 3-6 carbon atoms. Cycloalkyl may also be referred to as a carbocyclic ring and vice versa additionally referring to the number of carbon atoms present. Unless otherwise provided, cycloalkyl refers to cyclic hydrocarbon groups having between 3 and 6 ring carbon atoms or between 3 and 4 ring carbon atoms. Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. As used herein, the term “halogen” or “halo” refers to fluoro, chloro, bromo, and iodo.
As used herein, the term "heteroaryl" refers to a 5-14 membered monocyclic- or bicyclic- or tricyclic-aromatic ring system, having 1 to 8 heteroatoms. Typically, the heteroaryl is a 5-10 membered ring system containing 1 to 4 hereroatoms selected from N, S or O ( e.g ., 5-7 membered monocycle or an 8-10 membered bicycle) or a 5-7 membered ring system. Preferably, the term “heteroaryl” is a 5-7 membered monocycle. Typical heteroaryl groups include 2- or 3-thienyl, 2- or 3-furyl, 2- or 3-pyrrolyl, 2-, 4-, or 5-imidazolyl, 3-, 4-, or 5- pyrazolyl, 2-, 4-, or 5-thiazolyl, 3-, 4-, or 5-isothiazolyl, 2-, 4-, or 5-oxazolyl, 3-, 4-, or 5-isoxazolyl, 3- or 5- 1,2,4-triazolyl, 4- or 5-1 , 2, 3-triazolyl, tetrazolyl, 2-, 3-, or 4-pyridyl, 3- or 4-pyridazinyl, 3-, 4-, or 5-pyrazinyl, 2-pyrazinyl, and 2-, 4-, or 5-pyrimidinyl.
The term “heteroaryl” also refers to a group in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Non limiting examples include indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, quinolizinyl, quinoliyl, isoquinoliyl, benzisoqinolinyl, thieno[2,3-b]furanyl, furo[3,2-b]-pyranyl, pyrido[2,3-d]-o-oxazinyl, pyrazolo[4,3-d]-oxazolyl, imidazo[4,5-d] thiazolyl, 3pyrazino[2,3-d]pyridazinyl, imidazo[2,1-b] thiazolyl, benzo[b]thienyl, benzimidazolyl, and benzothiazolyl. Typical fused heteroaryl groups include, but are not limited to quinolinyl, isoquinolinyl, indolyl, benzo[b]thienyl, benzoxazolyl, benzimidazolyl, and -benzothiazolyl.
A substituted heteroaryl is a heteroaryl group containing one or more substituents.
As used herein, the term “heterocyclyl” refers to a heterocyclic group that is saturated or partially saturated and is preferably a monocyclic or a polycyclic ring (in case of a polycyclic ring particularly a bicyclic, tricyclic or spirocyclic ring); and has 3 to 24, more preferably 4 to 16, most preferably 5 to 10, and most preferably 5 or 6 ring atoms; wherein one or more, preferably one to four, especially one or two ring atoms are a heteroatom (the remaining ring atoms therefore being carbon). The bonding ring (i.e. the ring connecting to the molecule) preferably has 4 to 12, especially 5 to 7 ring atoms. The term heterocyclyl excludes heteroaryl. The heterocyclic group can be attached at a heteroatom or a carbon atom. The heterocyclyl can include fused or bridged rings as well as spirocyclic rings. Examples of heterocycles include tetrahydrofuran (THF), dihydrofuran, 1,4-dioxane, morpholine, 1,4-dithiane, piperazine, piperidine, 1,3- dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane, dithiolane, 1,3-dioxane, 1 ,3-dithiane, oxathiane, thiomorpholine, and the like.
A substituted heterocyclyl is a heterocyclyl group independently substituted by 1-4, such as one, or two, or three, or four substituents. As used herein, the term "aryl" refers to an aromatic hydrocarbon group having 6-20 carbon atoms in the ring portion. Typically, aryl is monocyclic, bicyclic or tricyclic aryl having 6- 20 carbon atoms. Furthermore, the term "aryl” as used herein, refers to an aromatic substituent which can be a single aromatic ring, or multiple aromatic rings that are fused together. Non- limiting examples include phenyl, naphthyl or tetrahydronaphthyl.
A substituted aryl is an aryl group substituted by 1-5 (such as one, or two, or three) substituents independently selected from the group consisting of hydroxyl, thiol, cyano, nitro, C1-C4-alkyl, C1- C4-alkenyl, C1-C4-alkynyl, C1-C4-alkoxy, C1-C4-thioalkyl, C1-C4-alkenyloxy, C1-C4-alkynyloxy, halogen, C1-C4-alkylcarbonyl, carboxy, C1-C4-alkoxycarbonyl, amino, C1-C4-alkylamino, di- C1- C4-alkylamino, C1-C4-alkylaminocarbonyl, di- C1-C4-alkylaminocarbonyl, C1-C4- alkylcarbonylamino, C1-C4-alkylcarbonyl(C1-C4-alkyl)amino, sulfonyl, sulfamoyl, alkylsulfamoyl, C1-C4-alkylaminosulfonyl, where each of the afore-mentioned hydrocarbon groups ( e.g ., alkyl, alkenyl, alkynyl, alkoxy residues) may be further substituted by one or more residues independently selected at each occurrence from halogen, hydroxyl or C1-C4-alkoxy groups.
As used herein, the terms “about” and “substantially” indicate with respect to features such as endotherms, endothermic peak, exotherms, baseline shifts, etc., that their values can vary. With reference to X-ray diffraction peak positions, “about” or “substantially” means that typical peak position and intensity variability are taken into account. For example, one skilled in the art will appreciate that the peak positions (2q) will show some inter-apparatus variability, typically as much as 0.2°. Occasionally, the variability could be higher than 0.2° depending on apparatus calibration differences. Further, one skilled in the art will appreciate that relative peak intensities will show inter-apparatus variability as well as variability due to degree of crystallinity, preferred orientation, prepared sample surface, and other factors known to those skilled in the art, and should be taken as qualitative measure only. For DSC, variation in the temperatures observed will depend upon the rate of temperature change as well as sample preparation technique and the particular instrument employed. Thus, the endotherm/melting point values reported herein relating to DSC/TGA thermograms can vary ± 5°C (and still be considered to be characteristic of the particular crystalline form described herein). When used in the context of other features, such as, for example, percent by weight (% by weight), reaction temperatures, the term “about” indicates a variance of ± 5%.
As used herein, “substantially phase pure,” when used in reference to any crystalline form of the compound of Formula I, means a compound having a phase purity of greater than about 90% by weight, including greater than about 91 , 92, 93, 94, 95, 96, 97, 98, and about 99% by weight, and also including equal to about 100% by weight of the compound of Formula (I), based on the weight of the compound on an anhydrous basis. The term “phase pure” or “phase purity” herein refers to phase homogeneity with respect to a particular solid state form of the compound of Formula (I), and does not necessarily imply a high degree of chemical purity absent an express statement to that effect. Phase purity may be determined according to methods known in the art, for example, using XRPD to do quantitative phase analysis using one or more approaches known in the art, for example, via an external standard method, direct comparisons of line (peak) characteristics which are attributed to different phases in a particular spectra, or via an internal standard method. However XRPD quantification of phase purity can be complicated by the presence of amorphous material. Accordingly, other methods that may be useful for determining phase purity include, for example, solid state NMR spectroscopy, Raman and/or infrared spectroscopy. One of skilled in the art would readily understand these methods and how to employ these additional (or alternative) methods for determining phase purity.
As used herein, the terms “salt” or “salts” refers to an acid addition or base addition salt of a compound for use in the method of the invention. “Salts” include, in particular, “pharmaceutically acceptable salts”. The term “pharmaceutically acceptable salts” refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable. In many cases, the compounds for use in the methods of the invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups, or groups similar thereto.
Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalacturonate, propionate, stearate, succinate, sulfosalicylate, tartrate, tosylate and trifluoroacetate salts.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
The pharmaceutically acceptable salts for use in the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable. Lists of additional suitable salts can be found, e.g., in “Remington's Pharmaceutical Sciences”, 20th ed., Mack Publishing Company, Easton, Pa., (1985); and in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, second revised edition Feb 2011).
Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds (i.e. LTA4H inhibitor as described herein) include isotopes of hydrogen, carbon, sulfur, nitrogen, oxygen, phosphorous, fluorine, iodine and chlorine, such as 2H, 3H, 11C, 13C, 14C, 15N, 18F 31P, 32P, 35S, 36CI, 125l respectively. The invention includes various isotopically labeled compounds as defined herein, for example, those into which radioactive isotopes, such as 3H and 14C, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically labeled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, a 18F or labeled compound may be particularly desirable for PET or SPECT studies. Isotopically-labeled compounds of Formula (I), or a pharmaceutically acceptable salt thereof, can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index. It is understood that deuterium in this context is regarded as a substituent of a compound of the Formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by the isotopic enrichment factor. The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
Pharmaceutically acceptable solvates for use in the method of the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D2O, d6-acetone, d6- DMSO (dimethylsulfoxide).
Compounds for use in the method of the invention, i.e. compounds of Formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers. These co-crystals may be prepared from compounds of Formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of Formula (I) with the co-crystal former under crystallization conditions and isolating co-crystals thereby formed. Suitable co-crystal formers include those described in WO 2004/078163. Hence, the invention further provides co-crystals comprising a compound of Formula (I) for use in the method of the present invention.
As used herein, the term “administering” in relation to a compound, e.g., an LTA4H inhibitor (e.g. a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a specific compound described herein), or another agent, is used to refer to delivery of that compound to a patient by any administration route.
As used herein, the term "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 20th Ed. Mack Printing Company, 1990, pp. 1289-1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
The term "a therapeutically effective amount" of a compound for use in the method of the invention refers to an amount of said compound that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms of liver disease, slow or delay disease progression of liver disease, or prevent liver disease. In one non-limiting embodiment, the term “a therapeutically effective amount” refers to the amount of the compound that, when administered to a subject, is effective to (1) at least partially alleviating, inhibiting, preventing and/or ameliorating liver disease. In another non- limiting embodiment, the term “a therapeutically effective amount” refers to the amount of the compound that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of LTA4H; or reducing or inhibiting the expression of LTA4H partially or completely.
As used herein, the term “subject” refers to an animal. Typically, the animal is a mammal. A subject also refers to, for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. The term “subject” is used interchangeably with “patient” when it refers to human.
As used herein, the phrase “population of patients” is used to mean a group of patients. In some embodiments of the disclosed methods, the LTA4H inhibitor (e.g., compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a compound disclosed in WO2015/092740 or any compound described herein) is used to treat a population of liver disease patients.
As used herein, “selecting” and “selected” in reference to a patient is used to mean that a particular patient is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria. Similarly, “selectively treating” refers to providing treatment to a patient having a particular disease, where that patient is specifically chosen from a larger group of patients on the basis of the particular patient having a predetermined criterion. Similarly, “selectively administering” refers to administering a drug to a patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criterion. By selecting, selectively treating and selectively administering, it is meant that a patient is delivered a personalized therapy based on the patient’s personal history (e.g., prior therapeutic interventions, e.g., prior treatment with biologies), biology (e.g., particular genetic markers), and/or manifestation (e.g., not fulfilling particular diagnostic criteria), rather than being delivered a standard treatment regimen based solely on the patient’s membership in a larger group. Selecting, in reference to a method of treatment as used herein, does not refer to fortuitous treatment of a patient having a particular criterion, but rather refers to the deliberate choice to administer treatment to a patient based on the patient having a particular criterion. Thus, selective treatment/administration differs from standard treatment/administration, which delivers a particular drug to all patients having a particular disease, regardless of their personal history, manifestations of disease, and/or biology. In some embodiments, the patient is selected for treatment based on having a liver disease.
As used herein, the term “inhibit”, "inhibition" or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
As used herein, the term “treat”, “treating" or "treatment" of any disease or disorder refers in one embodiment to ameliorating the disease or disorder (i.e. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms or pathological features thereof). In another embodiment “treat”, "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter or pathological features of the disease, e.g. including those which may not be discernible by the subject. In yet another embodiment, “treat”, "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g. stabilization of at least one discernible or non-discernible symptom), physiologically (e.g. stabilization of a physical parameter) or both. In yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the disease or disorder, or of at least one symptoms or pathological features associated thereof. In yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying progression of the disease to a more advanced stage or a more serious condition, such as e.g. liver cirrhosis; or to preventing or delaying a need for liver transplantation.
For example, treating NASH may refer to ameliorating, alleviating or modulating at least one of the symptoms or pathological features associated with NASH; e.g. hepatosteatosis, hepatocellular ballooning, hepatic inflammation and fibrosis; e.g. may refer to slowing progression, reducing or stopping at least one of the symptoms or pathological features associated with NASH, e.g. hepatosteatosis, hepatocellular ballooning, hepatic inflammation and fibrosis.
As used herein, the term “liver disease or disorder” encompasses one, a plurality, or all of non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), primary biliary cirrhosis (PBC), bile duct obstruction, cholelithiasis and liver fibrosis.
As used herein, the term NAFLD may encompass the different stages of the disease: hepatosteatosis, NASH, fibrosis and cirrhosis.
As used herein, the term NASH may encompass steatosis, hepatocellular ballooning and lobular inflammation.
As used herein, the term "prevention” refers delaying the onset or development or progression of the disease or disorder. More specifically, the term “preventing” the liver disease refers preventing or delaying liver cirrhosis or a need for liver transplantation.
As used herein, a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
As used herein, the term "a,” "an,” "the” and similar terms used in the context of the present invention (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context.
Any asymmetric atom (e.g., carbon or the like) of the compound(s) for use in the method of the invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R)- or (S)- configuration. Substituents at atoms with unsaturated double bonds may, if possible, be present in cis- (Z)- or trans- (E)- form.
Accordingly, a compound for use in the method of the present invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof. For greater clarity, the term “possible isomers” shall not include positional isomers.
Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
Any resulting racemates of final products or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound. In particular, a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di- 0,0'-p- toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid. Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral stationary phase.
Furthermore, the compounds for use in the method of the invention, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization. The compounds for use in the method of the invention may by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embraces the use of compounds as described herein, both solvated and unsolvated forms. The term "solvate" refers to a molecular complex of a compound for use in the method of the invention (including pharmaceutically acceptable salts thereof) with one or more solvent molecules. Such solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like. The term "hydrate" refers to the complex where the solvent molecule is water.
The compounds for use in the method of the present invention, include salts, hydrates, solvates and polymorph thereof. As herein defined, “combination” refers to either a fixed combination in one unit dosage form (e.g., capsule, tablet, or sachet), free (i.e. non-fixed) combination, or a kit of parts for the combined administration where a LTA4H inhibitor of the present invention and one or more “combination partner” (i.e. the additional therapeutic agent, also referred to as or “co-agent”) may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
The terms “co-administration” or “combined administration” or the like as utilized herein are meant to encompass administration of the additional therapeutic agent to a single subject in need thereof (e.g. a patient), and the additional therapeutic agent are intended to include treatment regimens in which the LTA4H inhibitor and the additional therapeutic agent are not necessarily administered by the same route of administration and/or at the same time. Each of the components of the combination of the present invention may be administered simultaneously or sequentially and in any order. Co-administration comprises simultaneous, sequential, overlapping, interval, continuous administrations and any combination thereof.
The term “pharmaceutical combination” as used herein means a pharmaceutical composition that results from the combining (e.g. mixing) of more than one active ingredient and includes both fixed and free combinations of the active ingredients.
The term “fixed combination” means that the active ingredients, i.e. i) a LTA4H inhibitor, e.g. Compound of any one of Formulae (I) to (V) as defined herein, or a compound according to any one of the embodiments below (in free form or e.g. as a pharmaceutically acceptable salt) and ii) the additional therapeutic agent (as herein defined), are both administered to a patient simultaneously in the form of a single entity or dosage.
The term “free combination” means that the active ingredients as herein defined are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, and in any order, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The term “free combination” is sometimes also referred to as “non fixed combinations”.
By "simultaneous administration", it is meant that the LTA4H inhibitor and the additional therapeutic agent (as herein defined), are administered on the same day. The two active ingredients can be administered at the same time (e.g. for fixed or free combinations) or one at a time (e.g. for free combinations).
According to the invention, "sequential administration", may mean that during a period of two or more days of continuous co-administration only one of the LTA4H inhibitor and the additional therapeutic agent is administered on any given day.
By "overlapping administration", it is meant that during a period of two or more days of continuous co-administration, there is at least one day of simultaneous administration and at least one day when only one of LTA4H inhibitor and the additional therapeutic agent, e.g., is administered.
By "interval administration", it is meant a period of co-administration with at least one void day, i.e with at least one day where neither the LTA4H inhibitor nor the additional therapeutic agent is administered.
By "continuous administration", it is meant a period of co-administration without any void day. The continuous administration may be simultaneous, sequential, or overlapping, as described above.
LTA4H inhibitors for use in the method of the invention
In embodiment one, the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor.
In embodiment 1A, the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor as described in WO2014/164658.
In embodiment 1 B, the present invention relates to a method of treating or preventing a liver disease, e.g. NAFLD, NASH, liver fibrosis and PBC, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor which is 4-(((1S,4S)-5-(4-(4-(oxazol-2-yl)phenoxy)benzyl)-2,5-diazabicyclo[2.2.1]heptan-2- yl)methyl)benzoic acid, or represented by:
Figure imgf000020_0001
pharmaceutically acceptable salt thereof.
In embodiment 1C, the present invention relates to a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a LTA4H inhibitor which is Acebilustat, also known as CTX 4430.
In embodiment 2, the present invention relates to a method of treating or preventing a liver disease in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000020_0002
wherein,
R1 is OH or NH2; Y is O, S or CH2;
X1, X2, X3 and X4 are N; or
X1, X2, X3 and X4 are selected from N, NH, C, CH and O with the proviso that at least two of X1, X2, X3 and X4 are N or NH; R2 is C1-C6 alkyl optionally substituted by phenyl; C3-C6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C1-C6 alkyl optionally substituted by halogen, C1-C6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable salt thereof.
Embodiment 2A relates to a method according to embodiment 2, comprising administering to the subject in need thereof, a therapeutically effective amount of a compound of formula (II), or a pharmaceutically acceptable salt thereof,
Figure imgf000021_0001
wherein the variables R1, R2 and Y have the meaning as defined in embodiment 2.
Embodiment 2B relates to a method according to embodiment 2, comprising administering to the subject in need thereof, a therapeutically effective amount of a compound of formula (III) or a pharmaceutically acceptable salt thereof,
Figure imgf000021_0002
wherein the variables R1, R2 and Y have the meaning as defined in embodiment 2.
Embodiment 2C relates to a method according to embodiment 2 comprising administering to the subject a therapeutically effective amount of a compound of formula (IV), or a pharmaceutically acceptable salt thereof,
Figure imgf000022_0001
wherein the variables R1, R2 and Y have the meaning as defined in embodiment 2.
Embodiment 2D relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of formula (V), or a pharmaceutically acceptable salt thereof;
Figure imgf000022_0002
wherein the variables R1, R2 and Y have the meaning as defined in embodiment 2.
Embodiment 2E of the present invention relates to the method according to any one of embodiments 2 and 2A to 2D, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is O; and
R2 is phenyl optionally being substituted by halogen, cyano, C1-C6alkyl optionally substituted by halogen, C1-C6alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or
R2 is a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen.
In a preferred aspect of embodiment 2E, the present invention relates to the method according to embodiment 2E, comprising administering to the subject a therapeutically effective amount of a compound of Formula (V), or a pharmaceutically acceptable salt thereof, wherein
R1 is H, Y is O and R2 is phenyl optionally substituted by one or two halogen.
Embodiment 2F of the present invention relates to the method according to any one of embodiments 2 and 2A to 2D, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is CH2; and
R2 is phenyl optionally being substituted by halogen, cyano, C1-C6alkyl optionally substituted by halogen, C1-C6alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or
R2 is a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by cyano, halogen or C1-C6alkyl optionally substituted by halogen.
Embodiment 2G of the present invention relates to a method according to any one of embodiments 2 and 2A to 2D, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein Y is O; and
R2 is C1-C6 alkyl optionally substituted by phenyl; or C3-C6cycloalkyl.
Embodiment 2H of the present invention relates to a method according to any one of embodiment 2 and 2A to 2D, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein Y is CH2; and
R2 is C1-C6alkyl optionally substituted by phenyl; or C3-C6cycloalkyl.
Embodiment 2I relates to a method according to any one of the embodiments 2 and 2A to 2H, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable salt thereof, wherein Y is attached to the para-position of the phenyl moiety.
Embodiment 2J relates to a method according to any one of the embodiments 2 and 2A to 2H, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein Y is attached to the meta-position of the phenyl moiety.
Embodiment 2K relates to a method according to any one of the embodiments 2 and 2A to 2J, comprising administering to the subject a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof, wherein R1 is OH.
Embodiment 2L relates to a method in accordance to any one of the embodiments 2 and 2A to 2K, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein the amino group on the beta-amino acid side chain has the (R)-configuration.
Embodiment 2M relates to a method in accordance to any one of the embodiments 2 and 2A to 2K, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I), (II), (III), (IV) or (V), or a pharmaceutically acceptable salt thereof; wherein the amino group on the beta-amino acid side chain has the (S)-configuration.
Embodiment 2N relates to a method according to embodiment 2, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof, wherein the compound is disclosed in WO2015/092740 [attorney docket number PAT056044-WO-PCT]; i.e. the compound is selected from: (R)-3-amino-4-(5-(4-(benzo[d]thiazol-2-yloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-((5-chloropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-(oxazol-2-yl)-phenoxy)phenyl)-2H-tetrazol-2-yl)-butanoic acid; (R)-3-amino-4-(5-(3-(4-chlorophenoxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-fluorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(3-chloro-4-fluorophenoxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(p-tolyloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (S)-3-amino-4-(5-(3-phenoxyphenyl)-2H-tetrazol-2-yl)butanoic acid; (S)-3-amino-4-(5-(4-(benzo[d]thiazol-2-yloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(3-phenethoxyphenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-phenethoxyphenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(benzyloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(3-(benzyloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-butoxyphenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(pentyloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(3-((5-(trifluoromethyl)pyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-((5-(trifluoromethyl)pyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(3-(benzo[d]thiazol-2-yloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(3-(3,5-difluorophenoxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (S)-3-amino-4-(5-(4-(p-tolyloxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-fluorophenoxy) phenyl)-1,3,4-oxadiazol-2-yl)butanoic acid; (R)-3-amino-4-(5-(4-(4-chlorophenoxy) phenyl)-1,3,4-oxadiazol-2-yl)butanoic acid; (R)-3-amino-4-(3-(4-(4-chlorophenoxy)phenyl)-1,2,4-oxadiazol-5-yl)butanoic acid; (R)-3-amino-4-(3-(4-(4-chlorophenoxy)phenyl)-1,2,4-oxadiazol-5-yl)butanamide; (S)-3-amino-4-(4-(4-(4-chlorophenoxy)phenyl)-1H-pyrazol-1-yl)butanoic acid; and (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
In embodiment 3, the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) wherein the compound is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid; or a pharmaceutically acceptable salt thereof. In embodiment 3A, the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (R)-3-amino-4-(5-(4-phenethoxyphenyl)-2H-tetrazol-2- yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
In embodiment 3B, the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (R)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid; or a pharmaceutically acceptable salt thereof.
In embodiment 3C, the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H- tetrazol-2-yl)butanoic acid (compound 2); or a pharmaceutically acceptable salt thereof.
The compounds of any one of Formulae (I) to (V), or a pharmaceutically acceptable salt thereof, and the compounds according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3C, or a pharmaceutically acceptable salt thereof, for use in the method of the invention are disclosed in WO2015/092740, which is incorporated by reference herein.
In embodiment 3D, the invention relates to a method according to embodiment 2, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) wherein the compound is the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form (i.e. non-salt form); or a pharmaceutically acceptable salt thereof.
In embodiment 3E, the invention relates to the method according to embodiment 3D, wherein the crystalline form is characterized by at least one of the following characteristics:
(i) an x-ray powder diffraction pattern comprising representative peaks in terms of 2q at 22.6 ± 0.2 °2q, 24.1 ± 0.2 °2q and 26.3 ± 0.2 °2q, measured at a temperature of about 25°C and an x- ray wavelength, l, of 1 .5418 Å;
(ii) an x-ray powder diffraction pattern comprising four or more 2q values selected from the group consisting of 11 .3 ± 0.2 °2q, 12.8 ± 0.2 °2q, 15.2 ± 0.2 °2q, 19.7 ± 0.2 °2q, 20.0 ± 0.2 °2q,
20.3 ± 0.2 °2q, 21 .0 ± 0.2 °2q, 22.6 ± 0.2 °2q, 24.1 ± 0.2 °2q, 24.4 ± 0.2 °2q, 25.1 ± 0.2 °2q,
26.3 ± 0.2 °2q, 28.5 ± 0.2 °2q, and 30.0 ± 0.2 °2q, measured at a temperature of about 25°C and an x-ray wavelength, l, of 1 .5418 Å; (iii) an x-ray powder diffraction pattern comprising five or more 2q values selected from the group consisting of 11 .3 ± 0.2 °2q, 12.8 ± 0.2 °2q, 15.2 ± 0.2 °2q, 19.7 ± 0.2 °2q, 20.0 ± 0.2 °2q,
20.3 ± 0.2 °2q, 21 .0 ± 0.2 °2q, 22.6 ± 0.2 °2q, 24.1 ± 0.2 °2q, 24.4 ± 0.2 °2q, 25.1 ± 0.2 °2q,
26.3 ± 0.2 °2q, 28.5 ± 0.2 °2q, and 30.0 ± 0.2 °2q, measured at a temperature of about 25°C and an x-ray wavelength, l, of 1 .5418 Å;
(iv) an x-ray diffraction spectrum substantially the same as the x-ray powder diffraction spectrum shown in FIG. 1 ;
(v) a differential scanning calorimetry (DSC) thermogram substantially the same as that shown in FIG. 2; (Tonset = 197.4°C) and
(vi) a thermo gravimetric analysis (TGA) diagram substantially the same as that shown in FIG.
3. (the weight loss by TGA is about 0.32% at 150°C)
The crystalline form of embodiment 3D and 3E is disclosed in PAT058189-WO-PCT, which is PCT/CN2018/000278, filed on July 31st 2018, which is incorporated by reference herein.
Combination partners
According to the invention, a compound of any one of Formulae (l)-(V), or a pharmaceutically acceptable salt thereof, or a compound according to any one of embodiments 1 , 2, 2A-2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof, can be administered as a monotherapy of in combination with one or more additional therapeutic agent (e.g. an anti- steatotic agent, and anti-inflammatory agent, and anti-fibrotic agent or combination thereof). Accordingly, the combination partner (additional therapeutic agent) of the invention can be a farnesoid X receptor (FXR) agonist, e.g. compound of Formula A described below; or a pharmaceutically acceptable salt thereof.
The bile acid receptor, farnesoid X receptor (FXR), is a member of the nuclear hormone receptor superfamily, primarily expressed in liver, intestine and kidney. FXR acts as a sensor of elevated bile acids and initiates homeostatic responses to control bile acid levels and modulate other metabolic processes such as gluconeogenesis and lipogenesis (Pattni et al., clin Trans Gastroenterol, 2012, 3: e18; Walters et al., Aliment Pharmacol Ther. 2015, 41 (1):54-64). FXR agonism modulates bile acid synthesis and detoxifying metabolism. In the setting of NASH, FXR agonism in both the liver and the gut has the potential to provide multimodal benefits which, in brief, include: reduction of fat accumulation in liver via decreased de novo lipogenesis, reduction of hepatic inflammation, anti-fibrotic effects mediated by stellate cells via decreased oxidative stress, and reduction of bacterial and lipopolysaccharide translocation from gut which induces inflammation (Schapp et al., Nat Rev Gastroenterol Hepatol, 2014, 11 (1):55-67).
Furthermore, Clinical validation of a FXR agonist for the treatment of NASH has been shown in clinical trials with obeticholic acid (OCA), a semi-synthetic variant of the natural bile acid chenodeoxycholic acid. In a small study in 20 patients with NAFLD and type 2 diabetes mellitus in which OCA was given for 6 weeks, OCA 25 mg improved insulin sensitivity and reduced circulating alanine aminotransferase (ALT) concentrations (Mudaliar et al. Gastroenterology 2013, 145(3):574-82). In a larger trial (n=219), it was shown that 45% of NASH patients receiving 25 mg OCA QD (once a day) for 72 weeks had improved liver histology compared to 23% of NASH patients receiving placebo in the same period (Neuschwander-Tetri et al., Lancet 2015, 385(9972):956-65). Finally, preliminary data from the ongoing clinical study CLJN452A2202 in patients with NASH indicates that the FXR agonist,
T ropifexor 90 pg daily for 12 weeks improved liver fat and ALT versus placebo.
Therefore, LTA4H inhibitor provides inflammatory effect in addition to those of a FXR agonist (such as Tropifexor) via inhibition of inflammation including hepatic neutrophil and macrophage infiltration and enhanced expression of the pro-resolving factor Lipoxin A4, which results in additional benefits in the treatment of liver disease.
Suitable FXR agonists for use in the combination include, but are not limited to, obeticholic acid (so called OCA, Intercept), GS9674, elafibranor (GFT505), GW4064, UPF987, FXR-450, fexaramine, methylcolate, methyl deoxycholate, 5b-cholanic acid, 5b-chloanic acid 7a, 12a diol, NIHS700, marchantin A, marchantin E, MFA-1 INT767 (also called 6a-ethyl-CDCA disclosed in WO2014/085474), MET409 (Metacrine), EDP-305 (Enanta), 2-[(1R,3r,5S)-3-({5- cyclopropyl-3-[2-(trifluoromethoxy)phenyl]-1 ,2-oxazol-4-yl}methoxy)-8-azabicyclo[3.2.1]octan-8- yl]-4-fluoro-1 ,3-benzothiazole-6-carboxylic acid (also known under the name Tropifexor), or a pharmaceutically acceptable salt thereof, or a compound disclosed in WO 2012/087519, or a compound disclosed in WO 2015/069666.
Preferably, the FXR agonist for use in combination with a LTA4H inhibitor, is a compound disclosed in patent application No. WO2012/087519, or a pharmaceutically acceptable salt thereof. More preferably, the FXR agonist for use in the combination with a LTA4H inhibitor is disclosed in WO2012/087519 and is 2-[3-({5-cyclopropyl-3-[2- (trifluoromethoxy)phenyl]-1,2-oxazol-4-yl}methoxy)-8-azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3- benzothiazole-6-carboxylic acid, a stereoisomer, an enantiomer, a pharmaceutically acceptable salt, solvate, prodrug, ester thereof and/or an amino acid conjugate thereof. In one aspect, the FXR agonist for use in the combination is 2-[(1R,3r,5S)-3-({5-cyclopropyl-3-[2- (trifluoromethoxy)phenyl]-1,2-oxazol-4-yl}methoxy)-8-azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3- benzothiazole-6-carboxylic acid which is also represented by Formula (A):
Figure imgf000029_0001
In yet a preferred embodiment, the FXR agonist for use in the combination is 2-[(1 R, 3r, 5S)-3-({5-cyclopropyl-3-[2-(trifluoromethoxy)phenyl]-1 ,2-oxazol-4-yl}methoxy)-8- azabicyclo[3.2.1]octan-8-yl]-4-fluoro-1 ,3-benzothiazole-6-carboxylic acid in its free form which is also known as Tropifexor or LJN452.
In another embodiment, the present invention relates to a pharmaceutical combination comprising a therapeutically effective amount of a compound of any one of Formulae (l)-(V), or a compound according to any one of embodiment 1 , 2, 2A-2N, 3, and 3A-3E, or a pharmaceutically acceptable salt thereof, and a FXR agonist, separately or together. In one aspect of this embodiment the invention relates to a pharmaceutical combination comprising a therapeutically effective amount of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof and Tropifexor. In one aspect of the previous embodiment, the pharmaceutical combination is a fixed combination. In another aspect of the previous embodiment, the pharmaceutical combination is a non-fixed combination.
Modes of administration
The pharmaceutical composition of the invention can be formulated to be compatible with its intended route of administration (e.g. oral compositions generally include an inert diluent or an edible carrier). Other non-limiting examples of routes of administration include parenteral (e.g. intravenous), intradermal, subcutaneous, oral (e.g. inhalation), transdermal (topical), transmucosal, and rectal administration. The pharmaceutical compositions compatible with each intended route are well known in the art. In a preferred embodiment, the pharmaceutical composition of the invention is formulated to be compatible with an oral administration.
In one embodiment, the pharmaceutical composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the prevention or the treatment of liver disease, can be in unit dosage of about 1 -500 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1 -250 mg or about 1 -150 mg or about 1 -100 mg, or about 1 -50 mg of active ingredients. The therapeutically effective dosage of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, the pharmaceutical composition is dependent on the species of the subject, the body weight, age and individual condition, the severity of the contrast-induced nephropathy disorder. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
Preferred formulation is a capsule or a tablet composition comprising from about 1 mg to about 160mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant. In a preferred embodiment, the capsule or tablet composition comprises from about 5 mg to about 80mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant. In yet another embodiment , the capsule or tablet composition comprises about 1 mg, about 5mg, about 10mg, about 20mg, about 30mg, about 40mg and about 50mg of a LTA4H inhibitor or of a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof, and one or more excipients independently selected from fillers, desintegrants, binders, and optionally lubricant and glidant.
Diseases
As hereinabove defined, the fibrotic or cirrhotic disease and/or disorder can be a liver disease or disorder, e.g. as defined below herein, or renal fibrosis. As hereinabove defined, the liver diseases or disorders can be cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibrosis, dyslipidemia, atherosclerosis, diabetes, diabetic nephropathy, colitis, newborn jaundice, prevention of kernicterus, veno-occlusive disease, portal hypertension, metabolic syndrome, hypercholesterolemia, progressive fibrosis of the liver caused by any of the diseases above or by infectious hepatitis.
The liver diseases or disorders can also refer to liver transplantation.
In one embodiment of the invention, the pharmaceutical composition (as herein defined, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof) is for the treatment or prevention of a fibrotic disease or disorder, e.g. a liver disease or disorder, e.g. a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of PBC, NAFLD, NASH, drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis. In one embodiment of the invention, the pharmaceutical combination (as herein defined) is for the treatment or prevention of fibrosis, e.g. renal fibrosis, autoimmune hepatitis or liver fibrosis.
In another embodiment of the invention, the pharmaceutical combination (as herein defined) is for the treatment or prevention of a fibrotic disease or disorder, e.g. a liver disease or disorder, e.g. a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of PBC, NAFLD, NASH, drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis. In one embodiment of the invention, the pharmaceutical combination (as herein defined) is for the treatment or prevention of fibrosis, e.g. renal fibrosis, autoimmune hepatitis or liver fibrosis.
According to one embodiment of the invention, the liver diseases or disorders refer to NAFLD, e.g. any stages of NAFLD, e.g. any of steatosis, NASH, fibrosis and cirrhosis. In one embodiment of the invention, there is provided a pharmaceutical combination of the invention for the improvement of liver fibrosis without worsening of steatohepatitis
In another embodiment of the invention, there is provided a pharmaceutical combination of the invention for obtaining a complete resolution of steatohepatitis without worsening, e.g. improving, of liver fibrosis.
In another embodiment of the invention, there is provided a pharmaceutical combination of the invention for preventing or treating steatohepatitis and liver fibrosis.
In yet another embodiment of the invention, there is provided a pharmaceutical combination of the invention for reducing at least one of the features of the NAS score (Brunt et al. Hepatology, 2011 , 53(3):810-820), i.e. one of hepatosteatosis, hepatic inflammation and hepatocellular ballooning; e.g. at least two features of the NAS score, e.g. hepatosteatosis and hepatic inflammation, or hepatosteatosis and hepatocellular ballooning, or hepatocellular ballooning and hepatic inflammation.
In a further embodiment of the invention, there is provided a pharmaceutical combination of the invention for reducing at least one or two features of the NAS score and liver fibrosis, e.g. for reducing hepatic inflammation and liver fibrosis, or hepatosteatosis and liver fibrosis or hepatocellular ballooning and liver fibrosis.
In yet a further embodiment of the invention there is provided a pharmaceutical combination for treating or preventing, stage 3 fibrosis to stage 1 fibrosis, e.g. stage 3 and/or stage 2 and/or stage 1 fibrosis.
Dosing regimens
Depending on the patient general condition, the targeted disease or disorder and the stage of such disease or disorder, the dosing regimen, i.e. administered doses and/or frequency of each component of the pharmaceutical combination may vary.
The frequency of dosing of the the LTA4H inhibitor of the invention (e.g. compound of any one of Formulae (I) to (V), or a pharmaceutically acceptable salt thereof, or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof), and the additional therapeutic agent, e.g as a fixed dose combination, may be once per day, twice per day, three times per day, four times per day, or every two days, every three days, e.g. once a day. According to the invention, the LTA4H inhibitor (e.g. compound of any one of Formula (I) to (V) or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof) and the additional therapeutic agent may not be administered following the same regimen, i.e. may not be administered at the same frequency and/or duration and/or dosage, e.g. at the same frequency and/or dosage. This can be the case e.g. for free combinations. As one example, the LTA4H inhibitor can be administered twice a day and the additional therapeutic agent, e.g. Compound of Formula A (in free form or as a pharmaceutically acceptable salt, solvate, prodrug and/or ester thereof) once per day.
In one embodiment of the invention, the co-administration is carried out for at least one week, at least one month, at least 6 weeks, at least 12 weeks, at least three months, at least 6 months, at least one year. For example, the pharmaceutical combination of the invention is administered lifelong to the patient. The frequency of administration, and/or the doses of the LTA4H inhibitor and of the additional therapeutic agent, may vary during the whole period of administration.
During the treatment, there can be one or more periods of time, e.g. days, during which nor the LTA4H inhibitor of the invention neither the additional therapeutic agent, e.g. a Compound of Formula A, or a pharmaceutically acceptable salt thereof, are administered to the patient (i.e. periods, e.g. days, void of combination treatment), or during which only one drug amongst the LTA4H inhibitor or the additional therapeutic agent is administered to the patient.
In case of a sequential co-administration, the LTA4H inhibitor may be administered prior the additional therapeutic agent, or reciprocally. The time interval between administration of the LTA4H inhibitor and of the additional therapeutic agent may vary from a few minutes to a few days, e.g. a few minutes, e.g. a few hours, e.g. 1 day to 1 week.
According to the invention, LTA4H inhibitor, (e.g. Compound of any one of Formula (I) to (V) or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof (as hereinabove defined, e.g. in free form, crystalline or amorphous form), is administered at a dose of about 1mg to about 160mg, preferably about 4mg to about 100mg; e.g. about 10mg to about 100mg, e.g. about 5mg to about 80mg, e.g. about 20mg to 60mg delivered orally; e.g. about 20mg, e.g about 30mg, e.g. about 40mg, e.g. about 80mg. Such doses may be for oral administration. Such doses may be for daily administration, or twice daily administration or every two days administration, e.g. for daily oral administration, twice daily oral administration, e.g. about 5mgQD to about 40mg BID (twice a day), e.g. about 10mg BID to about 30mg BID, e.g. about 10mg BID, or about 15mg BID, preferably about 20mg BID; or every two days oral administration. Dose selection for LTA4H inhibitor was determined from human safety and tolerability, pharmacokinetic and pharmacodynamic data obtained during first-in human clinical study.
According to the invention, the second therapeutic agent, i.e. the non-bile acid derived FXR agonist, e.g. Compound of Formula A (as hereinabove defined, e.g. in free form or as a pharmaceutically acceptable salt thereof), is administered at a dose of about 1 mg to about 250mg, e.g. about 5mg to about 150mg, e.g. about 10mg to about 150mg, e.g. about 20mg to 150mg, e.g. about 90mg to about 250mg; preferably about 140mg to about 200mg delivered orally, e.g. about 30mg, e.g. about 60mg, e.g. about 90mg, e.g. about 100mg, e.g about 120mg, e.g. about 140mg, e.g. about 150mg, e.g. about 160mg, e.g. about 170mg, e.g. about 180mg e.g. about 190, e.g. about 200mg, e.g. about 210mg, e.g. about 220mg, or e.g. 240mg. Such doses may be for oral administration. Such doses may be for daily administration, or twice daily administration or every two days administration, e.g. for daily oral administration, twice daily oral administration or every two days oral administration. Dose selection for FXR agonist was determined from human safety and tolerability, pharmacokinetic and pharmacodynamic data obtained during first-in human clinical study.
Methods of Treatment and Uses of LTA4H inhibitors for liver disease
Disclosed herein are methods of treating or preventing liver disease comprising orally administering to a patient in need thereof a dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg) of a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), daily. Said doses can be administered to the patient either with a once a day dosing regimen or twice a day dosing regimen. In another embodiment, the method comprises orally administering to a patient in need thereof a dose of about 10mg QD to about 40mg BID of a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof). In yet another embodiment, the method comprises orally administering to a patient in need thereof a dose of about 10mg to about 30 mg twice a day (e.g. about 10mg BID, about 15mg BID, preferably about 20mg BID) of a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof). In a preferred aspect of this embodiment, the method comprises orally administering a dose of about 20 mg of LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), to said patient twice a day (BID).
Disclosed herein are methods of treating or preventing liver diseases, comprising orally administering to a patient in need thereof a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg) of a LTA4H inhibitor, wherein the LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof. In another embodiment, the method comprises orally administering a dose of about 10mg QD to about 40mg BID of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2- yl)butanoic acid, or a pharmaceutically acceptable salt thereof, to said patient. In a preferred aspect of this embodiment, the method comprises orally administering a dose of about 10mg to about 30 mg twice a day (e.g. about 10mg BID, about 15mg BID, preferably about 20mg BID) of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, to said patient. In a most preferred aspect of this embodiment, the method comprises orally administering a dose of about 20mg of (S)-3-amino- 4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, to said patient twice a day.
Disclosed herein are methods of treating or preventing liver disease, comprising orally administering to a patient in need thereof a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg) of a LTA4H inhibitor, wherein the LTA4H inhibitor is a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form, as described herein. In another embodiment, the method comprises orally administering a dose of about 10mg QD to about 40mg BID of a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, as described herein, to said patient. In a preferred aspect of this embodiment, the method comprises orally administering a dose of about 10mg to about 30 mg twice daily (e.g. about 10mg BID, about 15mg BID, or preferably about 20mg BID) of a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H- tetrazol-2-yl)butanoic acid, as described herein, to said patient. In a most preferred aspect of this embodiment, the method comprises orally administering a dose of about 20mg of a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2- yl)butanoic acid, as described herein, to said patient twice a day.
Disclosed herein is a LTA4H inhibitor (i.e. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), for use in the treatment or prevention of a liver disease or disorder, wherein the LTA4H inhibitor is administered orally to a patient in need thereof in a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg). In another embodiment, LTA4H inhibitor (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof) is administered orally to a patient in need thereof in a dose of about 10mg QD to about 40mg BID. In a preferred aspect of this embodiment, LTA4H inhibitor (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof) is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day (e.g. about 10mg BID, or about 15mg BID, or preferably about 20mg BID). In yet a most preferred aspect of this embodiment, LTA4H inhibitor (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof) is administered orally to a patient in need thereof in a dose of about 20 mg twice a day.
Disclosed herein is a LTA4H inhibitor which is (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a liver disease or disorder, wherein the LTA4H is administered orally to a patient in need thereof a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg). In another embodiment, (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H- tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, is administered orally to a patient in need thereof in a dose of about 10mg QD to about 40mg BID. In a preferred aspect of this embodiment, (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H- tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day (e.g. about 10mg BID, about 15mg BID, about 20mg BID). In yet a most preferred embodiment, (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, is administered orally to a patient in need thereof in a dose of about 20mg twice a day.
Disclosed herein is a LTA4H inhibitor which is a crystalline form of (S)-3-amino-4-(5-(4- ((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, for use in the treatment or prevention of a liver disease or a disorder wherein the LTA4H is administered orally to a patient in need thereof in a daily dose of about 1 mg to about 160mg, or about 4mg to about 100mg, or about 10mg to about 100mg, or about 20mg to about 60mg or about 5mg to about 80mg (e.g. about 20mg, about 30mg, about 40mg or about 80mg). In another embodiment, the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, is administered orally to a patient in need thereof at a dose of about 10mg QD to about 40mg BID. In a preferred aspect of this embodiment, the crystalline form of (S)-3-amino-4-(5-(4-((5- chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, is administered orally to a patient in need thereof in a dose of about 10mg to about 30mg twice a day (e.g about 10mg BID, about 15mg BID, or preferably about 20mg BID). In yet a most preferred embodiment, the crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3- fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form as described herein, is administered orally to a patient in need thereof at a dose of about 20mg twice a day.
In preferred embodiments of the disclosed methods, uses and kits, the dose of the LTA4H inhibitor is about 10mg to about 30mg BID or about 20 mg BID.
In preferred embodiments of the disclosed methods, uses and kits, prior to treatment with a LTA4H inhibitor as disclosed herein, the patient has been previously treated with a systemic agent selected from obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
In some embodiments of the disclosed methods, uses and kits, prior to treatment with a LTA4H as described herein, the patient has not been previously treated with a systemic agent such as obeticholic acid, anti-diabetic agents, insulin, beta blockers, thiazide diuretics, fibrates, statins, niacin and ezetimibe.
In one embodiment of the disclosed methods, uses and kits, the LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), is administered in combination with at least one of an anti-steatotic agent, an anti-inflammatory agent and anti-fibrotic agent. In a particular aspect of this embodiment of the disclosed methods, uses and kits, the LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), is administered in combination with a FXR agonist. In a preferred aspect of this embodiment, the FXR agonist is T ropifexor.
In preferred embodiments of the disclosed methods, uses and kits, the dose of the LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), is about 10mg to about 30mg BID. In other preferred embodiments of the disclosed methods, uses and kits, the dose of the LTA4H inhibitor (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), is about 20 mg.
In preferred embodiments of the disclosed methods, uses and kits, the patient is a NAFLD patient with a biomarker phenotype consistent with ongoing liver inflammation and fibrosis such as a NASH-like phenotype as defined based on the presence of all five of the following:
• ALT ³ 43 IU/L (males) or ³ 28 IU/L (females) at screening; and
• BMI ³ 27 kg/m2 (in patients with a self-identified race other than Asian) or ³ 23 kg/m2 (in patients with a self-identified Asian race); and
• History of Type 2 diabetes mellitus, and
• ELF Test score ³ 8.5 and £ 10.5; and
• Liver fat ³ 8% at baseline as determined by the reading of the central MRI laboratory of locally produced images.
In preferred embodiments of the disclosed methods, uses and kits, the patient is an adult of 18 years and older. In some embodiments of the disclosed methods, uses and kits, the patient must weigh at least 40kg and no more than 150kg.
In preferred embodiments of the disclosed methods, uses and kits, prior to treatment with a LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test ³8.5 and £10.5.
In preferred embodiments of the disclosed methods, uses and kits, prior to treatment with a LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF) ³8%.
In preferred embodiments of the disclosed methods, uses and kits, prior to treatment with a LTA4H inhibitor as described herein (e.g. a compound according to any one of embodiments 1 , 2, 2A to 2N, 3, and 3A to 3E, or a pharmaceutically acceptable salt thereof), the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) ³43 IU/L for male and ³28 IU/L for female. In particular, the ALT does not exceed 5 times the upper limit of normal.
In preferred embodiments of the disclosed methods, uses and kits, the patient achieves a change from baseline as measured by ALT, percent liver fat (MRI-PDFF) or ELT Test by week 12 of treatment.
In preferred embodiments of the disclosed methods, uses and kits, the patient achieves at least a 3% decrease of ELF by week 12 of treatment. In a particular aspect of this embodiment, the patient further achieves at least a 10% reduction in ALT, and preferably at least 35% reduction in ALT at week 12 of treatment.
In another embodiment of the disclosed methods, uses and kits, the patient achieves at least a 3% decrease of ELF together with at least 10% reduction of ALT together with at least 10% reduction of percent liver fat by week 12 of treatment. In another embodiment of the disclosed methods, uses and kits, the patient achieves at least a 3% decrease of ELF together with at least 10% reduction of ALT together with at least 15% reduction of percent liver fat by week 12 of treatment. In a non-limiting example, the patient achieves a 3% reduction of ELF, a 10% reduction of percent liver fat and a 30% reduction of ALT by week 12 of treatment.
In another preferred embodiment of the disclosed methods, uses and kits, the patient achieves between 2% and <3% decrease of ELF together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat at week 12 of treatment. In another preferred embodiment of the disclosed methods, uses and kits, the patient achieves between 2% and <3% decrease of ELF together with a reduction of at least 35% ALT by week 12 of treatment. In a non-limiting example, the patient achieves by week 12: a 2.5% reduction of ELF, a 10% reduction of percent liver fat and a 40% reduction of ALT.
In another preferred embodiment of the disclosed methods, uses and kits, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat by week 12 of treatment. In one aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 30% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 35% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 35% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment. In a non-limiting example, the patient achieves an ELF reduction of 1 %, together with a 30% fat reduction and a 40% ALT reduction by week 12 of treatment.
In yet another embodiment of the disclosed methods, uses and kits, the patient achieve by week 12 of treatment one of the following: a) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or b) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or c) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or; d) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or e) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 35% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or f) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF).
In another preferred embodiment of the disclosed methods, uses and kits, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 10% of percent liver fat by week 12 of treatment. In one aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 15% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 30% of percent liver fat by week 12 of treatment. In yet another aspect of this embodiment, the patient achieves a decrease of ELF of less than 2%, together with a reduction of at least 10% ALT and a reduction of at least 35% of percent liver fat by week 12 of treatment. In a non-limiting example, the patient achieves an ELF reduction of 1 %, together with a 30% fat reduction and a 40% ALT reduction by week 12 of treatment.
In preferred embodiments of the disclosed methods, uses and kits, the patient is treated with a LTA4H inhibitor, as described herein, for at least 12 weeks, at least 16 weeks, at least 24 weeks, at least 48 weeks or at least 52 weeks. Most preferably, the patient is treated for at least 12 weeks.
In preferred embodiments of the disclosed methods, uses and kits, the LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
In preferred embodiments of the disclosure, the LTA4H inhibitor is a crystalline form of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid in its free form (herein designated as Form B).
In preferred embodiments, the LTA4H inhibitor is Form B as described in embodiment 3E, in a substantially pure phase.
In some embodiments, the invention relates to a pharmaceutical combination comprising a therapeutically effective amount of a compound of any one of Formulae (I)- (V) or a compound according to any one of embodiments 2, 2A to 2N, 3 and 3A to 3E, or a pharmaceutically acceptable salt thereof and a FXR agonist, for simultaneous, sequentially or separate administration. In a special aspect of this embodiment, the compound of Formula (I) is (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof, and the FXR agonist is tropifexor.
In some embodiments, the combination is a fixed dose combination.
In other embodiments, the combination is a free combination.
In one embodiment, the invention relates to a combination as described above for use in the treatment and prevention of a liver disease. In a specific aspect of this embodiment, the invention provides a combination for use in treating and preventing a liver disease, wherein the LTA4H inhibitor is administered at a dose in a range of about 10mg to about 100mg. In yet another aspect of this embodiment, the invention provides a combination for use in treating and preventing a liver disease, wherein the FXR agonist is administered at a dose in a range of about 1 mg to about 250mg.
Aspects, advantageous features and preferred embodiments of the present invention summarized in the following items, respectively alone or in combination, contribute to solving the object of the invention. Item 1 . A method of treating or preventing a liver disease, comprising administering to a subject in need thereof a therapeutically effective amount of a LTA4H inhibitor.
Item 2. The method according to item 1 , wherein the LTA4H inhibitor is a compound of
Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000043_0001
wherein,
R1 is OH or NH2;
Y is O, S or CH2;
X1, X2, X3 and X4 are N; or
X1, X2, X3 and X4 are selected from N, NH, C, CH and O with the proviso that at least two of X1, X2, X3 and X4 are N or NH;
R2 is C1-C6 alkyl optionally substituted by phenyl; C3-C6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C1-C6 alkyl optionally substituted by halogen, C1-C6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen; or a pharmaceutically acceptable salt thereof.
Item 3. The method according of item 1 or 2, wherein the LTA4H inhibitor is (S)-3-amino-
4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
Item 4. The method according to any of the above items, wherein the LTA4H inhibitor is disposed in a pharmaceutical formulation, wherein said pharmaceutical formulation comprises one or more pharmaceutically acceptable carriers, each of which is independently selected from a filler, a lubricant, a binder, a desintegrant and a glidant. Item 5. The method according to item 4, wherein the pharmaceutical formulation is in tablet or capsule form.
Item 6. The method according to any of the above items, wherein the LTA4H inhibitor is administered in combination with a FXR agonist, and wherein each of the components of the combination are administered simultaneously, or sequentially, or in any order.
Item 7. The method according to item 6, wherein the FXR agonist is Tropifexor.
Item 8. The method according to item 6 or 7 wherein the FXR agonist is administered at a dose range of about 1 mg to about 250mg.
Item 9. The method according to any of the above items, wherein the LTA4H inhibitor is administered at a dose of about 10 mg to about 30 mg twice a day.
Item 10. The method according to any of the above items, wherein the patient is a NAFLD patient selected according to at least one of the following criteria: a) prior to the treatment with LTA4H inhibitor, the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test ³8.5 and £10.5; b) prior to treatment with the LTA4H inhibitor, the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI- PDFF) ³8%; c) prior to treatment with the LTA4H inhibitor, the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) ³43 IU/L for male and ³28 IU/L for female.
Item 11 . The method according to any of the above items, wherein said patient achieves by week 12 of treatment one of the following: g) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or h) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or i) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or; j) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or k) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 35% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or
L) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF).
Item 12. The method according to any one of the above items wherein the liver disease or disorder is a chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibrosis, dyslipidemia, atherosclerosis, diabetes, diabetic nephropathy, colitis, newborn jaundice, prevention of kernicterus, veno-occlusive disease, portal hypertension, metabolic syndrome, hypercholesterolemia, progressive fibrosis of the liver caused by any of the diseases above or by infectious hepatitis; e.g. NAFLD, NASH, liver fibrosis, or PBC.
Item 13. A LTA4H inhibitor for use in the treatment and/or the prevention of a liver disease or disorder in a patient in need of such treatment and/or prevention.
Item 14. A LTA4H inhibitor for use according to item 13, wherein said LTA4H inhibitor is a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000046_0001
wherein,
R1 is OH or NH2;
Y is O, S or CH2;
X1, X2, X3 and X4 are N; or
X1, X2, X3 and X4 are selected from N, NH, C, CH and O with the proviso that at least two of X1, X2, X3 and X4 are N or NH;
R2 is C1-C6 alkyl optionally substituted by phenyl; C3-C6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C1-C6 alkyl optionally substituted by halogen, C1-C6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen. Item 15. A LTA4H inhibitor, for use according to item 13 or 14, wherein said LTA4H inhibitor is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
Item 16. A LTA4H inhibitor for use according to any one of items 13-15, wherein the LTA4H inhibitor is disposed in a pharmaceutical formulation, wherein said pharmaceutical formulation comprises one or more pharmaceutically acceptable carriers, each of which is independently selected from a filler, a lubricant, a binder, a desintegrant and a glidant.
Item 17. A LTA4H inhibitor for use according to item 16, wherein the pharmaceutical formulation is in tablet or capsule form.
Item 18. A pharmaceutical composition comprising a LTA4H inhibitor which is a compound of Formula (I), or a pharmaceutically acceptable salt thereof in accordance to item 14, together with one or more pharmaceutically acceptable carriers for use in the treatment and/or prevention of liver disease or disorder in a patient in need of such treatment and/or prevention.
Item 19. A pharmaceutical combination comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof according to item 14, and a FXR agonist.
Item 20. A pharmaceutical combination according to item 19, wherein the compound of Formula (I) is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof and the FXR agonist is Tropifexor.
Item 21 . A pharmaceutical combination according to item 19 or 20, for use in the treatment and/or prevention of a liver disease or disorder in simultaneous, sequentially or separate administration.
Item 22. A LTA4H inhibitor for use according to any one of items 13 to 17 or a pharmaceutical composition for use according to item 18, a pharmaceutical combination for use according to item 19-21 , wherein the LTA4H inhibitor is administered at a dose of about 10 mg to about 30mg twice a day. Item 23. A LTA4H inhibitor for use according to any one of items 13 to 17 or a pharmaceutical composition for use according to item 18, or a pharmaceutical combination for use according to item 19-21 , wherein the FXR agonist is administered at a dose in a range of about 1mg to about 250mg.
Item 24. A LTA4H inhibitor for use according to any one of items 13 to 17, 22 and 23, or a pharmaceutical composition for use according to any one of items 18, 22 and 23, or a pharmaceutical combination for use according to any one of items 19 to 23, wherein the patient is a NAFLD patient selected according to at least one of the following criteria: a) prior to the treatment with LTA4H inhibitor, the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test ³8.5 and £10.5; b) prior to treatment with the LTA4H inhibitor, the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI- PDFF) ³8%; c) prior to treatment with the LTA4H inhibitor, the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) ³43 IU/L for male and ³28 IU/L for female.
Item 25. A LTA4H inhibitor for use according to any one of items 13 to 17 and 22-24, or a pharmaceutical composition for use according to any one of items 18 and 22-24, or a pharmaceutical combination for use according to any one of items 19 to 24, wherein said patient achieves by week 12 of treatment at least one of the following: a) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or b) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or c) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or; d) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or e) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 35% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or f) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF).
Item 26. A LTA4H inhibitor for use according to any one of items 13 to 17 and 22-25, or a pharmaceutical composition for use according to any one of items 18 and 22-25, or a pharmaceutical combination for use according to any one of items 19 to 25, wherein a liver disease or disorder is selected from chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibrosis, dyslipidemia, atherosclerosis, diabetes, diabetic nephropathy, colitis, newborn jaundice, prevention of kernicterus, veno-occlusive disease, portal hypertension, metabolic syndrome, hypercholesterolemia, progressive fibrosis of the liver caused by any of the diseases above or by infectious hepatitis; e.g. NAFLD, NASH, liver fibrosis, or PBC. General
The details of one or more embodiments of the disclosure are set forth in the accompanying description above. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. Other features, objects, and advantages of the disclosure will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms include plural references unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents and publications cited in this specification are incorporated by reference. The following Examples are presented in order to more fully illustrate the preferred embodiments of the disclosure. These examples should in no way be construed as limiting the scope of the disclosed subject matter, as defined by the appended claims.
EXAMPLES
Example 1 : Crystalline form B of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid
Figure imgf000050_0001
Example 29 as described in WO2015/092740 (28g, 35 mmol) and a solvent mixture containing 360g water and 40g THF was mixed together and stirred for 20 minutes. The mixture was filtered and the filtrate was adjusted to pH = 5 with aqueous NaHCO3. The stirring was continued for 18h before the mixture was filtered to afford the free acid (lnt-1) in wet cake 25.6g, which was used for preparation of polymorph Form B without further purification.
505mg of the free acid (lnt-1) are weighed into a 20 mL glass vial and 6 ml. of methanol are added. The slurry is heated to 50°C and stirred for 4 days using a magnetic stirrer. The suspension is cooled to room temperature and filtered. The recovered solid is dried at 40°C for 2.5 h under vacuum. The white solid was analyzed by XRPD, DSC and TGA (Figures 1-3 respectively).
Alternative methods for making crystalline Form B are described in Patent application No. PCT/CN2018/000278, filed on July 31st 2018 (Attorney docket number PAT058189-WO-PCT).
Powder X-Ray Diffraction
X-ray powder diffraction (XRPD) patterns were obtained using a Bruker Discovery D8 in reflection geometry. Powders were analyzed using a zero background Si-wafer sample holder. The radiation was Cu Ka (I = 1.5418 Å). Patterns were measured between 2° and 40° 2theta.
Table 1
X-ray powder diffraction data for crystalline form B
Figure imgf000052_0001
DSC:
Differential scanning calorimetry was conducted for each crystalline form using a TA Instruments (DSC 2500). For each analysis, 2-4 mg of sample was placed in an aluminium T- zero crucible that closed with a pin-hole lid. The heating rate was 10°C per minute in the temperature range between 30 and 300°C. Temperatures are reported in degrees Celsius (°C) and enthalpies are reported in Joules per gram (J/g). Plots are showing endothermic peaks as down. The endothermic melt peak (melting point) was evaluated for extrapolated onset temperature. The accuracy of the measured sample temperature with this method is within about ±1 °C, and the heat of fusion can be measured within a relative error of about ±5%.
Illustrative DSC trace generated using crystalline Forms B is shown in Figures 2.
Form B: Melting endotherm: Tonset = 197.4° C (melting under decomposition)
Thermoaravimetric Analysis (TGA):
TGA curves were obtained using a TA-instrument Q5000. 5-15mg of sample was placed into an aluminum crucible and sealed hermetically. The sealed crucible was pierced by the robotic auto sampler immediately before analysis. The TGA curve was measured at 10°C/min between 30- 300°C. The LoD (Loss of drying) was calculated between 40°C and 150°C. The weight loss is plotted against the measured sample temperature. Temperatures are reported in degrees Celsius (°C) and weight loss in %.
Illustrative TGA trace generated using crystalline Forms B is shown in Figures 3.
Example 2:
Formulation of compound of Example 1 as a capsule
Figure imgf000053_0001
Figure imgf000054_0001
Example 3: Liver transcriptomic analysis of mice fed a high fat, high fructose NASH (HF/NASH) diet
C57BL/6J mice were fed a high fat, high fructose NASH (HF/NASH) diet up to 24 weeks to induce NASH like pathology in liver including steatosis , lobular inflammation and fibrosis accompanied by elevated liver injury enzymes, ALT and AST.
Transcriptomic analysis of 6-10 liver tissue of BL/6 mice taken at different time points after exposure to a HF/NASH diet versus normal control diet reveals a continuous upregulation of the expression of the 5-lipoxygenase pathway genes ALOX5 and ALOX5AP. Furthermore a strong expression of the LTA4H gene could be observed, expression of which, however, was not further elevated upon high fat, high fructose diet. ALOX5, ALOX5AP and LTA4H genes encode for the enzymatic cascade that is responsible for biosynthesis of the pro-inflammatory lipid mediator LTB4.
From snap frozen liver tissue a homogenate was prepared and RNA extracted. cDNA of the samples was prepared from the same starting amount of RNA using a high capacity cDNA reverse Transcription Kit (Applied Biosystems). 10 ng/ul of cDNA for each sample and qPCR was conducted using TaqMan technology. TBP (TATA-Binding Protein) was used as control gene. The changes of mRNA levels compared with untreated sample were calculated according to the manufacturer’s instructions (DCt).
Figure 4 shows normalized expression of the ALOX5 gene (5-Lipoxygenase enzyme). A significant upregulation of ALOX5 in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow could be observed as of week 12. Expression continued to rise until week 24. **** P<0.0001 ; *** P<0.001 ; ** P<0.01 ; * P<0.05, applying a two- tailed unpaired t-test.
Figure 5 shows normalized expression of the ALOX5AP gene (5-Lipoxygenase accessory protein, FLAP). A significant upregulation of ALOX5AP in 8-10 liver biopsies from mice treated with high fat, high fructose diet versus mice on normal chow could be observed as of week 12. Expression continued to rise until week 24. **** P<0.0001 ; *** P<0.001 ; ** P<0.01 ; * P<0.05, applying a two-tailed unpaired t-test.
Figure 6 shows normalized expression of the LTA4H gene (Leukotriene A4 hydrolase). A strong expression of LTA4H versus control gene TBP was observed in all liver biopsies. Expression was not different between mice on high fat, high fructose diet versus normal chow. **** P<0.0001 ; *** P<0.001 ; ** P<0.01 ; * P<0.05, applying a two-tailed unpaired t-test.
Example 3b: Treatment with (S)-3-amino-4-(5-(4-(4-chlorophenoxy)-phenyl)-2H-tetrazol-2- yl)butanoic acid (compound 2) attenuates the development of high fat, high fructose- induced NASH
At the age of 8 weeks, BL/6 mice were exposed for 20 weeks to a high fat, high fructose diet. Control mice were left on normal chow. 8 weeks after starting the high fat, high fructose NASH diet, mice were treated with 3 mg/kg/day or 10 mg/kg/day p.o. of the LTA4H inhibitor compound 2 or vehicle control until week 20. As compared to mice on normal chow, mice on high fat, high fructose diet showed substantially increased body weight, accompanied by histomorphological signs of NASH, such as liver steatosis, lobular liver inflammation and liver enzyme aberration. Mice treated with the LTA4H inhibitor compound 2 showed significantly reduced signs of liver inflammation, in particular reduced numbers of liver neutrophils and macrophages. Also elevation of liver enzymes ALT and AST was significantly reduced upon treatment with compound 2, indicating the protective effect of LTA4H inhibition in this diet-induced NASH model.
Figure 7: IBA1 positive liver tissue macrophages of mice treated with two doses of the LTA4H inhibitor compound 2 were compared to vehicle treated mice and mice on normal chow. The IBA1 (pan-macrophage marker)-stained area was quantified and showed a 38% and and 42% reduction in liver tissue macrophages in mice treated with 3mg/kg/ and10 mg/kg compound 2, respectively as compared to vehicle control. **p<0.01 ; ***p<0.001 ; ****p<0.0001 compared to vehicle control; one-way ANOVA with a post- hoc Dunnett's test.
Figure 8: LY6B positive liver tissue neutrophils of mice treated with two doses of the LTA4H inhibitor compound 2 were compared to vehicle treated mice and mice on normal chow. The LY- 6b stained area was quantified and showed a 42% and 49% reduction in liver tissue neutrophiles in mice treated with 3mg/kg and 10 mg/kg compound 2 as compared to vehicle control. *p<0.05 ; **p<0.01 ; ****p<0.0001 compared to vehicle control; one-way ANOVA with a post-hoc Dunnett's test.
Figure 9: Liver enzymes ALT and AST were determined in blood of mice fed a high fat, high fructose NASH diet and treated with 3 mg/kg and 10 mg/kg compound 2 versus vehicle control and mice on normal chow. AST blood levels were reduced by 31% and 45% with 3 mg/kg and 10 mg/kg compound 2, respectively. ALT level were reduced by 30 % with 3 mg/kg compound 2 and 46% with 10 mg/kg compound 2. Depicted are international units of enzyme per liter blood with standard deviation. *p< 0.05; **p<0.01 ; ***p<0.001 ; ****p<0.0001 compared to vehicle control; one-way ANOVA with a post-hoc Dunnett's test. Figure 10: Mice were treated with either vehicle, 3 mg/kg or 10 mg/kg compound 2. 22 hours after the last dose, blood was obtained, diluted 1 :3 and stimulated for 15 min ex-vivo with 10 mg/ml calcium ionophore to induce LTB4 production. Release of LTB4 was quantified via LTB4 EIA and demonstrated over 90% inhibition of LTB4 in both treated groups. Depicted are averages of 4 mice per group +/- standard error of the mean.
Example 4: Clinical data from first in Human (FIH) trial
Compound of example 1 has been studied in a FIH study designed to characterize its preliminary safety, tolerability, and PK in adult healthy subjects. The study consisted of a part 1 , single ascending dose (SAD) and a part 2 multiple ascending dose (MAD). Compound of example 1 was administered orally in single either QD or BID, in fed or fasted conditions over a dose range of 5 mg to 2 times 100 mg. Compound of example 1 was also administered orally in multiple-dose administration, in fasted conditions for 12 days over a dose range of 5 mg QD to 80 mg BID.
Part 1 (SAD)
Part 1 was a randomized, subject-blinded and Investigator-partial-blinded, placebo-controlled, single ascending oral dose study. The dose was administered either in one take or split into two intakes at 12 hours apart (N =69). Eight subjects were randomized into each cohort (except in Cohort 7 where seven subjects were randomized and in Cohort 9 where six subjects were randomized) to receive either compound of example 1 or matching placebo in a 6:2 ratio (active: placebo), testing nine dose levels.
Subjects were assigned to one of the following cohort:
Cohort 1 : Single oral dose of 5 mg or matching placebo Cohort 2: Single oral dose of 10 mg or matching placebo Cohort 3: Single oral dose of 20 mg or matching placebo Cohort 4: Single oral dose of 30 mg or matching placebo Cohort 5: Single oral dose of 45 mg or matching placebo Cohort 6: Single oral dose of 70 mg or matching placebo
Cohort 7: total oral dose of 140 mg or matching placebo split into two doses of 70 mg taken 12h apart
Cohort 8: total oral dose of 200 mg or matching placebo split into two doses of 100 mg taken 12h apart Cohort 9: total oral dose of 80mg or matching placebo split into two doses of 40 mg taken 12h apart
Part 1 (Single ascending dose) design (qd) - Overview of the study
Figure imgf000058_0001
Part 1 (Single ascending dose) design (Split daily intake) - Overview of the study
Figure imgf000058_0002
Part 2 (MAD)
Part 2 was a randomized, subject-blinded and investigator-partial-blinded, placebo-controlled, MAD study in which, eight subjects each were randomized into five cohorts to receive either compound of example 1 or matching placebo in a 6:2 ratio (active: placebo).
Subjects were assigned to one of the following cohort:
Cohort 1 : multiple once daily (qd) oral dose of 5 mg or matching placebo
Cohort 2: multiple once daily (qd) oral dose of 15 mg or matching placebo
Cohort 3: multiple oral daily dose of 20mg or matching placebo administered twice daily (bid) Cohort 4: multiple oral daily dose of 40mg or matching placebo administered twice daily (bid) Cohort 5: multiple oral daily dose of 80mg or matching placebo administered twice daily (bid)
Part 2 multiple ascending dose design (qd) - Overview of the study
Figure imgf000059_0001
Part 2 multiple ascending dose design (bid) - Overview of the study
Figure imgf000059_0002
Human safety and tolerability
In Part 1 (Single Ascending Dose, SAD) of the above study, healthy subjects received a single dose of compound of example 1 up to a maximal total daily dose of 200 mg (two times 100 mg administered 12 h apart). In Part 2 (Multiple Ascending dose, MAD) of study, the subjects received doses up to 80 mg BID for 12 days. No adverse events led to study discontinuation. The highest doses in both parts were safe and no maximal tolerated dose was established.
Some subjects who were treated with compound of example 1 or placebo in the SAD and MAD portions of this study experienced asymptomatic lipase elevations. Three treated subjects (one from the SAD part at the lowest dose of 5 mg, two from the MAD at the highest dose of 80 mg BID) and one placebo treated subject experienced increases in lipase. In the three subjects with lipase elevations, the lipase increase was accompanied by mild amylase increase (£1.5 ULN). All subjects were asymptomatic and events were transient, rapidly returning to normal levels while subjects continued to receive compound of example 1. No significant findings in physical exam, vital signs or ECGs have been related to compound of example 1.
Therefore, compound of example 1 was well tolerated in healthy subjects at doses up to 80 mg bid (160 mg daily dose) over 12 days.
Human pharmacokinetic data
Pharmacokinetic behavior of compound of example 1 was evaluated in healthy subjects following single and multiple oral doses. The PK parameters calculated were standard parameters used for measuring drug exposure in the systemic circulation after receiving single or multiple doses of compound of example 1 .
Part 1 (SAD)
The mean plasma concentration time profiles for compound of example 1 are shown in Figure 11.
Following single oral administration of the compound at 5, 10, 20, 30, 45, 70, two times 70 (split intake, 2 identical doses were administered 12 h apart), two times 100 (split intake) and two times 40 mg (split intake), the plasma exposure to compound of example 1 increased with dose and a median Tmax ranging from 1 to 1 .5 hours post dose indicated a fast absorption. After the concentration peak plasma concentrations decreased initially very rapidly; at later timepointsthe rate of concentration decline decreased strongly and the mean apparent elimination half-life (T1/2) ranged from 245 to 513 hours. With an increase in dose from 5 to 100 mg (i.e. 20-fold), Cmax increased by 21 .7-fold; AUC0-24h increased by 75 fold, with a 40-fold increase in daily dose (5 mg to 100 mg bid). The percentage coefficient of variation (CV%) ranged from 18.5 to 41 .4 for Cmax and 6.4 to 26.3 for AUC0-24h.
For the dose range of 5 mg to 2 times 100 mg (40-fold) for AUC and 5 mg to 100 mg for Cmax (20-fold), the estimated slope and the corresponding 90% Cl for Cmax was 1.08 (1.01 , 1.15), for AUCIast the slope was 1 .00 with 90% Cl (0.957; 1 .04) and for AUC0-24 the slope was 1 .20 with 90% Cl of (1.15, 1.24). Dose proportionality over the whole dose range was demonstrated for AUCIast but not for Cmax and AUC0-24h where data suggest a slightly over proportional increase of exposure with dose. However, for Cmax and AUC0-24h PK can be considered dose proportional for an up to 4.50-fold and 2.57-fold increase in dose, respectively.
Part 2 (MAD)
The mean plasma concentration time profiles of the compound are shown in Figure 12a (Day 1) and Figure 12b (Day 12).
Following oral administration of the first dose of compound of example 1 on Day 1 , for all dose groups (5 and 15 mg qd fasted; 20, 40 and 80 mg bid fed), the plasma concentrations of compound of example 1 increased in a dose dependent manner with a median T max ranging from 1 to 2.5 hours.
With an increase in the dose of compound of example 1 from 5 mg qd to 80 mg bid (i.e. 16-fold per dosing interval), Cmax and AUC on Day 12 (AUC0-12h,ss for 20, 40 and 80 mg bid cohorts and AUC0-24h,ss for 5 and 15 mg qd cohort) increased by 17.9-fold and 20.9-fold respectively. For the dose range of 5 to 80 mg (16 fold), the estimated slope and the corresponding 90% Cl for Cmax.ss was 1 .19 (1 .08,1 .30) and for AUCtau.ss the slope was 1 .07 with 90% Cl (1 .01 ; 1.12). Dose proportionality over the whole dose range was not demonstrated for both Cmax.ss and AUCtau.ss. However, Cmax.ss and AUCtau PK can be considered dose proportional for and up to 2.12 and 6.29 fold increase in dose, respectively. Dose proportionality criteria were met for AUCtau over the dose range 5 mg qd to 40 mg bid. The % CV ranged from 17.1 to 30.2 % for Cmax and 11.7 to 18.1 for AUC0-24h.
Across the investigated dose range Cmax.ss was between 1.39 to 1.19 times greater than after single dose (mean Cmax (Day 12)/ Cmax (Day 1)). For AUC (AUC0-12 and AUC0-24), the average ratios were between 2.08 to 1 .27. This indicates minor accumulation of compound of example 1 from day 1 to steady state. For the lowest dose of 5 mg qd 2.1-fold mean accumulation to steady state was observed but for all higher doses accumulation was £ 1 .4-fold. Little accumulation on day 1 of the bid regimen (mean Race, day 1 from 1.09 - 1.29) suggests that time to steady state is short. In 4 of the 5 dose groups morning pre-dose concentrations are higher on Day 3 as compared to Day 2, but the concentration differences are small. From Day 3 to Day 4 no consistent or major increase in morning pre-dose concentrations is observed. This demonstrates that steady state is achieved fast i.e. approximately on day 3. For BID oral dosing of compound of example 1 , in the fed state, steady state exposure is reached within 3-4 days of dosing. At steady state, dose proportionality criteria were met for AUCtau from 5 mg QD to 40 mg BID.
Human pharmacodynamic data
PD blood biomarker: Leukotriene B4 (LTB4) in blood
Determination of Leukotriene B4 (LTB4) in human plasma of ex vivo stimulated whole blood
In animal models the degree of LTB4 inhibition in ex-vivo stimulated blood correlated well with degree of therapeutic effect (Figure 10), in particular reduction of neutrophils in inflamed tissues. The peripheral PD readout of ex-vivo stimulated blood LTB4 is predictive for therapeutic efficacy and hence suitable to monitor target inhibition in the FIH trial.
Therefore, LTB4 concentrations were determined in plasma of ex-vivo stimulated whole blood by LC-MS/MS method.
Bioanalytical method for LTB4
Ex-vivo stimulation was performed at the clinical site according to the following procedure: Whole blood (500mL) were stimulated for 30 min using Calcium lonophore A23187. Then plasma supernatant is harvested, and frozen at -80°C for at least 24 hours prior to use.
LTB4 quantification by LC-MS/MS was performed as follow:
Human plasma was processed with organic precipitation followed by separation by reverse phase high performance liquid chromatography with tandem mass spectrometric detection.
Chromatography:
Reversed phase separation on Agilent Technologies 1290 Infinity HPLC and Zorbax SB-C18, Rapid Resolution HD 1.8mm (100 x 2.1 mm) column at 60°C and flow rate of 400 mL/min Total analytical run time of 11 .0 minutes.
Detection:
AB Sciex QTrap 5500 MS/MS Turbo Spray Ion Drive negative ion mode.
Sample from each subject and time point was prepared and analyzed in three aliquots. Aliquot one and two contained plasma from ex-vivo stimulated whole blood, while aliquot three as negative control contained plasma from non-stimulated whole blood. Samples with CV>25% for the first two aliquots and/or with detected values in aliquot three were excluded from the results.
LTB4 concentration data were calculated as mean of concentration of aliquot one and two. Concentration of aliquot three was not be included in the calculation. The change from baseline and percent inhibition (percent of baseline) for each biomarker was calculated. Baseline was the mean of the Baseline and Day 1 , -1 h measurements.
To avoid being treated as missing values when calculating changes from baseline and percent inhibition, values below the LLOQ were replaced by 0.5 x LLOQ and values above the upper limit of quantification (ULOQ) were replaced by 1 .5 x ULOQ.
Results:
Results from the first cohorts of the SAD indicated that blood cell stimulated LTB4 release remained at 25-50% reduction from baseline at 24 hours with 5 mg. Inhibition at 24 hours was observed to increase with increasing dose. An approximately 50% inhibition at 24 h after dosing was observed with the 10 mg dose and transient maximum inhibition (>90%) following Cmax was observed at the 20 mg dose. (Figure 13)
Additional cohorts in the SAD part confirmed the dose dependent inhibition of LTB4. Approximately 81%, 86% and 89% inhibition at 24h after dosing was observed with the 30 mg, 45 mg and 70 mg dose, respectively.
Steady state data from cohorts 1 to 3 in the MAD part, confirm the dose dependent inhibition of LTB4. Approximately 75%, 90% and 99% inhibition was observed in samples collected pre-dose at Day 9 (steady state) with the 5 mg (Q.D.), 15 mg (Q.D.) and 20 mg (B.I.D.) dose, respectively. (Figure 14)
Target inhibition of 90-99% was maintained at 15 mg (Q.D.) and 20 mg (B.I.D.) doses, in the MAD part of the study.
In conclusion, steady state leukotriene A4 hydrolase target inhibition assessed ex-vivo in blood samples was on average 75% at 5mg QD, 90% at 15mg QD and 99% at 20mg QD bid.
Example 5: Efficacy and safety of (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2- yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid (Form B) in adult patients with NAFLD who manifest a NASH-like biomarker phenotype Provided below are the details of the clinical trial design to demonstrate the efficacy of (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid alone or in combination with Tropifexor, as compared to placebo.
Clinical Study design
This is a non-confirmatory, multicenter, open label, platform study in NAFLD patients with a NASH-like biomarker phenotype.
The NEXSCOT study uses a platform type design to investigate “multiple targeted therapies in the context of a single disease in a perpetual manner” (Woodcock and LaVange 2017; N Engl J Med, 377(1), 62-70). Each single or combination regimen will be considered a unique treatment arm. One or more treatment arms which are bundled together for review and execution are considered a cohort. The treatment arm(s) within a cohort will be studied in a way that allows for the comparison of the treatment effect. Typically, a particular treatment arm or cohort will be independent and will not necessarily provide decision making information for future treatment arms or cohorts.
Each cohort will consist of a 33-day screening period (Day -60 to Day -28), a baseline period of 27 days (Day -27 to Day -1), a treatment period of 12 weeks (Day 1 to Day 85), and a study completion evaluation approximately 28 days after the last drug administration (Day 113). Patients will be advised to maintain their recommended diet for NAFLD during the study.
Patients who meet the eligibility criteria at screening will have baseline assessments performed, including determination of the percent liver fat content by MRI.
Once eligibility has been confirmed from screening and baseline assessments, patients will be randomized into any of the treatment arms open to enrollment for which the patient meets the eligibility criteria using an Interactive Response Technology (IRT) system. If a patient does not meet the specific eligibility criteria of a particular treatment arm, the patient will be randomized to any open and actively enrolling arms for which the patient is eligible. Randomization will be stratified by race and BMI
Study patient
The study enrolls NAFLD patients with characteristics consistent with NASH:
Two risk factors for NASH: elevated BMI and history of Type II diabetes mellitus, and • Three phenotypic biomarkers for NASH as shown in Table 2: elevated % liver fat diagnostic of NAFLD as measured by MRI-PDFF, liver inflammation as measured by ALT, and ongoing liver fibrotic process as measured by ELF Test. These phenotypic biomarkers are consistent with the pathobiology of NASH and recent studies have supported these biomarkers being highly predictive of NASH (Harrison et al 2018a; J Hepatol Suppl, 68(1), p. S38; Sanyal et al 2017; J Hepatol Suppl, 66(1), p. S89).
Table 2 Biomarker inclusion criteria compared to normal range
Figure imgf000065_0001
In this study, ELF Test measurement will be conducted at screening (Inclusion criteria), and patients with an ELF Test result within the range of 8.5-10.5 will be eligible for participation in the study. The goal of using these ELF Test inclusion criteria is to enrich the study population for those subjects who have an ongoing fibrotic process in the liver but do not yet have severe fibrosis or cirrhosis, typically considered to be F1-3. This ELF Test inclusion range is based on a weight- of-evidence analysis of the following reports:
In their review of the current state-of-the-art of soluble liver fibrosis biomarkers, Vilar-Gomez et al. (2018; J Hepatol, 68(2), p. 3015) noted that the area under the receiver operating characteristic curve (AUROC) for ELF Test using 8.5-10.18 cut-off values to detect liver fibrosis (F³2) was 0.82 in a meta-analysis of adult and pediatric patients (n=1329) with chronic liver disease, including NAFLD. • Lichtinghagen et al (2013; J Hepatol, 59(2), p. 236) proposed the following ELF Test cut-off values based on the results of this test in healthy volunteers (n=400) and the relationship of this test with liver biopsy staged fibrosis in patients with hepatitis C (n=39):
• £7.7 excluded fibrosis with high sensitivity
• ³9.8 had high specificity for identification of fibrosis
• ³11.3 discriminated cirrhosis
• The National Institute for Health and Care Excellence (2016) proposed that NAFLD patients should have ELF Test as a measure of advanced liver fibrosis and proposed that a value of ³10.51 was consistent with advanced liver fibrosis.
• Gungoren et al (2018, J Lab Percis Med, 3(21), p.1-7) showed that using an ELF Test cut-off of ³8.84 was associated with a liver fibrosis score of F³2 at an AUROC of 0.89 in 46 patients with auto-immune hepatitis who have liver biopsy.
Study treatment
The cohort 1 consist of two treatment arms
Arm 1 : (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid 20mg BID
Arm 2: (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid 20mg BID and Tropifexor 200mg
Objectives
Figure imgf000066_0001
Figure imgf000067_0001
Safety and tolerability will be the paramount and primary objective, assessed by safety endpoints (including vital signs, physical examination, laboratory measurements, ECG) and adverse events.
Secondary objectives are to measure efficacy. The secondary endpoints are
• ALT, % liver fat (MRI-PDFF), and ELF Test: These biomarkers will provide evidence of efficacy and are further described below.
• Cardiometabolic risk factors: NAFLD and NASH patients are at high risk for cardiovascular morbidity and mortality. Therefore, it is important to characterize any negative or positive effects of single drugs or drug combinations on cardiometabolic risk factors including body weight, Homeostasis Model Assessment (HOMA), hemoglobin A1C (HbA1c), fasting glucose, fasting insulin, and fasting lipid profile.
• Pharmacokinetics: The potential pharmacokinetic interactions of drugs used in combination will be measured.
Given the relatively low variability of the secondary endpoints ALT, % liver fat (MRI-PDFF), and ELF Test, a sample size of 20 patients should provide sufficient power to resolve relevant, high effect size changes from baseline of -30%, -30% and -3%, respectively. Drugs and/or combinations with reasonable expectation of substantive efficacy. Placebo effect
Currently, there are no approved treatments for NASH. However, over the last five years, the results of a number of studies in this indication have been reported in the medical literature (Sanyal et al 2017, J Hepatol Suppl, 66(1), p S89; Harrison et al 2018a; Harrison et al 2018b, Lancet, 391 (10126), p. 1174) Based on these results, considerable knowledge of the magnitude of both placebo and active treatment effects in NASH studies now exists.
The placebo effects reported in four relatively short-term (≤ 16 week) NASH studies for three main efficacy endpoints, ALT, % liver fat (MRI-PDFF) and ELF Test, are summarized in Table 3.
Table 3 Examples of placebo effect in published studies in NASH and NASH-like patient populations
Figure imgf000068_0001
Efficacy endpoint Conversely, a review of multiple, recent studies in patients with NASH or a NASH-like phenotype shows that high efficacy, active treatment is associated with relative percent decreases in % liver fat (MRI-PDFF) and ALT of ³ 30% (Sanyal et al 2017; Harrison et al 2018a; Harrison et al 2018b). Similarly, a decrease in ELF Test by ³ 0.3 units or 3% can be considered evidence of a substantive, beneficial reversal in the ongoing liver fibrotic process (Harrison et al 2018a).

Claims

WE CLAIM
1. A LTA4H inhibitor for use in the treatment and/or the prevention of a liver disease or disorder in a patient in need of such treatment and/or prevention.
2. A LTA4H inhibitor for use according to claim 1 , wherein said LTA4H inhibitor is a compound of Formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000070_0001
wherein,
R1 is OH or NH2;
Y is O, S or CH2;
X1, X2, X3 and X4 are N; or
X1, X2, X3 and X4 are selected from N, NH, C, CH and O with the proviso that at least two of X1, X2, X3 and X4 are N or NH;
R2 is C1-C6 alkyl optionally substituted by phenyl; C3-C6 cycloalkyl; phenyl optionally being substituted by halogen, cyano, C1-C6 alkyl optionally substituted by halogen, C1-C6 alkoxy, or a 5 - 6 membered heteroaryl ring containing 1 to 3 heteroatoms selected from N, O and S; or a 5 - 10 membered mono- or bicyclic heteroaryl containing 1 to 4 heteroatoms selected from N, O and S, said heteroaryl being optionally substituted by halogen, cyano or C1-C6 alkyl optionally substituted by halogen.
3. A LTA4H inhibitor, for use according to claim 1 or 2, wherein said LTA4H inhibitor is (S)-3- amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof.
4. A LTA4H inhibitor for use according to any one of claims 1-3, wherein the LTA4H inhibitor is disposed in a pharmaceutical formulation, wherein said pharmaceutical formulation comprises one or more pharmaceutically acceptable carriers, each of which is independently selected from a filler, a lubricant, a binder, a desintegrant and a glidant.
5. A LTA4H inhibitor for use according to claim 4, wherein the pharmaceutical formulation is in tablet or capsule form.
6. A pharmaceutical composition comprising a LTA4H inhibitor which is a compound of Formula (I), or a pharmaceutically acceptable salt thereof in accordance to claim 2, together with one or more pharmaceutically acceptable carriers for use in the treatment and/or prevention of liver disease or disorder in a patient in need of such treatment and/or prevention.
7. A pharmaceutical combination comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof according to claim 2, and a FXR agonist.
8. A pharmaceutical combination according to claim 7, wherein the compound of Formula (I) is (S)-3-amino-4-(5-(4-((5-chloro-3-fluoropyridin-2-yl)oxy)phenyl)-2H-tetrazol-2-yl)butanoic acid, or a pharmaceutically acceptable salt thereof and the FXR agonist is Tropifexor.
9. A pharmaceutical combination according to claim 7 or 8, for use in the treatment and/or prevention of a liver disease or disorder in simultaneous, sequentially or separate administration.
10. A LTA4H inhibitor for use according to any one of claims 1 -5 or a pharmaceutical composition for use according to claim 6, a pharmaceutical combination for use according to any one of claims 7- 9, wherein the LTA4H inhibitor is administered at a dose of about 10 mg to about 30mg twice a day.
11 . A LTA4H inhibitor for use according to any one of claims 1 -5 or a pharmaceutical composition for use according to claim 6, or a pharmaceutical combination for use according to any one of claims 7-9, wherein the FXR agonist is administered at a dose in a range of about 1mg to about 250mg.
12. A LTA4H inhibitor for use according to any one of claims 1-5, 10 and 11 , or a pharmaceutical composition for use according to any one of claims 6, 10 and 11 , or a pharmaceutical combination for use according to any one of claims 7 to 11 , wherein the patient is a NAFLD patient selected according to at least one of the following criteria: a) prior to the treatment with LTA4H inhibitor, the patient has circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test ³8.5 and £10.5; b) prior to treatment with the LTA4H inhibitor, the patient has an intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI- PDFF) ³8%; c) prior to treatment with the LTA4H inhibitor, the patient has circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT) ³43 IU/L for male and ³28 IU/L for female.
13. A LTA4H inhibitor for use according to any one of claims 1-5 and 10-12, or a pharmaceutical composition for use according to any one of claims 6 and 10-12, or a pharmaceutical combination for use according to any one of claims 7 to 12, wherein said patient achieves by week 12 of treatment at least one of the following: a) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or b) at least 3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, together with at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or c) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or; d) between 2% and <3% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT); or e) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 10% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 35% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF); or f) less than 2% decrease in circulating markers of ongoing liver fibrosis as measured by Enhance Liver Fibrosis (ELF) Test, and at least 35% decrease in circulating markers of liver and/or systemic inflammation as measured by Alanine aminotransferase (ALT), and at least 15% in intrahepatic lipid content (liver fat content) as measured by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF).
14. A LTA4H inhibitor for use according to any one of claims 1-5 and 10-13, or a pharmaceutical composition for use according to any one of claims 6 and 10-13, or a pharmaceutical combination for use according to any one of claims 7 to 13, wherein a liver disease or disorder is selected from chronic liver disease, e.g. a liver disease or disorder selected from the group consisting of cholestasis, intrahepatic cholestasis, estrogen-induced cholestasis, drug-induced cholestasis, cholestasis of pregnancy, parenteral nutrition-associated cholestasis, primary biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), progressive familiar cholestasis (PFIC), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis (ASH), drug-induced bile duct injury, gallstones, liver cirrhosis, alcohol-induced cirrhosis, cystic fibrosis-associated liver disease (CFLD), bile duct obstruction, cholelithiasis, liver fibrosis, renal fibrosis, dyslipidemia, atherosclerosis, diabetes, diabetic nephropathy, colitis, newborn jaundice, prevention of kernicterus, veno-occlusive disease, portal hypertension, metabolic syndrome, hypercholesterolemia, progressive fibrosis of the liver caused by any of the diseases above or by infectious hepatitis; e.g. NAFLD, NASH, liver fibrosis, or PBC.
PCT/IB2020/058229 2019-09-06 2020-09-04 Methods of treating liver disease using lta4h inhibitors WO2021044351A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962896917P 2019-09-06 2019-09-06
US62/896,917 2019-09-06

Publications (1)

Publication Number Publication Date
WO2021044351A1 true WO2021044351A1 (en) 2021-03-11

Family

ID=72432977

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2020/058229 WO2021044351A1 (en) 2019-09-06 2020-09-04 Methods of treating liver disease using lta4h inhibitors

Country Status (2)

Country Link
TW (1) TW202122078A (en)
WO (1) WO2021044351A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US20090111794A1 (en) * 2007-10-31 2009-04-30 Bacani Genesis M Aryl-substituted bridged or fused diamines as modulators of leukotriene A4 hydrolase
WO2012087519A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating fxr
WO2014085474A1 (en) 2012-11-28 2014-06-05 Intercept Pharmaceuticals, Inc. Treatment of pulmonary disease
WO2014164658A1 (en) 2013-03-12 2014-10-09 Celtaxsys, Inc. Methods of inhibiting leukotriene a4 hydrolase
WO2015069666A1 (en) 2013-11-05 2015-05-14 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2015092740A1 (en) 2013-12-20 2015-06-25 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2017145041A1 (en) * 2016-02-22 2017-08-31 Novartis Ag Methods for using fxr agonists
US20180162802A1 (en) * 2016-12-09 2018-06-14 Celtaxsys, Inc. Monamine and monoamine derivatives as inhibitors of leukotriene a4 hydrolase

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
US20090111794A1 (en) * 2007-10-31 2009-04-30 Bacani Genesis M Aryl-substituted bridged or fused diamines as modulators of leukotriene A4 hydrolase
WO2012087519A1 (en) 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating fxr
WO2014085474A1 (en) 2012-11-28 2014-06-05 Intercept Pharmaceuticals, Inc. Treatment of pulmonary disease
WO2014164658A1 (en) 2013-03-12 2014-10-09 Celtaxsys, Inc. Methods of inhibiting leukotriene a4 hydrolase
WO2015069666A1 (en) 2013-11-05 2015-05-14 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2015092740A1 (en) 2013-12-20 2015-06-25 Novartis Ag Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2017145041A1 (en) * 2016-02-22 2017-08-31 Novartis Ag Methods for using fxr agonists
US20180162802A1 (en) * 2016-12-09 2018-06-14 Celtaxsys, Inc. Monamine and monoamine derivatives as inhibitors of leukotriene a4 hydrolase

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY, pages: 1289 - 1329
ANONYMOUS: "Celtaxsys announces the issue of four new patents expanding its pipeline of selective leukotriene B4 modulation anti-inflammatory medicines", 1 February 2018 (2018-02-01), XP002801241, Retrieved from the Internet <URL:https://www.globenewswire.com/news-release/2018/02/01/1330002/0/en/Celtaxsys-announces-the-issue-of-four-new-patents-expanding-its-pipeline-of-selective-leukotriene-B4-modulation-anti-inflammatory-medicines.html?print=1> [retrieved on 20201126] *
BORGESON ET AL., CELL METAB, vol. 22, no. 10, 2015, pages 125 - 37
BRUNT ET AL., HEPATOLOGY, vol. 53, no. 3, 2011, pages 810 - 820
ELIAS ET AL., DIABETES, vol. 65, no. 8, 2016, pages 2139 - 50
GUNGOREN ET AL., J LAB PERCIS MED, vol. 3, no. 21, 2018, pages 1 - 7
HARRISON ET AL., LANCET, vol. 391, no. 10126, 2018, pages 1174
INTERCEPT ANNOUNCES NEW FLINT TRIAL DATA SHOWING OCA TREATMENT INCREASES FIBROSIS RESOLUTION AND CIRRHOSIS PREVENTION IN HIGH-RISK NASH PATIENTS, 23 April 2015 (2015-04-23)
LI ET AL., MOL ENDOCRINOL, vol. 28, no. 8, 2014, pages 1316 - 28
LICHTINGHAGEN ET AL., J HEPATOL, vol. 59, no. 2, 2013, pages 236
MARTÍNEZ-CLEMENTE MARCOS ET AL: "The 5-lipoxygenase/leukotriene pathway in obesity, insulin resistance, and fatty liver disease", CURRENT OPINION IN CLINICAL NUTRITION AND METABOLIC CARE NOV 2010, LIPPINCOTT WILLIAMS & WILKINS, US, vol. 14, no. 4, 1 July 2011 (2011-07-01), pages 347 - 353, XP009194872, ISSN: 1473-6519, DOI: 10.1097/MCO.0B013E32834777FA *
MOTHE-SATNEY ET AL., DIABETES, vol. 61, no. 9, 2012, pages 2311 - 9
MUDALIAR ET AL., GASTROENTEROLOGY, vol. 145, no. 3, 2013, pages 574 - 82
NEUSCHWANDER-TETRI ET AL., LANCET, vol. 385, no. 9972, 2015, pages 956 - 65
OH ET AL., NAT REV DRUG DISC, vol. 15, 2016, pages 161 - 172
PATTNI ET AL., CLIN TRANS GASTROENTEROL, vol. 3, 2012, pages e18
RATZIU ET AL., J GASTROINTESTIN LIVER DIS, vol. 19, no. 4, 2010, pages 415 - 23
SANYAL ARUN ET AL: "Tropifexor, a farnesoid X receptor agonist for the treatment of non-alcoholic steatohepatitis: Interim results based on baseline body mass index from first two parts of Phase 2b study FLIGHT-FXR", JOURNAL OF HEPATOLOGY, & INTERNATIONAL LIVER CONGRESS / 54TH ANNUAL MEETING OF THE EUROPEAN-ASSOCIATION-FOR-THE-STUDY-OF-THE-LIVER (EASL); VIENNA, AUSTRIA; APRIL 10 -14, 2019, vol. 70, no. 1, Suppl. 1, 1 April 2019 (2019-04-01), pages E796 - E797, XP002800455 *
SANYAL ET AL., J HEPATOL, vol. 66, no. 1, 2017, pages S89
SCHAPP ET AL., NAT REV GASTROENTEROL HEPATOL, vol. 11, no. 1, 2014, pages 55 - 67
SPITE ET AL., J IMMUNOL, vol. 187, no. 4, 2011, pages 1942 - 9
STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", February 2011, WILEY-VCH
TITOS E ET AL: "Protection from hepatic lipid accumulation and inflammation by genetic ablation of 5-lipoxygenase", PROSTAGLANDINS AND OTHER LIPID MEDIATORS, ELSEVIER, US, vol. 92, no. 1-4, 1 June 2010 (2010-06-01), pages 54 - 61, XP027047553, ISSN: 1098-8823, [retrieved on 20100321] *
VILAR-GOMEZ ET AL., J HEPATOL, vol. 68, no. 2, 2018, pages 3015
WALTERS ET AL., ALIMENT PHARMACOL THER, vol. 41, no. 1, 2015, pages 54 - 64
WONG ET AL., GASTROENTEROLOGY, vol. 148, no. 3, 2015, pages 547 - 55
WOODCOCKLAVANGE, N ENGL J MED, vol. 377, no. 1, 2017, pages 62 - 70
YOUNOSSI ET AL., HEPATOLOGY, vol. 64, no. 5, 2016, pages 1577 - 1586

Also Published As

Publication number Publication date
TW202122078A (en) 2021-06-16

Similar Documents

Publication Publication Date Title
US20230234968A1 (en) Methyl-substituted benzobisoxazole compound and use thereof
CN107406424B (en) Estrogen receptor modulators and uses thereof
JP6865038B2 (en) Treatment of NAFLD and NASH
CN108349893A (en) Farnesoid X receptor agonists and application thereof
JP2017105751A (en) Method of treating cancer and bone cancer pain
US20230241049A1 (en) Rbp4 antagonists for treatment and prevention of non-alcoholic fatty liver disease and gout
WO2015138986A1 (en) Fxr agonists and methods for making and using
EA019252B1 (en) SUBSTITUTED (1,2,3,4-TETRAHYDROCYCLOPENTA[b]INDOL-3-YL)ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
WO2020259528A1 (en) Method for treating idiopathic pulmonary fibrosis
EA028378B1 (en) Deuterated derivatives of ivacaftor
JP2019530742A (en) Disubstituted pyrazole compounds for the treatment of diseases
WO2012175049A1 (en) Amide compound, preparation method and uses thereof
JP2022500393A (en) Farnesoid X receptor agonist and its use
AU2019242387A1 (en) Calpain modulators and therapeutic uses thereof
WO2019024776A1 (en) Pharmaceutical composition and use thereof
US20080269189A1 (en) Method for Avoiding Edema in the Treatment or Prevention of Ppary-Responsive Diseases, Including Cancer
US11883539B2 (en) Pharmaceutical compositions of berberine with EPA and DHA, and methods thereof
CN1652787A (en) Compositions and methods for combating lower urinary tract dysfunctions with delta opioid receptor agonists
TWI729094B (en) Endosulfonamide compounds and methods of use
WO2021044351A1 (en) Methods of treating liver disease using lta4h inhibitors
JP2024510431A (en) Aromatic compounds, their preparation and applications
TW201818964A (en) Methods of using tryptophan hydroxylase inhibitors
TWI828839B (en) Methods of treating hidradenitis suppurativa using lta4h inhibitors
EP4166137A1 (en) Therapeutic agent for fatty liver disease
CN113880747A (en) Indole derivative and application thereof

Legal Events

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

Ref document number: 20768718

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20768718

Country of ref document: EP

Kind code of ref document: A1