EP4054567A1 - Traitement de troubles hépatiques - Google Patents

Traitement de troubles hépatiques

Info

Publication number
EP4054567A1
EP4054567A1 EP20886094.0A EP20886094A EP4054567A1 EP 4054567 A1 EP4054567 A1 EP 4054567A1 EP 20886094 A EP20886094 A EP 20886094A EP 4054567 A1 EP4054567 A1 EP 4054567A1
Authority
EP
European Patent Office
Prior art keywords
compound
patient
liver
formula
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP20886094.0A
Other languages
German (de)
English (en)
Other versions
EP4054567A4 (fr
Inventor
Martijn Fenaux
Yujin Wang
Weidong Zhong
Kevin Klucher
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Terns Pharmaceuticals Inc
Original Assignee
Terns Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Terns Pharmaceuticals Inc filed Critical Terns Pharmaceuticals Inc
Publication of EP4054567A1 publication Critical patent/EP4054567A1/fr
Publication of EP4054567A4 publication Critical patent/EP4054567A4/fr
Pending legal-status Critical Current

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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

Definitions

  • This invention relates to methods and compositions for treating a liver disorder in a patient.
  • FLD Fatty liver disease
  • NAFLD non-alcoholic fatty liver disease
  • NASH non-alcoholic steatohepatitis
  • liver disorder in a patient in need thereof comprising administering a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • NAFLD non alcoholic fatty liver disease
  • NASH non-alcoholic steatohepatitis
  • Also provided herein are methods of impeding or slowing the progression of NASH in a patient in need thereof comprising administering a therapeutically effective amount of a compound of Formula (I) pharmaceutically acceptable salt thereof.
  • FXR Famesoid X Receptor
  • the administration does not result in pruritus in the patient greater than Grade 2 in severity. In some embodiments, the administration does not result in pruritus in the patient greater than Grade 1 in severity. In some embodiments, the administration does not result in pruritus in the patient.
  • the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver
  • the liver disorder is NAFLD. In some embodiments, the liver disorder is NASH. In some embodiments, the liver disorder is PSC. In some embodiments, the liver disorder is PBC.
  • the administration results in a liver concentration to plasma concentration ratio of the compound of Formula (I) of 10 or greater.
  • the therapeutically effective amount is 0.5 pg/day - 600 mg/day. In some embodiments, the therapeutically effective amount is 0.5 pg/day - 20 mg/day. In some embodiments, the therapeutically effective amount is 0.5 pg/day - 4 mg/day.
  • the administration comprises administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, daily for a treatment period of one or more weeks.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered once daily or twice daily.
  • the amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, administered on day 1 of the treatment period is greater than or equal to the amount administered on all subsequent days of the treatment period.
  • the amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, administered on day 1 of the treatment period is equal to the amount administered on all subsequent days of the treatment period.
  • the treatment period is one or more months. In some embodiments, the treatment period is the remaining lifespan of the patient.
  • the patient is obese. In some embodiments, the patient is not obese. In some embodiments, the patient also has diabetes mellitus and/or a cardiovascular disorder. In some embodiments, the patient is at risk for developing an adverse effect affecting one or more of the kidneys, lung, heart, and skin. In some embodiments, the patient is 2-17 years old. In some embodiments, the patient is 18-54 years old. In some embodiments, the patient is 65 or more years old. In some embodiments, the patient has had a liver transplant.
  • the patient s alkaline phosphatase, gamma-glutamyl transferase (GGT), alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) levels are elevated.
  • the method does not comprise administering an antihistamine, an immunosuppressant, a steroid, rifampicin, an opioid antagonist, or a selective serotonin reuptake inhibitor (SSRI).
  • SSRI selective serotonin reuptake inhibitor
  • NAS score of the patent is decreased upon administration.
  • TGR5 signaling is not activated upon administration.
  • the administration results in decreased level of expression of a marker of fibrosis.
  • the expression level of Ccr2, Collal, Colla2, Colla3, Cxcr3, Den, Hgf, Ilia, Inhbe, Lox, Loxll, Loxl2, Loxl3, Mmp2, Pdgfb, Plau, Serpinel, Perpinhl, Snai, Tgfbl, Tgfb3, Thbsl, Thbs2, Timp2, and/or Timp3 is reduced.
  • the administration results in decreased level of expression of a marker of liver inflammation.
  • the level of Adgrel, Ccr2, Ccr5, 111A, and/or Tlr4 is reduced.
  • FIG. 1 A shows plasma concentrations of Compound I at various time points after intravenous (IV) administration to rats (1 mg/kg), dogs (1 mg/kg) and monkeys (0.3 mg/kg).
  • FIG. IB shows plasma concentrations of Compound I at various time points after oral administration to mice (10 mg/kg), rats (10 mg/kg), dogs (3 mg/kg) and monkeys (5 mg/kg).
  • FIG. 2A shows the liver to plasma ratio of the concentration of Compound I, obeticholic acid (OCA), cilofexor, or tropifexor after 2 mg/kg IV administration to Sprague- Dawley (SD) rats.
  • OCA obeticholic acid
  • SD Sprague- Dawley
  • FIG. 2B shows the tissue to plasma ratio of the concentration of Compound I for kidney, lung, and liver after 2 mg/kg IV administration of Compound I to SD rats with or without co-administration of rifampicin.
  • FIG. 3 shows the tissue distribution of radiolabeled Compound I in plasma, liver, small intestine, cecum, kidney, lungs, heart, and skin after 5 mg/kg oral administration of Compound I to Long-Evans rats.
  • FIG. 4 shows the pharmacodynamics of Compound I administration, as measured by 7-alpha-hydroxy-4-cholesten-3-one (7a-C4), after administration of 0.3 mg/kg, 1 mg/kg or 5 mg/kg oral dose to cynomolgus monkeys.
  • FIG. 5A shows the pharmacokinetics of Compound I administration, after administration of 1 mg/kg oral dose for one day, or 7 consecutive daily doses, to cynomolgus monkeys.
  • FIG. 5B shows the pharmacodynamics of Compound I administration, as measured by 7-alpha-hydroxy-4-cholesten-3-one (7a-C4), after administration of 1 mg/kg oral dose for one day, or 7 consecutive daily doses, to cynomolgus monkeys.
  • FIG. 6 shows RT-qPCR results measuring liver SHP1, liver OSTb, ileum SHP1, and ileum FGF15 RNA expression after administering 10 mg/kg Compound I, 30 mg/kg OCA, or vehicle control to C5BL/6 mice.
  • FIG. 7A shows the number of differentially expressed genes (vs. vehicle-treated: fold- change >1.5-fold; p ⁇ 0.05) modulated by the administration of 10 mg/kg Compound I (500 total genes modulated) or 30 mg/kg OCA to C5BL/6 mice (44 total genes modulated), as well as the shared number differentially expressed genes that are modulated by both compounds (37 total genes).
  • FIG. 7B shows average expression levels (as shown by CPM value) of select FXR- related genes in C5BL/6 mice treated with 10 mg/kg Compound I or 30 mg/kg OCA, or a vehicle control.
  • FIG. 7C shows the number of pathways enriched (p ⁇ 0.05 ) by the administration of 10 mg/kg Compound I (32 pathways) or 30 mg/kg OCA to C5BL/6 mice (6 pathways), as well as the number of enriched pathways by either compound (2 pathways).
  • FIG. 7D shows the 25 pathways most statistically enriched upon administration of 10 mg/kg Compound I to C57BL/6 mice, and compares the enrichment of those pathways to the enrichment upon administration of 30 mg/kg OCA.
  • FIG. 8 shows the design of a study testing the efficacy of Compound I on a mouse model of NASH.
  • FIG. 9 shows the NAFLD Activity Score (NAS) of control mice and mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 10A shows the steatosis score of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 10B shows the inflammation score of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. IOC shows the ballooning score of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 11 A shows a histological section of fibrosis in control mice and NASH mice treated with 100 mg/kg Compound I.
  • FIG. 1 IB shows the amount of fibrosis in control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 12A shows the serum alanine amino transferase (ALT) levels of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 12B shows aspartate amino transferase (AST) of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 12C shows serum triglyceride levels of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 12D shows serum total cholesterol levels of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 13 A shows live triglyceride levels of control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 13B shows representative histology of steatosis assessment for control mice and NASH mice treated with 100 mg/kg Compound I.
  • FIG. 14A shows COL1 A expression in the liver in control mice and NASH mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 14B shows expression levels of inflammatory genes in control mice and NASH mice treated with 30 mg/kg Compound I.
  • FIG. 14C shows expression of fibrosis genes in control mice and NASH mice treated with 30 mg/kg Compound I.
  • FIGs. 15A and 15B show mean serum concentrations of Compound I in different dosing groups on Day 1 and Day 7, respectively, after administration of Compound I.
  • FIGs. 16A and 16B show changes of mean serum levels of 7a-C4 from the pre-dose baseline in different dosing groups on Day 1 and Day 7, respectively, after administration of Compound I or placebo.
  • FIGs. 17A and 17B show changes of mean serum levels of FGF-19 from the pre-dose baseline in different dosing groups on Day 1 and Day 7, respectively, after administration of Compound I or placebo.
  • FIG. 18 shows changes of mean serum levels of low-density lipoprotein (LDL) from the pre-dose baseline in different dosing groups after administration of Compound I or placebo.
  • LDL low-density lipoprotein
  • FLD Fatty liver disease encompasses a spectrum of disease states characterized by excessive accumulation of fat in the liver often accompanied with inflammation. FLD can lead to non-alcoholic fatty liver disease (NAFLD), which may be characterized by insulin resistance. If untreated, NAFLD can progress to a persistent inflammatory response or non-alcoholic steatohepatitis (NASH), progressive liver fibrosis, and eventually to cirrhosis.
  • NASH non-alcoholic steatohepatitis
  • Fatty liver diseases such as NAFLD and NASH
  • NAFLD is the second most common reason for liver transplantation. Accordingly, the need for treatment is urgent, but due to the lack of obvious symptoms to the patient, patients may lack the motivation to maintain treatment regimens, particularly burdensome treatment regimens, such as injected medicines, medications that are administered many times a day, or any that produce dangerous or irritating side effects.
  • the Farnesoid X Receptor is a nuclear hormone receptor that controls the conversion of cholesterol into bile acids and maintains homeostasis of multiple metabolic pathways and, therefore, is considered an important clinical target for NASH.
  • FXR agonists such as cilofexor, tropifexor, obeti cholic acid (OCALIVA®), and ED-305 (Enanta) have all reported pruritus as a side effect. Pruritus can cause patient discomfort, decrease patient quality of life, and affect adherence to treatment regimens, which can be particularly of issue for conditions requiring chronic drug administration.
  • the treatment does not result in pruritus.
  • the treatment results in less pruritus than is associated with a corresponding treatment with an FXR agonist selected from the group consisting of obeticholic acid (OCA), cilofexor, or tropifexor.
  • OCA obeticholic acid
  • cilofexor cilofexor
  • tropifexor obeticholic acid
  • the method comprises administering to the patient a therapeutically effective amount of a compound of Formula (I): or a pharmaceutically acceptable salt or enantiomer thereof in some embodiments, the method comprises administering to the patient a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt or enantiomer thereof, in combination with a therapeutically effective amount of another agent.
  • the compound of Formula (I) is disclosed in US 2010/0152166, the content of which is incorporated by reference in its entirety, and specifically with respect to the compound of Formula (I) or a pharmaceutically acceptable salt or enantiomer thereof, as well as methods of making and using the foregoing
  • the liver disorder is liver inflammation, fibrosis, or steatohepatitis.
  • the liver disorder is selected from: liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), NAFLD, and NASH.
  • the liver disorder is NASH.
  • the liver disorder is liver inflammation.
  • the liver disorder is liver fibrosis.
  • the liver disorder is alcohol induced fibrosis.
  • the liver disorder is steatosis.
  • the liver disorder is alcoholic steatosis. In some embodiments, the liver disorder is NAFLD. In some embodiments, the liver disorder is primary sclerosing cholangitis (PSC). In some embodiments, the liver disorder is primary biliary cirrhosis (PBC)
  • NASH can progress, e.g., to one or more of liver cirrhosis, hepatic cancer, etc.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination. For example, a composition consisting essentially of the elements as defined herein would not exclude other elements that do not materially affect the basic and novel characteristic(s) of the claimed invention.
  • Consisting of shall mean excluding more than trace amount of, e.g., other ingredients and substantial method steps recited. Embodiments defined by each of these transition terms are within the scope of this invention.
  • “Combination therapy” or “combination treatment” refers to the use of two or more drugs or agents in treatment, e.g., the use of a compound of Formula (I) together with a second agent useful to treat liver disorders, such as NAFLD, NASH, and symptoms and manifestations of each thereof is a combination therapy.
  • Administration in “combination” refers to the administration of two agents (e.g., a compound of formula (I), and a second agent) in any manner in which the pharmacological effects of both manifest in the patient at the same time.
  • administration in combination does not require that a single pharmaceutical composition, the same dosage form, or even the same route of administration be used for administration of both agents or that the two agents be administered at precisely the same time.
  • Both agents can also be formulated in a single pharmaceutically acceptable composition.
  • a non-limiting example of such a single composition is an oral composition or an oral dosage form.
  • a compound of Formula (I) can be administered in combination therapy with a second agent in accordance with the present disclosure.
  • “Second agent” as used herein refers to an agent, which is other than a compound of Formula (I), and which is useful in a method described herein.
  • the term second is meant as a term to distinguish the agent from a compound of Formula (I) or a pharmaceutically acceptable salt or enantiomer thereof, and is not intended to signify an order of administration.
  • excipient means an inert or inactive substance that may be used in the production of a drug or pharmaceutical, such as a tablet containing a compound of the invention as an active ingredient.
  • Various substances may be embraced by the term excipient, including without limitation any substance used as a binder, disintegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, solutions for parenteral administration, materials for chewable tablets, sweetener or flavoring, suspending/gelling agent, or wet granulation agent.
  • Binders include, e.g ., carbomers, povidone, xanthan gum, etc.
  • coatings include, e.g.
  • lubricants include, e.g. , magnesium stearate, stearic acid, sodium stearyl fumarate, etc.
  • materials for chewable tablets include, e.g. , dextrose, fructose dc, lactose (monohydrate, optionally in combination with aspartame or cellulose), etc.
  • suspending/gelling agents include, e.g. , carrageenan, sodium starch glycolate, xanthan gum, etc.
  • sweeteners include, e.g.
  • wet granulation agents include, e.g. , calcium carbonate, maltodextrin, microcrystalline cellulose, etc.
  • Patient refers to mammals and includes humans and non-human mammals.
  • patient examples include, but are not limited to mice, rats, hamsters, guinea pigs, pigs, rabbits, cats, dogs, goats, sheep, cows, and humans. In some embodiments, patient refers to a human.
  • “Pharmaceutically acceptable” refers to safe and non-toxic, preferably for in vivo , more preferably, for human administration.
  • “Pharmaceutically acceptable salt” refers to a salt that is pharmaceutically acceptable.
  • a compound described herein may be administered as a pharmaceutically acceptable salt.
  • Salt refers to an ionic compound formed between an acid and a base.
  • salts include, without limitation, alkali metal, alkaline earth metal, and ammonium salts.
  • ammonium salts include, salts containing protonated nitrogen bases and alkylated nitrogen bases.
  • Exemplary and non-limiting cations useful in pharmaceutically acceptable salts include Na, K, Rb, Cs, NEE,
  • salts include, without limitation, salts of organic acids, such as carboxylic acids and sulfonic acids, and mineral acids, such as hydrogen halides, sulfuric acid, phosphoric acid, and the likes.
  • exemplary and non-limiting anions useful in pharmaceutically acceptable salts include oxalate, maleate, acetate, propionate, succinate, tartrate, chloride, sulfate, bisulfate, mono-, di-, and tribasic phosphate, mesylate, tosylate, and the likes.
  • “Therapeutically effective amount” or dose of a compound or a composition refers to that amount of the compound or the composition that results in reduction or inhibition of symptoms or a prolongation of survival in a patient. The results may require multiple doses of the compound or the composition.
  • Treating” or “treatment” of a disease in a patient refers to 1) preventing the disease from occurring in a patient that is predisposed or does not yet display symptoms of the disease; 2) inhibiting the disease or arresting its development; or 3) ameliorating or causing regression of the disease.
  • the terms “optional” or “optionally” as used throughout the specification means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not.
  • “the nitrogen atom is optionally oxidized to provide for the N-oxide (N ⁇ K)) moiety” means that the nitrogen atom may but need not be oxidized, and the description includes situations where the nitrogen atom is not oxidized and situations where the nitrogen atom is oxidized.
  • a liver disorder in a patient e.g., a human patient
  • a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • liver disorder in a patient (e.g., a human patient) in need thereof with an FXR agonist, comprising administering a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the liver disorder is NAFLD or NASH.
  • the liver disorder is NAFLD.
  • the liver disorder is NASH.
  • the patient has had a liver biopsy.
  • the method further comprising obtaining the results of a liver biopsy.
  • NAFLD non alcoholic fatty liver disease
  • NASH non-alcoholic steatohepatitis
  • the compound of Formula (I), as demonstrated in the examples described herein, is preferentially distributed to the liver, which, without being bound by theory, would allow the compound to reach its FXR target in the liver with fewer off-target adverse effects.
  • the examples herein show that the compound of Formula (I) has an approximately 20-fold higher concentration in the liver than in the plasma, kidney, lungs, heart, and skin. This trait would likely be particularly beneficial for vulnerable populations, such as children, the elderly, and people with comorbidities.
  • pruritus is a well -documented adverse effect of several FXR agonists and can result in patient discomfort, a decrease in patient quality of life, and an increased likelihood of ceasing treatment. Pruritus is particularly burdensome for indications, such as those described herein, including NASH, for which chronic drug administration is likely.
  • the tissue specificity of the compound of Formula (I), in particular the preference for liver over skin tissue is a striking and unpredicted observation that makes it more likely that the compound will not cause pruritus in the skin, a theory that has been substantiated by human trials thus far.
  • a liver disorder in a patient in need thereof e.g., a human patient
  • a Fasoid X Receptor (FXR) agonist that preferentially distributes in liver tissue over one or more of kidney, lung, heart, and skin tissues
  • the method comprising administering a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • a liver disorder in a patient in need thereof with a Farnesoid X Receptor (FXR) agonist (such as the compound of Formula (I) or a pharmaceutically acceptable salt thereof), wherein the FXR agonist does not activate TGR5 signaling.
  • FXR Farnesoid X Receptor
  • the level of an FXR-regulated gene is increased.
  • the level of small heterodimer partner (SHP), bile salt export pump (BSEP) and fibroblast growth factor 19 (FGF-19) is increased.
  • the liver disorder is NASH.
  • a method of reducing liver damage comprising administering an FXR agonist (such as a compound of Formula (I) or a pharmaceutically acceptable salt thereof) to an individual in need thereof.
  • an FXR agonist such as a compound of Formula (I) or a pharmaceutically acceptable salt thereof
  • fibrosis is reduced.
  • the level of expression of one or more markers for fibrosis is reduced.
  • the level of Ccr2, Collal, Colla2, Colla3, Cxcr3, Den, Hgf, Ilia, Inhbe, Lox, Loxll, Loxl2, Loxl3, Mmp2, pdgfb, Plau, Serpinel, Perpinhl, Snai, Tgfbl, Tgfb3, Thbsl, Thbs2, Timp2, and/or Timp3 expression is reduced.
  • the level of collagen is reduced.
  • the level of collagen fragments is reduced.
  • the level of expression of the fibrosis marker is reduced at least 2, at least 3, at least 4, or at least 5-fold. In some embodiments, the level of expression of the fibrosis marker is reduced about 2-fold, about 3-fold, about 4-fold, or about 5-fold.
  • a method of reducing liver damage comprising administering an FXR agonist (such as a compound of Formula (I) or a pharmaceutically acceptable salt thereof) to an individual in need thereof.
  • inflammation is reduced.
  • one or more markers of inflammation are reduced.
  • the level of expression of Adgrel, Ccr2, Ccr5, 111A, and/or Tlr4 is reduced.
  • the level of expression of the inflammation marker is reduced at least 2-, at least 3-, at least 4-, or at least 5-fold.
  • the level of expression of the fibrosis marker is reduced about 2-fold, about 3-fold, about 4-fold, or about 5-fold.
  • the administration results in a liver concentration to plasma concentration ratio of the compound of Formula (I) of 10 or greater, such as 11 or greater, 12 or greater, 13 or greater, 14 or greater, or 15 or greater.
  • the administration does not result in pruritus in the patient greater than Grade 2 in severity. In some embodiments, the administration does not result in pruritus in the patient greater than Grade 1 in severity. In some embodiments, the administration does not result in pruritus in the patient.
  • the grading of adverse effects is known.
  • Grade 1 pruritus is characterized as “Mild or localized; topical intervention indicated.”
  • Grade 2 pruritus is characterized as “Widespread and intermittent; skin changes from scratching (e.g., edema, papulation, excoriations, lichenification, oozing/crusts); oral intervention indicated; limiting instrumental ADL.”
  • Grade 3 pruritus is characterized as “Widespread and constant; limiting self care ADL or sleep; systemic corticosteroid or immunosuppressive therapy indicated.” Activities of daily living (ADL) are divided into two categories: “Instrumental ADL refer to preparing meals, shopping for groceries or clothes, using the telephone, managing money, etc.,” and “Self care ADL refer to bathing, dressing and undressing, feeding self, using the toilet, taking medications, and not bedridden.”
  • a liver disorder in a patient e.g., a human patient
  • an FXR agonist that does not result in detectable pruritus in the patient in need thereof
  • the method comprising administering to the patient in need thereof a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • the patient is a human. Obesity is highly correlated with NAFLD and NASH, but lean people can also be affected by NAFLD and NASH. Accordingly, in some embodiments, the patient is obese. In some embodiments, the patient is not obese. Obesity can be correlated with or cause other diseases as well, such as diabetes mellitus or cardiovascular disorders. Accordingly, in some embodiments, the patient also has diabetes mellitus and/or a cardiovascular disorder. Without being bound by theory, it is believed that comorbidities, such as obesity, diabetes mellitus, and cardiovascular disorders can make NAFLD and NASH more difficult to treat. Conversely, the only currently recognized method for addressing NAFLD and NASH is weight loss, which would likely have little to no effect on a lean patient.
  • the risk for NAFLD and NASH increases with age, but children can also suffer from NAFLD and NASH, with literature reporting of children as young as 2 years old (Schwimmer, et ak, Pediatrics, 2006, 118:1388-1393).
  • the patient is 2-17 years old, such as 2-10, 2-6, 2-4, 4-15, 4-8, 6-15, 6-10, 8-17, 8-15, 8-12, 10-17, or 13-17 years old.
  • the patient is 18-64 years old, such as 18-55, 18-40, 18-30, 18-26, 18-21, 21-64, 21-55, 21-40, 21-30, 21-26, 26-64, 26-55, 26-40, 26-30, 30-64, 30-55, 30-40, 40-64, 40- 55, or 55-64 years old.
  • the patient is 65 or more years old, such as 70 or more, 80 or more, 90 or more.
  • NAFLD and NASH are common causes of liver transplantation, but patients that already received one liver transplant often develop NAFLD and/or NASH again. Accordingly, in some embodiments, the patient has had a liver transplant.
  • the patient’s alkaline phosphatase, gamma-glutamyl transferase (GGT), alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) levels are elevated.
  • the GGT, ALT, and/or AST levels are elevated prior to treatment with a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the patient’s ALT level is about 2-4-fold greater than the upper limit of normal levels.
  • the patient’s AST level is about 2-4-fold greater than the upper limit of normal levels.
  • the patient’s GGT level is about 1.5-3-fold greater than the upper limit of normal levels. In some embodiments, the patient’s alkaline phosphatase level is about 1.5-3-fold greater than the upper limit of normal levels.
  • Normal levels of ALT in the blood range from about 7-56 units/liter.
  • Normal levels of AST in the blood range from about 10-40 units/liter.
  • Normal levels of GGT in the blood range from about 9-48 units/liter.
  • Normal levels of alkaline phosphatase in the blood range from about 53-128 units/liter for a 20- to 50-year-old man and about 42-98 units/liter for a 20- to 50-year-old woman.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof reduces level of AST, ALT, and/or GGT in an individual having elevated AST, ALT, and/or GGT levels.
  • the level of ALT is reduced at least 2-, at least 3-, at least 4-, or at least 5-fold. In some embodiments, the level of ALT is reduced about
  • the level of AST is reduced at least 2-, at least 3-, at least 4-, or at least 5-fold. In some embodiments, the level of AST is reduced about 1.5 to about
  • the level of GGT is reduced at least 2, at least 3, at least 4, or at least 5-fold. In some embodiments, the level of GGT is reduced about 1.5 to about 3-fold.
  • administration of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a subject does not substantially change the level of low-density lipoprotein (LDL) (e.g., serum level of LDL) in the subject.
  • LDL low-density lipoprotein
  • administration of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to a subject results in a reduced NAFLD Activity (NAS) score.
  • NAS NAFLD Activity
  • steatosis, inflammation, and/or ballooning is reduced upon treatment.
  • the compounds disclosed herein reduce liver fibrosis.
  • the compounds reduce serum triglycerides.
  • the compounds reduce liver triglycerides.
  • the patient is at risk of developing an adverse effect prior to administering the compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the adverse effect is an adverse effect which affects the kidney, lung, heart, and/or skin.
  • the adverse effect is pruritus.
  • the patient has had one or more prior therapies.
  • the liver disorder progressed during the therapy.
  • the patient has had one or more prior therapies with another FXR agonist other that a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the patient suffered from pruritus during at least one of the one or more prior therapies.
  • the therapeutically effective amount is below the level that induces an adverse effect in the patient, such as below the level that induces pruritus, such as grade 2 or grade 3 pruritus.
  • Formula (I), or a pharmaceutically acceptable salt thereof is 500 pg/day - 600 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 500 pg/day - 300 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 500 pg/day - 150 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 500 pg/day - 100 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 500 pg/day - 20 mg/day. In some embodiments, the therapeutically effective amount of the compound of
  • Formula (I), or a pharmaceutically acceptable salt thereof is 1 mg/day - 600 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 1 mg/day - 300 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 1 mg/day - 150 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 1 mg/day - 100 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 1 mg/day - 20 mg/day.
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is 5 mg/day - 300 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 5 mg/day - 150 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 5 mg/day - 100 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 5 mg/day - 20 mg/day.
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is 5 mg/day - 15 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 25 mg/day - 300 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 25 mg/day - 150 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 25 mg/day - 100 mg/day.
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is 500 pg/day - 5 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 500 pg/day - 4 mg/day. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 5 mg/day - 600 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 75 mg/day - 600 mg/day.
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is about 1 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 2 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 5 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 10 mg/day. In another embodiment, the therapeutically effective amount is about 15 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 25 mg/day.
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is about 75 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 200 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 400 mg/day. In another embodiment, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is about 600 mg/day.
  • the administration comprises administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, daily for a treatment period of one or more weeks. In some embodiments, the administration comprises administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, twice daily for a treatment period of one or more weeks. In some embodiments, the administration comprises administering the compound of Formula (I), or a pharmaceutically acceptable salt thereof, every other day for a treatment period of one or more weeks.
  • the amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, administered on day 1 of the treatment period is greater than or equal to the amount administered on all subsequent days of the treatment period. In some embodiments, the amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, administered on day 1 of the treatment period is equal to the amount administered on all subsequent days of the treatment period.
  • the treatment period is at least 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 1 year, 2 years, 3 years, 4 years, or more. In some embodiments, the treatment period is from about a week to about a month, from about a month to about a year, from about a year to about several years.
  • the treatment period at least any of about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 1 year, 2 years, 3 years, 4 years, or more. In some embodiments, the treatment period is the remaining lifespan of the patient.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered with rifampicin.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered as a monotherapy, i.e., administered in absence of another agent, which: is useful in treating or substantially treating a liver disorder, impedes or slows the progression of non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH); or impedes or slows the progression of NASH, in a patient in need thereof.
  • the method does not comprise treating pruritus in the patient.
  • the method does not comprise administering an antihistamine, an immunosuppressant, a steroid (such as a corticosteroid), rifampicin, an opioid antagonist, or a selective serotonin reuptake inhibitor (SSRI).
  • an antihistamine such as a corticosteroid
  • a steroid such as a corticosteroid
  • rifampicin such as a corticosteroid
  • opioid antagonist such as a selective serotonin reuptake inhibitor (SSRI).
  • SSRI selective serotonin reuptake inhibitor
  • the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered once daily. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered twice daily. In some embodiments, the therapeutically effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is 75 mg - 200 mg twice daily per patient. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered as a pharmaceutically acceptable composition comprising at least one pharmaceutically acceptable excipient, carrier, or diluent.
  • the administration modulates one or more of the following: a metabolic pathway, bile secretion, retinol metabolism, drug metabolism-cytochrome P450, fat digestion and absorption, glycerolipid metabolism, chemical carcinogenesis, glyceropholipid metabolism, nicotine addiction, linoleic acid metabolism, ABC transporters, metabolism of xenobiotics by cytochrome P450, sphingolipid metabolism, glutathione metabolism, folate biosynthesis, morphine addiction, glycosphingolipid biosynthesis-lacto and neolacto series, arachidonic acid metabolism, tyrosine metabolism, maturity onset diabetes of the young, DNA replication, cholesterol metabolism, drug metabolism-other enzymes, and ether lipid metabolism.
  • a metabolic pathway bile secretion, retinol metabolism, drug metabolism-cytochrome P450, fat digestion and absorption
  • glycerolipid metabolism chemical carcinogenesis
  • glyceropholipid metabolism nicotine addiction
  • linoleic acid metabolism ABC transport
  • -the administration modulates one or more of the following: a metabolic pathway, retinol metabolism, fat digestion and absorption, glycerolipid metabolism, chemical carcinogenesis, glyceropholipid metabolism, ABC transporters, metabolism of xenobiotics by cytochrome P450, sphingolipid metabolism, glutathione metabolism, folate biosynthesis, and morphine addiction.
  • the administration modulates expression of one or more of the following: Abcb4, Apoa5, Cyp7al, Cyp8bl, Nr0b2, and Sic51b.
  • the compound utilized herein is of Formula (I): or a tautomer thereof, or an isotopomer of each thereof, or an enantiomer or diastereomer of the foregoing, or a pharmaceutically acceptable salt of each of the above.
  • the compound utilized herein is the compound of Formula (I).
  • the compound utilized herein is a pharmaceutically acceptable salt of the compound of Formula (I).
  • the compounds utilized herein may be prepared by a combination of a variety of stepwise procedures known in the art, such as, e.g., US 2010/0152166 (incorporated herein by reference).
  • the compound of Formula (I) is 6- ⁇ 4-[5-cyclopropyl-3-(2,6- dichloro-phenyl)-isoxazol-4-ylmethoxy]-piperidin4-yl ⁇ -l-methyl-lH-indole-3-carboxylic acid: or a pharmaceutically acceptable salt or enantiomer thereof.
  • a compound as detailed herein may in some aspects be in a purified form and compositions comprising a compound in purified forms are detailed herein.
  • Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds.
  • a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • substantially pure intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof.
  • a composition of a substantially pure compound intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound or a salt thereof.
  • the composition contains no more than 25% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 20% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 10% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 5% impurity.
  • a composition of substantially pure compound or a salt thereof wherein the composition contains no more than 3% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 1% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 0.5% impurity.
  • a composition of substantially pure compound means that the composition contains no more than 15%, such as no more than 10%, no more than 5%, no more than 3%, or no more than 1% impurity, which impurity may be the compound in a different stereochemical form.
  • a composition of substantially pure (S) compound means that the composition contains no more than 15% or no more than 10% or no more than 5% or no more than 3% or no more than 1% of the (R) form of the compound.
  • the compounds herein are synthetic compounds prepared for administration to an individual such as a human.
  • compositions are provided containing a compound in substantially pure form.
  • the disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier or excipient.
  • compositions or pharmaceutical compositions for use in any of the methods of provided herein are suitable for any compound or form thereof detailed herein.
  • Pharmaceutical compositions of any of the compounds detailed herein are embraced by this disclosure.
  • the disclosure includes pharmaceutical compositions comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid.
  • Pharmaceutical compositions according to the invention may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
  • the compound or pharmaceutical composition may be formulated for any available delivery route, including an oral, mucosal (e.g ., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form.
  • oral e.g ., nasal, sublingual, vaginal, buccal or rectal
  • parenteral e.g., intramuscular, subcutaneous or intravenous
  • topical or transdermal delivery form e.g., topical or transdermal delivery form.
  • a compound or pharmaceutical composition may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g, nasal spray or inhalers), gels, suspensions (e.g, aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water- in-oil liquid emulsions), solutions and elixirs.
  • suitable carriers include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices
  • the compound described herein can be used in the preparation of a composition, such as a pharmaceutical composition, by combining the compound as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above.
  • a pharmaceutically acceptable carrier such as those mentioned above.
  • the carrier may be in various forms.
  • pharmaceutical compositions may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • compositions comprising the compound may also contain other substances which have valuable therapeutic properties.
  • Pharmaceutical compositions may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g, in Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, 21 st ed. (2005), which is incorporated herein by reference.
  • Compounds as described herein may be administered to individuals (e.g, a human) in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions.
  • oral compositions such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions.
  • carriers which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc.
  • Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid polyols, and so on.
  • pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • any of the compounds described herein can be formulated in a tablet in any dosage form described.
  • compositions comprising a compound provided herein are also described.
  • the composition comprises a compound and a pharmaceutically acceptable carrier or excipient.
  • a composition of substantially pure compound is provided.
  • a pharmaceutically acceptable composition comprising a compound of Formula (I), or a tautomer thereof, or an isotopomer of each thereof, or an enantiomer or diastereomer of the foregoing, or a pharmaceutically acceptable salt of each of the above, and at least one pharmaceutically acceptable excipient, carrier, or diluent for treating a liver disorder; for impeding or slowing the progression of non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH); or for impeding or slowing the progression of NASH, in a patient in need thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), NAFLD, and NASH.
  • NASH non-alcoholic fatty liver disease
  • PSC primary sclerosing
  • a unit dose form of the pharmaceutically acceptable formulations provided herein comprises a therapeutically effective amount of a compound of Formula (I). In some embodiments, the unit dose form comprises a therapeutically effective amount of a compound of Formula (I) and a therapeutically effective amount of another agent.
  • the unit dose form is for treating a liver disorder; for impeding or slowing the progression of non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH); or for impeding or slowing the progression of NASH, in a patient in need thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), NAFLD, and NASH.
  • the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), NAFLD, and NASH.
  • PSC primary sclerosing cholangitis
  • PBC primary biliary cir
  • the manufacture of a medicament is for the treatment of a liver disorder selected from the group consisting of liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • a liver disorder selected from the group consisting of liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • a liver disorder selected from the group consisting of liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic
  • a method of treating a liver disorder in a patient in need thereof comprising administering a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (
  • X Receptor (FXR) agonist comprising administering a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I) pharmaceutically acceptable salt thereof, and wherein the patient has discontinued one or more prior therapies with another FXR agonist other than a compound of Formula (I) or a pharmaceutically acceptable salt thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH).
  • FXR X Receptor
  • a method of impeding or slowing the progression of non- alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH) in a patient in need thereof comprising administering a therapeutically effective amount of a compound of Formula (I) pharmaceutically acceptable salt thereof.
  • a method of impeding or slowing the progression of NASH in a patient in need thereof comprising administering a therapeutically effective amount of a compound of Formula
  • X Receptor (FXR) agonist that preferentially concentrates in liver tissue over one or more of kidney, lung, heart, and skin tissues
  • the method comprising administering a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I) pharmaceutically acceptable salt thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH) [0123] 7.
  • the administration does not result in pruritus in the patient greater than Grade 2 in severity.
  • a method of treating a liver disorder with an FXR agonist that does not result in detectable pruritus in a patient in need thereof comprising administering a therapeutically effective amount of the FXR agonist, wherein the FXR agonist is a compound of Formula (I) pharmaceutically acceptable salt thereof, wherein the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD), and non-alcoholic steatohepatitis (NASH)
  • the liver disorder is selected from liver inflammation, liver fibrosis, alcohol induced fibrosis, steatosis, alcoholic steatosis, primary sclerosing cholangitis (PSC), primary biliary cirrhosis (PBC), non-alcoholic fatty liver disease (NAFLD),
  • Formula (I), or a pharmaceutically acceptable salt thereof, administered on day 1 of the treatment period is greater than or equal to the amount administered on all subsequent days of the treatment period.
  • Compound I refers to the compound of Formula (I) Example 1: In Vitro Metabolic Stability
  • the rate of hepatic metabolism of Compound I was assessed in cryopreserved hepatocytes to determine the in vitro half-life of the compound. 1 mM of Compound I was mixed with preconditioned mouse, rat, dog, monkey, or human hepatocytes (0.5 x 10 6 cells/mL) and allowed to incubate at 37 °C for 2 hours, with samples collected at several time points and assayed for Compound I.
  • a polarized monolayer of MDCK-II cells grown on a permeable support was used to test the ability of organic-anion-transporting polypeptide (OATP) 1B1 or OATP 1B3 to transport Compound I across the lipid bilayer and into the cells.
  • the MDCK-II cells were transfected one of (1) a vector to express OATP 1B1, (2) a vector to express OATP 1B3, or (3) a control vector. Expression was induced in the cells before culturing the cells at 37 °C in 5% CO2 atmosphere. After inducing expression, the cells were treated with 1 mM, 3 pM, and 10 pM Compound I, or 3 mM Compound I and 100 mM rifampin. Cellular uptake of Compound I was then measured. Results from this experiment demonstrated that Compound I is not an OATP 1B1 or OATP 1B3 substrate.
  • Compound I for oral administration to SD rats was formulated in a vehicle containing 10% DMSO, 10% Cremophor-EL, and 80% aqueous solution (10% 2-hydroxypropyl-P-cyclodextrin).
  • Compound I for oral administration to beagle dogs was formulated with an aqueous solution containing 1% carboxymethyl cellulose, 0.25% Tween-80, and 0.05% antifoam.
  • Compound I for oral administration to cynomolgus monkeys was formulated with 10% Solutol, 20% PEG400, 0.5% Tween-80 and 69.5% deionized water.
  • Serial blood samples were collected, and plasma concentrations of the Compound I were measured. Results are shown in FIG. 1 A (IV administration) and FIG. IB (oral administration), and in Table 2.
  • the results demonstrate that Compound I has low to moderate clearance in vivo.
  • the volume of distribution (Vdss) of Compound I is greater than the volume of total body water (0.70 L/kg) in rat and dog. Smaller V dss in monkeys is correlated with higher plasma protein binding.
  • Tissue distribution of Compound I administered to rats was determined and compared to distribution other Farnesoid X Receptor (FXR) agonists cilofexor, tropifexor, and obeticholic acid (OCA).
  • FXR Farnesoid X Receptor
  • OCA obeticholic acid
  • Blood, liver, kidney, and lung tissue samples were collected from the rats to determine a tissue/plasma ratio.
  • the liver tissue/plasma ratio for the compounds is shown in FIG. 2A, which demonstrates that substantially more of Compound I localizes to the liver tissue compared to the other tested compounds.
  • Radiolabeled ( 14 C) Compound I was also administered to Long-Evans rats at an oral dose of 5 mg/kg (100 pCi/kg). Plasma, liver, small intestine, cecum, kidney, lung, heart and skin tissue samples were collected up to 168 hours, and the amount of radioactive material at various time points was measured. Results are shown in FIG. 3. Liver, small intestine, and cecum had the most radioactive material.
  • PK/PD Pharmaeokinetics/pharmacodynamics
  • RNAseq analysis mRNA was extracted from total liver and sequenced using standard Illumina library preparation and sequencing protocols. Differentially expressed genes (DEG) were determined using RSEM and edgeR software packages and analyzed using Adicha Bio’s iPathwayGuide software. Results are shown in FIG. 7A-7D, which indicate that Compound I modulates a significantly higher number of genes and metabolic pathways relevant to NASH compared to OCA.
  • FIG. 7A shows that administration of Compound I modulates expression of 500 NASH-related genes, OCA modulates expression of 44 NASH-related genes, including 37 common NAS-related genes modulated by both Compound I and OCA, relative to vehicle control (fold change > 1.5; q-value ⁇ 0.05).
  • FIG. 7A shows that administration of Compound I modulates expression of 500 NASH-related genes, OCA modulates expression of 44 NASH-related genes, including 37 common NAS-related genes modulated by both Compound I and OCA, relative to vehicle control (fold change > 1.5; q-value ⁇
  • FIG. 7B shows average expression levels (as shown by CPM value) of select FXR-related genes in vehicle, OCA, and Compound I treated mice.
  • FIG. 7C shows that administration of Compound I causes enrichment of 32 global pathways and that administration of OCA causes enrichment of 6 global pathways, including 2 common global pathways to both Compound I and OCA administration.
  • FIG. 7D shows the 25 pathways most statistically enriched upon Compound I administration, and compares the enrichment of those pathways to the enrichment upon OCA administration.
  • RNAseq analysis of livers from mice treated with Compound I showed a more robust modulation of FXR-related genes and metabolic pathways relevant to non-alcoholic fatty liver disease compared to OCA treatment.
  • mice C57/BL6J mice were fed a high fat diet (D12492, Research Diet, fat/protein/carbohydrate 60/20/20 Kcal%, lOw) to induce obesity (>36g mouse) prior to daily oral Compound I and biweekly intraperitoneal carbon tetrachloride (CCU) treatment for four weeks.
  • FIG. 8. Compound I was administered at a dose of 10, 30, and 100 mg/kg.
  • Nonalcoholic Fatty Liver Disease Activity Score is a composite score used to assess NASH.
  • NAS is calculated based upon liver steatosis, inflammation, and ballooning and was determined by analysis of liver tissue histology using H&E stain. Specifically, inflammation score was calculated based upon H&E staining: Score 0, none; 1, ⁇ 2 foci per 200X field; 2, 2-4 foci per 200X field; 3, >4 foci per 200X field.
  • Steatosis score was calculated by H&E staining as follows: Score 0, ⁇ 5%; 1,5-33%; 2, >33-66%; 3, >66%).
  • Hepatocellular ballooning is a form of liver cell injury associated with cell swelling and is also measured by H&E stained liver sections. The ballooning score is calculated as follows: 0-no hepatocyte ballooning; 1-few ballooning hepatocytes; 2-many hepatocytes with prominent ballooning.
  • mice treated with 10, 30, or 100 mg/kg Compound I had a significantly lower NAS score as compared to untreated NASH mice. Treatment with Compound I also significantly reduced steatosis, inflammation and ballooning compared to untreated NASH mice.
  • FIG. 10A-C mice treated with 10, 30, or 100 mg/kg Compound I had a significantly lower NAS score as compared to untreated NASH mice. Treatment with Compound I also significantly reduced steatosis, inflammation and ballooning compared to untreated NASH mice.
  • FIG. 11 A shows representative histology for healthy mice, NASH mice, and NASH mice treated with Compound I at 100 mg/kg.
  • FIG. 1 IB shows quantification of the fibrosis area of mice treated with Compound I.
  • Treatment with 10, 30 or 100 mg/kg Compound I resulted in statistically significant reduced fibrosis compared to untreated NASH control.
  • Compound I administered at 10, 30, or 100 mg/kg resulted in decreased collagen, type 1, alpha 1 expression in the liver as compared to control NASH mice.
  • ALT alanine amino transferase
  • AST aspartate amino transferase
  • triglyceride total cholesterol levels.
  • FIG. 12A and FIG. 12B serum ALT and AST levels were reduced in mice treated with Compound I.
  • FIG. 12C shows a statically significant reduction in serum triglyceride concentration in mice treated with 100 mg/kg Compound I.
  • FIG. 12D shows statistically significant reduction of total cholesterol level in mice treated with 10, 30, and 100 mg/kg Compound I.
  • FIG. 13 A shows the concentration of liver triglycerides in control mice or mice treated with 10, 30, or 100 mg/kg Compound I. Mice treated with 100 mg/kg Compound I showed statistically significant reduced triglyceride levels.
  • FIG. 13B shows a representative histology section.
  • EC50 concentration of Compound I for FXR was determined by a fluorescence-based FXR coactivation assay.
  • Half-log serial dilutions of Compound I or OCA (obeticholic acid, a known FXR agonist) (10pM-3nM) were incubated with human FXR ligand binding domain produced in Sf9 insect cells, labeled coactivator SRC-1 peptide and TR-FRET Coregulator Buffer G for lh at 25°C.
  • TGR5 activity was measured using a cell-based cAMP assay. See Kawamata et al JBC 278 (11)935-440 (2003).
  • Half-log serial dilutions of Compound I or OCA (10mM-3hM) were added to Chinese Hamster Ovary cells expressing recombinant human TGR5. After 30min at RT, cAMP was measured using an HTRF readout. EC50 values for FXR- regulated gene expression were determined using a cell-based RNA assay. Half-log serial dilutions of Compound I or OCA (3mM-3hM) were added to human HuH7 hepatoma cells.
  • SHP small heterodimer partner
  • BSEP bile salt export pump
  • FGF-19 fibroblast growth factor 19
  • Compound l is a potent and selective FXR agonist.
  • Compound l is a potent and selective FXR agonist.
  • Compound I reduced expression of inflammatory and fibrosis related genes and strongly suppressed liver steatosis, inflammation, ballooning, and fibrosis in a mouse model of NASH.
  • FIGs. 16A and 16B show changes of mean serum levels of 7a-C4 from the pre-dose baseline in different dosing groups on Day 1 and Day 7, respectively.
  • Subjects in the placebo group experienced a post prandial 7a-C4 spike which was inhibited in Compound I dosed subjects. After a single dose on Day 1, a partial suppression of serum 7a-C4 levels was observed in all Compound I dosed groups. On Day 7, maximum suppression of serum 7a-C4 levels was observed six hours after dosing corresponding to a reduction of 74%, 82%, and 91% in the 25, 75, and 150 mg dosing groups, respectively.
  • FIGs. 17A and 17B show changes of mean serum levels of FGF-19 from the pre-dose baseline in different dosing groups on Day 1 and Day 7, respectively. Maximum increases from baseline of 718%, 486%, and 454% in serum FGF 19 levels were observed at four hours after dosing on Day 7.
  • FIG. 18 shows changes of mean serum levels of low-density lipoprotein (LDL) from the pre-dose baseline in different dosing groups. Serum LDL changes above baseline were minimal in Compound I dosed groups and did not exceed the LDL elevations observed in subjects dosed with placebo.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention concerne des méthodes et des compositions pour le traitement de troubles hépatiques, y compris, sans caractère limitatif la stéatohépatite non alcoolique, des symptômes et des manifestations de celle-ci, chez un patient.
EP20886094.0A 2019-11-08 2020-11-06 Traitement de troubles hépatiques Pending EP4054567A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962933277P 2019-11-08 2019-11-08
US202063004403P 2020-04-02 2020-04-02
PCT/US2020/059522 WO2021092474A1 (fr) 2019-11-08 2020-11-06 Traitement de troubles hépatiques

Publications (2)

Publication Number Publication Date
EP4054567A1 true EP4054567A1 (fr) 2022-09-14
EP4054567A4 EP4054567A4 (fr) 2024-01-10

Family

ID=75849579

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20886094.0A Pending EP4054567A4 (fr) 2019-11-08 2020-11-06 Traitement de troubles hépatiques

Country Status (11)

Country Link
EP (1) EP4054567A4 (fr)
JP (1) JP2023501386A (fr)
KR (1) KR20220098168A (fr)
CN (1) CN114667142A (fr)
AU (1) AU2020380968A1 (fr)
BR (1) BR112022008639A2 (fr)
CA (1) CA3160445A1 (fr)
CL (1) CL2022001167A1 (fr)
IL (1) IL292459A (fr)
MX (1) MX2022005407A (fr)
WO (1) WO2021092474A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20230119130A (ko) 2020-12-14 2023-08-16 리제너론 파마슈티칼스 인코포레이티드 인히빈 서브유닛 베타 e(inhbe) 억제제로 대사 장애 및 심혈관 질환을 치료하는 방법

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200906823A (en) * 2007-07-16 2009-02-16 Lilly Co Eli Compounds and methods for modulating FXR
US20140187633A1 (en) * 2012-12-31 2014-07-03 Amarin Pharmaceuticals Ireland Limited Methods of treating or preventing nonalcoholic steatohepatitis and/or primary biliary cirrhosis
TW201733582A (zh) * 2016-03-28 2017-10-01 大日本住友製藥股份有限公司 Fxr促效劑及arb之組合醫藥
WO2018170173A1 (fr) * 2017-03-15 2018-09-20 Metacrine, Inc. Agonistes du récepteur farnésoïde x et leurs utilisations
US20210121493A1 (en) * 2017-07-25 2021-04-29 Cedars-Sinai Medical Center Methods for treating liver diseases
US20210244744A1 (en) * 2018-08-30 2021-08-12 Terns Pharmaceuticals, Inc. Treating liver disorders

Also Published As

Publication number Publication date
MX2022005407A (es) 2022-05-24
CA3160445A1 (fr) 2021-05-14
BR112022008639A2 (pt) 2022-07-19
JP2023501386A (ja) 2023-01-18
IL292459A (en) 2022-06-01
AU2020380968A1 (en) 2022-05-12
WO2021092474A1 (fr) 2021-05-14
CN114667142A (zh) 2022-06-24
KR20220098168A (ko) 2022-07-11
EP4054567A4 (fr) 2024-01-10
CL2022001167A1 (es) 2023-02-10

Similar Documents

Publication Publication Date Title
US20240000765A1 (en) Combination treatment of liver disorders
Davis Cholestasis and endogenous opioids: liver disease and exogenous opioid pharmacokinetics
US20210379040A1 (en) Combination treatment of liver disorders
US10940125B2 (en) Methods and compositions for the treatment of steatosis-associated disorders
US20230181583A1 (en) Treating liver disorders with an ssao inhibitor
US20090093509A1 (en) Methods and Compositions for the Treatment of Pruritus
EP3743064B1 (fr) Traitement de la protéinurie
US20220387414A1 (en) Treating liver disorders
WO2021092474A1 (fr) Traitement de troubles hépatiques
TW202327589A (zh) 肝病之組合療法
EP4166137A1 (fr) Agent thérapeutique pour la stéatose hépatique non alcoolique
EP4223291A1 (fr) Composition pour le traitement de maladies médiées par le canal kca3.1 comprenant un composé de phénylalkylcarbamate
WO2023220404A1 (fr) Traitement de stéatohépatite non-alcoolique
Haddley Lu-AA-21004
EP1846105A2 (fr) Utilisation d'antagonistes alpha-2b pour le traitement de symptomes vasomoteurs

Legal Events

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

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

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

Free format text: ORIGINAL CODE: 0009012

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220511

AK Designated contracting states

Kind code of ref document: A1

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

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20231212

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 413/12 20060101ALI20231206BHEP

Ipc: C07D 261/08 20060101ALI20231206BHEP

Ipc: A61K 31/4545 20060101ALI20231206BHEP

Ipc: A61K 31/42 20060101AFI20231206BHEP