AU2019376907A1 - Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients - Google Patents

Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients Download PDF

Info

Publication number
AU2019376907A1
AU2019376907A1 AU2019376907A AU2019376907A AU2019376907A1 AU 2019376907 A1 AU2019376907 A1 AU 2019376907A1 AU 2019376907 A AU2019376907 A AU 2019376907A AU 2019376907 A AU2019376907 A AU 2019376907A AU 2019376907 A1 AU2019376907 A1 AU 2019376907A1
Authority
AU
Australia
Prior art keywords
aramchol
fibrosis
subject
inhibitors
score
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
AU2019376907A
Inventor
Allen BAHARAFF
Tali GORFINE
Liat Hayardeny-Nissimov
Jose M. Mato De La Paz
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.)
Galmed Research and Development Ltd
Original Assignee
Galmed Research and Development Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/182,767 external-priority patent/US20190175619A1/en
Application filed by Galmed Research and Development Ltd filed Critical Galmed Research and Development Ltd
Publication of AU2019376907A1 publication Critical patent/AU2019376907A1/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • 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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration

Landscapes

  • Health & Medical Sciences (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)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (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)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention relates to the treatment, inhibition and reduction of fibrosis, including hepatic fibrosis. More specifically, the invention provides compositions and methods useful for the treatment and inhibition of fibrotic disorders, hepato-fibrotic conditions associated with Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH), employing the use of 3β-arachidylamido-7α,12α-dihydroxy-5β-cholan-24-oic acid (Aramchol

Description

TREATMENT AND INHIBITION OF FIBROSIS IN NON-ALCOHOLIC FATTY
LIVER DISEASE PATIENTS
FIELD OF THE INVENTION
[0001] The present invention relates to unit dosage form of 3P-arachidylamido-7a,12a-dihydroxy- 5 -cholan-24-oic acid (Aramchol™) and use thereof in the treatment and/or inhibition of fibrosis.
BACKGROUND OF THE INVENTION
[0002] Aramchol™ is an amide conjugate of arachidic acid and 3-aminocholic acid, effective in reducing liver fat content as well as improving metabolic parameters associated with fatty liver disease. It is an oral, liver-targeted, fatty acid-bile acid conjugate that down-regulates stearoyl CoA type 1 desaturase. It belongs to a family of synthetic Fatty-Acid/Bile-Acid Conjugates (FABACs) and is being developed as a potentially disease modifying treatment for fatty liver disease and Non Alcoholic SteatoHepatitis (NASH).
[0003] Aramchol™ is chemically named 3P-arachidylamido-7a,12a-dihydroxy-5P-cholan-24-oic acid (or (4R)-4-((3S,5S,7R,10S,12S,13R,17R)-7,12-dihydroxy-3-icosanamido-10,13- dimethylhexadecahydro-lH-cyclopenta[a]phenanthren-17-yl)pentanoic acid, (IUPAC name)), and is represented by the following chemical structure:
[0004] Aramchol™, processes for its preparation, and use thereof are disclosed in U.S. 6,384,024;
U.S. 6,395,722; U.S. 6,589,946; U.S. 7,501,403; U.S. 8,110,564; U.S. 2012/0214872; and WO 2009/060452. Fibrosis
[0005] The formation of fibrous connective tissue is part of the normal healing process following tissue damage due to injury or inflammation. During this process, activated immune cells including macrophages stimulate the proliferation and activation of fibroblasts, which in turn deposit connective tissue. However, abnormal or excessive production of connective tissue may lead to accumulation of fibrous material such that it interferes with the normal function of the tissue. Fibrotic growth can proliferate and invade healthy surrounding tissue, even after the original injury heals. Such abnormal formation of excessive connective tissue, occurring in a reparative or reactive process, is referred to as fibrosis.
[0006] Physiologically, fibrosis acts to deposit connective tissue, which can obliterate the architecture and function of the underlying organ or tissue. Defined by the pathological accumulation of extracellular matrix (ECM) proteins, fibrosis results in scarring and thickening of the affected tissue, which interferes with normal organ function. In various conditions, the formation of fibrotic tissue is characterized by the deposition of abnormally large amounts of collagen. The synthesis of collagen is also involved in a number of other pathological conditions. For example, clinical conditions and disorders associated with primary or secondary fibrosis, such as systemic sclerosis, graft-versus host disease (GVHD), pulmonary fibrosis and autoimmune disorders, are distinguished by excessive production of connective tissue, which results in the destruction of normal tissue architecture and function. These diseases can best be interpreted in terms of perturbations in cellular functions, a major manifestation of which is excessive collagen synthesis and deposition. The role of collagen in fibrosis has prompted attempts to develop drugs that inhibit its accumulation.
Hepatic Fibrosis
[0007] Fibrosis of the liver, also referred to herein as hepatic fibrosis, may be caused by various types of chronic liver injury, especially if an inflammatory component is involved. Self -limited, acute liver injury (e.g., acute viral hepatitis A), even when fulminant, does not necessarily distort the scaffolding architecture and hence does not typically cause fibrosis, despite loss of hepatocytes. However, factors such as chronic alcoholism, malnutrition, hemochromatosis, and exposure to poisons, toxins or drugs, may lead to chronic liver injury and hepatic fibrosis due to exposure to hepatotoxic chemical substances. Hepatic scarring, caused by surgery or other forms of injury associated with mechanical biliary obstruction, may also result in liver fibrosis. [0008] Fibrosis itself is not necessarily symptomatic, however it can lead to the development of portal hypertension, in which scarring distorts blood flow through the liver, or cirrhosis, in which scarring results in disruption of normal hepatic architecture and liver dysfunction. The extent of each of these pathologies determines the clinical manifestation of hepato-fibrotic disorders. For example, congenital hepatic fibrosis affects portal vein branches, largely sparing the parenchyma. The result is portal hypertension with sparing of hepatocellular function.
Treatment
[0009] Attempts to develop anti-fibrotic agents for the treatment of various disorders have been reported. However, treatment of established fibrosis, formed after months or years of chronic or repeated injury, remains a challenge. In its initial stages, hepatic fibrosis may regress if the cause is reversible (e.g. with viral clearance). Thus, the majority of available treatment options are designed to remove the basis of the liver injury, such as by eliminating hepatitis B virus or hepatitis C virus in chronic viral hepatitis, abstaining from alcohol in alcoholic liver disease, removing heavy metals such as iron in hemochromatosis or copper in Wilson disease, and decompressing bile ducts in biliary obstruction.
[0010] Treatments aimed at reversing the fibrosis are usually too toxic for long-term use (e.g. corticosteroids, penicillamine) or have no proven efficacy (e.g. colchicine). Silymarin, present in milk thistle, is a popular alternative medicine used to treat hepatic fibrosis, appears to be safe but to lack efficacy.
Potential Therapeutic agents
[0011] Attempts to develop specific anti-fibrotic agents for the treatment of liver diseases have been reported. For example, U.S. Pat. No. 8,729,046 relates to methods for treating fibrosis of a tissue, including fibrosis of the liver, using combinations of nucleic acids or nucleic acid analogs. Specifically, the nucleic acids or analogs thereof are targeted to microRNAs of the miR23b cluster. U.S. 6,562,829 discloses compositions for treating hepatic fibrosis and methods of using and manufacturing the composition, the composition comprising a quinazolinone derivative, preferably Halofuginone. U.S. 8,858,954 is directed to pharmaceutical composition for preventing and treating liver fibrosis or nonalcoholic fatty liver disease, comprising 50 to 90% by weight of Cordyceps sinensis mycelium powder, and 10 to 50% by weight of condensed astragalus powder.
[0012] U.S. Pub. No. 2015/359805 relates to Farnesoid X receptor (FXR) modulators which can be used for the treatment of cholestatic disorders, in particular to bile acid derivatives wherein the C6 contains an ethyl and the C24 carboxy group is transformed into a sulphate group. Among the disorders suggested to be treated are alcoholic liver disease, living donor transplant liver regeneration, congenital hepatic fibrosis, choledocholithiasis, and granulomatous liver disease. U.S. 2014/187633 is directed to methods of treating and/or preventing non-alcoholic seatohepatitis (NASH) and/or primary biliary cirrhosis comprising administering to a subject in need thereof a pharmaceutical composition comprising eicosapentaenoic acid or a derivative thereof. The FXR agonist, obeticholic acid, which is a modified bile acid, is in phase III clinical trials for primary biliary cirrhosis. Use of this drug has been reported to be commonly associated with side effects such as pruritus.
[0013] Ursodeoxycholic acid (UDCA, Ursodiol) is the most frequently used treatment for primary biliary cirrhosis. It is one of the secondary bile acids, which are metabolic byproducts of intestinal bacteria. The drug is considered to assist in reducing the cholestasis and improves blood test results (liver function tests). However it has a minimal effect on symptoms and whether it improves prognosis is controversial. To relieve itching caused by bile acids in circulation, which would normally be removed by the liver, cholestyramine (a bile acid sequestrant) may be prescribed to primary biliary cirrhosis patients. The agent may assist in absorbing bile acids in the gut to be eliminated, rather than re-enter the blood stream. Alternative agents include stanozolol, naltrexone and rifampicin.
[0014] Obeticholic acid (OCA, Ocaliva) is a semi-synthetic bile acid analogue undergoing development in phase 2 and 3 studies for specific liver and gastrointestinal conditions. The FDA granted accelerated approval to Ocaliva on 27 May 2016 for the treatment of primary biliary cholangitis (PBC) in combination with ursodeoxycholic acid (UDCA) in adults with an inadequate response to UDCA, or as a single therapy in adults unable to tolerate UDCA. In addition, a phase 2 trial in NASH patients showed that administration of OCA reduced markers of liver inflammation and fibrosis and increased insulin sensitivity.
[0015] WO 2014/197738 and WO 2016/094570 relate to small molecule compounds, disclosed to be inhibitors of myofibroblast trans-differentiation and activation. Drugs and combinations suggested for the treatment of inter alia fatty liver were disclosed, for example, in WO 2016/112305 and EP2632925 (acetyl-CoA carboxylase inhibitors) as well as WO 2016/154258 (dual PPAR delta / gamma agonists). Some of the disclosed compounds were suggested to be used in combination with various other drugs.
[0016] Many patients do not respond to available treatments for fibrotic disorders, and long-term treatment is limited by toxicity and side effects. Therefore, a need remains for developing therapeutic modalities aimed at reducing fibrosis, especially hepatic fibrosis. The development of safe and effective treatments for established cirrhosis and portal hypertension and for attenuating fibrosis would be highly beneficial.
SUMMARY OF THE INVENTION
Fatty Acid Bile Acid Conjugates
[0017] Fatty acid bile salt conjugates, referred to also as Fatty Acid Bile Acid Conjugates (F ABACs), are a family of synthetic molecules that may be used to improve conditions related to bile acids or cholesterol metabolism. FABACs are believed to lower blood cholesterol concentration, reduce liver fat levels and dissolve gallstones ( Gilat et ai, Hepatology 2003; 38: 436-442; and Gilat et ai, Hepatology 2002; 35: 597-600). FABAC include 3 -arachidylamido-7a,12a-dihydroxy-5 - cholan-24-oic acid, also known as Aramchol™.
[0018] US Patents 6,384,024, 6,395,722, 6,589,946 disclose certain FABACs and their use in dissolving cholesterol gallstones in bile and treating arteriosclerosis. These and additional FABACs were disclosed in US Patents 7,501,403, 8,975,246 and 8,110,564 for use in treating fatty liver, in reducing blood cholesterol levels and in treating hyperglycemia, diabetes, insulin resistance and obesity. Further therapeutic uses of FABACs are disclosed in Safadi et al. (Clin Gastroenterol Hepatol. 2014 Dec;12(12):2085-91) and in WO 2015/019358 and WO 2015/019359. Amine salts of certain FABACs are disclosed in WO 2015/083164.
[0019] The invention relates to the treatment, inhibition and reduction of fibrosis, including hepatic fibrosis. More specifically, embodiments of the invention provide compositions and methods useful for the treatment and inhibition of fibrotic disorders, hepato-fibrotic conditions associated with Non- Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH), employing the use of 3 -arachidylamido-7a,12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™) or a pharmaceutically acceptable salt thereof. In other embodiments, the treatment and inhibition of hepato-fibrotic conditions caused by contact with hepatotoxic chemical substances or by mechanical obstruction is contemplated.The invention provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3b- arachidylamido-7a, 12a-di hydroxy-5 b-chol an -24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein the average AUCO-12 is between about 40,000 to about 90,000ng/ml.
[0021] In some embodiments, the average AUCo-12 is about 55,000 ng/ml. [0022] The invention further provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3 -arachidylamido- 7a, l2a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein the average AUC0-24 is between about 80,000 to about 180,000ng/ml.
[0023] In some embodiments, the average AUC0-24 is about 110,000ng/ml.
[0024] The invention further provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3 -arachidylamido- 7a, 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein Cavg per 24h is between about 3000 to about 7000 ng/ml.
[0025] In some embodiments, smd C vg per 24h is nbout 4500 n^/ml.
[0026] In one embodiment, this invention provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to the subject a daily dose of between 400 mg and 900 mg of 3 -arachidylamido-7a, 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, said dose is administered in a dose regimen of at least twice a day, thereby treating hepatic fibrosis in said subject. In another embodiment, the daily dose amount is 600 mg or 800 mg. In another embodiment, each administered dose is 300 mg or 400 mg. In another embodiment, the Aramchol™ daily dose is 600 mg and it is administered twice a day wherein each administered dose is 300 mg. In another embodiment, 300 mg of Aramchol™ is administered to the subject every 12 hours. In another embodiment, the human subject is afflicted with Non-Alcoholic Fatty Liver Disease (NAFLD). In another embodiment, the human subject has a ballooning score of at least 1. In another embodiment, the human subject has an inflammation score of at least 1. In another embodiment, the human subject has a steatosis score of at least 1. In another embodiment, the human subject has a ballooning score of at least 1, an inflammation score of at least land a steatosis score of at least 1. In another embodiment, the human subject is afflicted with Diabetes Mellitus type II or pre-diabetes. In another embodiment, the human subject is afflicted with Non- Alcoholic Steatohepatitis (NASH). In another embodiment, the human subject is not afflicted with Non-Alcoholic Steatohepatitis (NASH).
[0027] In another embodiment, the method further comprises lack of worsening of the subject’s NAFLD as defined by Activity (NAS) score. In another embodiment, the method further comprises lack of worsening of the subject’s Steatosis, Activity, and Fibrosis (SAF) Activity score. In another embodiment, the method further comprises reduction of liver fat in said subject. In another embodiment, the method further comprises improvement in subject’s Steatosis. In another embodiment, the method further comprises improvement in subject’s ballooning. In another embodiment, the method further comprises NAFLD resolution. In another embodiment, the method further comprises NAFLD resolution without worsening of fibrosis. In another embodiment, the method further comprises reduction of fibrosis without NAFLD worsening. In another embodiment, the method further comprises reduction of ALT levels in said subject. In another embodiment, the method further comprises reduction of AST levels in said subject. In another embodiment, the method further comprises reduction of HbAlc levels in said subject. In another embodiment, the method further comprises lack of subject’s progression to Cirrhosis. In another embodiment, the method further comprises inhibiting progression of Non-Alcoholic Fatty Liver Disease (NAFLD) and/or Non- Alcoholic Steatohepatitis (NASH). In another embodiment, the human subject is afflicted with Non- Alcoholic Steatohepatitis (NASH) and the method further comprises inhibiting the progression of NASH in said subject. In another embodiment, the human subject is afflicted with Non-Alcoholic Steatohepatitis (NASH) and the method further comprises NASH resolution in the subject.
[0028] In one embodiment, this invention provides a method for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject a daily dose of between 400 mg and 900 mg of 3 b -arac h i dy I a m i do - 7 a. 12a-di hydroxy-5 P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, said dose is administered in a dose regimen of at least twice a day, thereby inhibiting the development of hepatic fibrosis in said subject. In another embodiment, the daily dose amount is 600 mg or 800 mg. In another embodiment, each administered dose is 300 mg or 400 mg. In another embodiment, the Aramchol™ daily dose is 600 mg and it is administered twice a day wherein each administered dose is 300 mg. in another embodiment, 300 mg of Aramchol™ is administered to the subject every 12 hours. In another embodiment, the human subject has a NAFLD Activity (NAS) Score of at least 4, at least 5, at least 6, or at least 7. In another embodiment, the human subject has a ballooning score of at least 1. In another embodiment, the human subject has an inflammation score of at least 1. In another embodiment, the human subject has a steatosis score of at least 1. In another embodiment, the human subject has a ballooning score of at least 1, an inflammation score of at least 1, and a steatosis score of at least 1. In another embodiment, the human subject is afflicted with Diabetes Mellitus type II or pre-diabetes. In another embodiment, the human subject is afflicted with Non-Alcoholic Steatohepatitis (NASH). In another embodiment, the human subject is not afflicted with Non-Alcoholic Steatohepatitis (NASH).
[0029] In another embodiment, the method further comprises lack of worsening of the subject’s NAFLD as defined by Activity (NAS) score. In another embodiment, the method further comprises lack of worsening of the subject’s Steatosis, Activity, and Fibrosis (SAF) Activity score. In another embodiment, the method further comprises reduction of liver fat in said subject. In another embodiment, the method further comprises improvement in subject’s Steatosis. In another embodiment, the method further comprises improvement in subject’s ballooning. In another embodiment, the method further comprises NAFLD resolution. In another embodiment, the method further comprises NAFLD resolution without worsening of fibrosis. In another embodiment, the method further comprises reduction of fibrosis without NAFLD worsening. In another embodiment, the method further comprises reduction of ALT levels in said subject. In another embodiment, the method further comprises reduction of AST levels in said subject. In another embodiment, the method further comprises reduction of HbAlc levels in said subject. In another embodiment, the method further comprises lack of subject’s progression to Cirrhosis. In another embodiment, the method further comprises inhibiting progression of Non-Alcoholic Fatty Liver Disease (NAFLD) and/or Non- Alcoholic Steatohepatitis (NASH). In another embodiment, the human subject is afflicted with Non- Alcoholic Steatohepatitis (NASH) and the method further comprise inhibiting the progression of NASH in said subject. In another embodiment, the human subject is afflicted with Non-Alcoholic Steatohepatitis (NASH) and the method further comprises NASH resolution in the subject.
[0030] In one embodiment, this invention provides a method for treating fibrosis other than hepatic fibrosis in a human subject afflicted with fibrosis comprising administering to the subject a daily dose of between 400 mg and 900 mg of 3 b- arach i dy I a m i do - 7 a. 12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, said dose is administered in a dose regimen of at least twice a day, thereby treating fibrosis in said subject, wherein said fibrosis is being caused by a factor selected from the group consisting of pulmonary fibrosis, heart fibrosis, kidney fibrosis, dermal fibrosis, ocular fibrosis, mucosal fibrosis, fibrosis of the central nervous system, fibrosis in bone or bone marrow, fibrosis in an endocrine organ, fibrosis in the gastro-intestinal system, mediastinal fibrosis, postfibrinous fibrosis, proliferative fibrosis, retroperitoneal fibrosis, pancreatic fibrosis, or fibrosis associated with an autoimmune disease. [0031] In another embodiment, the methods according to this invention further comprise administering a therapeutically effect amount of a pharmaceutical composition comprising at least one compound selected from the group consisting of: a) ethyl eicosapentanoate (EPA-E), eicosapentaenoic acid (EPA) and its pharmaceutically acceptable amides, salts, esters and phospholipids; b) an inhibitor of Acetyl-CoA carboxylase (ACC) alone, or in combination with one or more additional therapeutic agents; c) pioglitazone hydrochloride or an enantiopure deuterium-enriched pioglitazone; d) a peroxisome proliferator activated receptor (PPAR) delta and gamma dual agonists; and e) angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacylglycerol 0-acyltransferase 1 (DGAT1) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, farnesoid X receptor (FXR) agonists such as obeticholic acid and Px-104, FXR/TGR5 dual agonists, galectin-3 inhibitors such as LIPC-1010 and GR-MD-02, glucagon-like peptide (GLPI) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 1 1— hydroxysteroid dehydrogenase (I I— HSD I) inhibitors, IL-I- antagonists, IL-6 antagonists, IL-I 0 agonists, IL-I 7 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PLC) inhibitors, PPARa agonists, PPAR gamma agonists such as rosiglitazone and pioglitazone, metformin, pentoxyfylline, vitamin E, selenium, omega-3 fatty acids and betaine, PPAR8 agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturasel inhibitors, thyroid hormone receptor- agonists, tumor necrosis factor a (TNFa) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, PTPib inhibitors, ASKI inhibitors, and vascular adhesion protein-1 inhibitors such as PXS4728A, metformin, GR-MD-02, cysteamine bitartrate, simtuzumab, emricasan, GFT-505, CER-002, KD3010, KD3020, MBX8025, LUM002, RP-103, and cenicriviroc. BRIEF DESCRIPTION OF THE DRAWINGS
[0032] Figure 1. demonstrates the effect of Aramchol™ on liver cirrhosis by macroscopic evaluation. Figure 1A - saline control; Figure IB - treatment with TAA (20mg/100 gr body weight) twice per week during 10 weeks; Figure 1C - treatment with TAA and Aramchol™ 1 mg/kg; Figure ID - treatment with TAA and Aramchol™ 5 mg/kg.
[0033] Figure 2. demonstrates the effect of Aramchol™ on liver fibrosis by microscopic evaluation (following Masson Goldner staining). Figure 2A - averaged fibrotic score (TAA - black, Aramchol™ - white, OCA - gray); Figure 2B - representative samples (TAA only - left; TAA and Aramchol™ 1 mg/kg - middle; TAA and Aramchol™ 5 mg/kg - right).
[0034] Figure 3. depicts the effect of Aramchol™ on COL1A1 expression in LX -2 human hepatic stellate cells.“Ctrl SI, S2 and S3” represent control (saline-treated cells) in three separate experiments; “A SI, S2 and S3” represent the result of Aramchol™ treated cells in these experiments.
[0035] Figure 4. depicts the effect of Aramchol™ on PPAR-g expression in LX-2 human hepatic stellate cells.“Ctrl SI, S2 and S3” represent control (saline-treated cells) in three separate experiments; “A SI, S2 and S3” represent the result of Aramchol™ treated cells in these experiments.
[0036] Figure 5. depicts the effect of Aramchol™ on collagen production from LX-2 human hepatic stellate cells compared to a DMSO control.
[0037] Figure 6. depicts the effect of Aramchol™ on liver steatosis in 0.1 MCD diet. Figure 6A - histology staining using Sudan III; Figure 6B - quantification of Sudan III stained cells.
[0038] Figure 7. depicts the effect of Aramchol™ on macrophage activation and infiltration in 0.1 MCD diet. Figure 7A - histology staining - F4/80 and CD64; Figure 7B - quantification of F4/80 and CD64 positive cells.
[0039] Figure 8. depicts the effect of Aramchol™ on fibrosis in 0.1 MCD Diet (histology - sirius red). Figure 8A - histology staining using sirius red; Figure 8B - quantification of sirius red stained cells.
[0040] Figure 9. depicts the effect of Aramchol™ on collagen production using liver extract from 0.1 MCD mice. [0041] Figures 10A and 10B depict the effects of Aramchol™ on liver biochemistry in 0.1 MCD mice. Figure 10A - quantification of metabolites in liver of control (grey) and Aramchol™-treated (black) 0.1 MCD mice; Figure 10B - schematic of relevant liver biochemical pathway.
[0042] Figure 11 depicts the bioavailability of lOOmg, 300mg and 900mg doses, relative to
30mg dose in healthy volunteers. Relative oral bioavailability in healthy volunteers decreases with increasing dose across the range 30-900mg.
[0043] Figures 12A and 12B depict the AUC vs. dose (Figure 12A) and AUC/Dose vs. dose (Figure 12B) grouped by study. AUC for the Phase IFb study in circles is estimated from the observed Cmin on D84*24h.
[0044] Figure 13A depicts the predicted versus observed concentration (straight line represents the dose level and study). Figure 13B depicts the predicted versus observed concentration-time after dose (circles = observed, sold lines = model). Figure 13C depicts the data and model parameters.
[0045] Figure 14A is the simulated plasma exposure from a 300mg BID dose using the model parameters (Figure 14B) fit to the observed data at 300mg QD in patients (Tlag is the delay in absorption; V/F is the volume of the central compartment divided by the bioavailability; Cl/F is the clearance from the compartment divided by bioavailability; and Ka is the rate of absorption into the compartment).
[0046] Figures 15A and 15B present the simulated plasma exposure from a 600mg QD dose (Figure 15A) using the model parameters (Figure 15B) fit to the observed data at 300mg QD in patients with 75% lower bioavailability estimated from the non-linear exposure in healthy volunteers between 300 and 900 mg (resulting in higher V/F and Cl/F), (Tlag is the delay in absorption; V/F is the volume of the central compartment divided by the bioavailability; Cl/F is the clearance from the compartment divided by bioavailability; and Ka is the rate of absorption into the compartment).
[0047] Figures 16A and 16 B depict the reduction in liver fat in human subjects receiving 400 mg, 600 mg doses of Aramchol™ and placebo. Figure 16A is the mean absolute change from baseline in liver fat; Figure 16B is the MRI Responders Analysis: Reduction >5% absolute change from baseline. Liver function was significantly reduced vs. placebo, Reduction of >5% absolute change displays a dose response pattern.
[0048] Figures 17A and 17B depict the reduction in ballooning in human subjects receiving 400 mg, 600 mg doses of Aramchol™ and placebo. Figure 17A is the Ballooning Improvement (>l point); and Figure 17B is the Ballooning 0 at end of study. A statistically significant effect on ballooning was demonstrated with Aramchol™ 600mg.
[0049] Figures 18A and 18B depict the NASH Resolution without worsening of Fibrosis demonstrated in human subjects receiving 400 mg, 600 mg doses of Aramchol™ and placebo. Figure 18A is NASH Resolution without Worsening of Fibrosis; and Figure 18B is the NASH Resolution. Significantly more patients treated with Aramchol™ 600mg showed NASH resolution without worsening of fibrosis with a dose response pattern.
[0050] Figures 19A and 19B depict the fibrosis improvement and the progression to Cirrhosis as demonstrated in human subjects receiving 400 mg, 600 mg doses of Aramchol™ and placebo. Figure 19A depicts the fibrosis improvement (>1 stage) without worsening of NASH; and Figure 19B depicts the progression to Cirrhosis. More patients showed fibrosis improvement without worsening of NASH with a dose response pattern. Less patients progressed to cirrhosis with Aramchol™ 600mg.
[0051] Figures 20A and 20B depict the Aramchol™ effect on ALT and AST levels as demonstrated in human subjects receiving 400 mg, 600 mg doses of Aramchol™ and placebo. Figure 20A depicts the change from baseline in ALT levels (U/L) ; and Figure 20B depicts the change from baseline in AST levels (U/L) . Both 400 mg, and 600 mg doses significantly reduced ALT, AST levels in subjects vs. placebo in a dose response manner.
[0052] Figures 21 A and 21B depict how Aramchol™ downregulates SCD-1 mRNA and fibrogenic genes and upregulates PPARy mRNA in HSCs. Based on RT-PCR (N = 3) *p<0.05, **p<0.01,
***p<0.001.
[0053] Figures 22A and 22B depict how Aramchol™ downregulates SCD-1 in HSCs. Figure 22A and Figure 22B depict the Western Blot and the densitometry respectively of downregulated SCD-1 protein in HSCs treated with Aramchol™ for 24 or 48 hrs. (N=3) *p<0.05, **p<0.01, ***p<0.001
[0054] Figure 23 depicts the confirmation by RNASeq for downregulation of fibrogenic genes and
SCD-1 mRNA, and upregulation of PPARy mRNA after 48 hrs of treatment with Aramchol™. The cholesterol efflux regulatory protein, ABCA1, is also upregulated.
[0055] Figure 24 depicts that Aramchol™ does not affect PPARy mRNA expression in primary mouse hepatocytes after 48 hours of treatment. (N=3) *p<0.05, **p<0.01, ***p<0.001 [0056] Figures 25A and 25B depict how Aramchol™ downregulates cholesterol biosynthesis and collagen formation in HSCs. Gene Set Enrichment Analysis of LX -2 cells after 24 (Figure 25A) or 48
(Figure 25B) hours of treatment.
DETAILED DESCRIPTION OF THE INVENTION
Fatty Acid Bile Acid Conjugate (FABAC)
[0057] According to an embodiment of the invention, the combinations, compositions, methods and packages of the invention may comprise Aramchol™ in its free acid form. According to an embodiment of the invention, Aramchol™ is in its salt form. The salt may be an amine -based salt. The amine-based salt may be selected from the group consisting of meglumine, lysine and tromethamine salts.
[0058] Other embodiments of the invention relate to compositions, methods and packages employing the use of a Fatty Acid Bile Acid Conjugate (FABAC), or salts thereof. According to some embodiments, the FABAC is of Formula I:
W - X - G (I)
[0059] wherein G represents a bile acid or a bile salt radical thereof; W represents one or two fatty acid radicals having 6-22 carbon atoms; and X represents a bonding member selected from the group consisting of: a heteroatom, a direct C-C bond and a C=C bond; each possibility represents a separate embodiment of the present invention.
[0060] According to some embodiments, the bonding member is selected from the group consisting of: NH, P, S, O and a direct C-C or C=C bond; each possibility represents a separate embodiment of the present invention. According to some embodiments, the bonding member is NH.
[0061] According to some embodiments, each of said one or two fatty acid radicals is a radical of a fatty acid selected from the group consisting of: arachidylic acid, stearic acid, behenic acid, palmitic acid, arachidonic acid, eicosapentaenoic acid and oleic acid; each possibility represents a separate embodiment of the present invention. According to some embodiments, said one or two fatty acid radicals are radicals of arachidylic acid; each possibility represents a separate embodiment of the present invention.
[0062] According to some embodiments, W represents two fatty acid radicals, each independently comprises 6-22 carbon atoms; and wherein each of said fatty acid radicals is independently bound to a bonding member X selected from the group consisting of: a heteroatom, a direct C-C bond and a C=C bond. According to some embodiments, W represents a single fatty acid radical.
[0063] According to some embodiments, the bile acid is selected from the group consisting of: cholic acid, ursodeoxycholic acid, chenodeoxycholic acid, deoxycholic acid, lithocholic acid and derivatives thereof; each possibility represents a separate embodiment of the present invention. In another embodiment the bile acid is cholic acid, chenodeoxycholic acid, or deoxycholic acid. In another embodiment the bile acid is other than ursodeoxycholic acid and lithocholic acid. According to some embodiments, the bile acid is cholic acid.
Indications.
[0064] The invention is based, in part, on the surprising discovery that Aramchol™ exerts a potent anti-fibrotic effect, independent of its reported activities on fatty liver and steatosis, and reduces and inhibits the development of fibrosis in various experimental models. Specifically, treatment with Aramchol™ (5 mg/kg) significantly inhibited the development of toxin-induced cirrhosis, necrosis and liver fibrosis in an in vivo thioacetamide (TAA) model. Aramchol™ was also found to be unexpectedly superior to obaticholic acid (OCA), which did not induce statistically significant reduction in these parameters under the tested experimental conditions. In addition, Aramchol™ significantly reduced COL1A1 expression in LX-2 human hepatic stellate cells via PPARy up- regulation. Aramchol™ was surprisingly found to be effective in reversing established fibrosis, and in reducing the production of collagen specifically in stellate cells.
[0065] Thus, independently from its reported activities on liver metabolism in subjects with NAFLD, Aramchol™ is surprisingly found herein to be effective in the treatment of new patient populations and patient subpopulations, such as in the treatment of patients with NAFLD or NASH who have not yet developed fibrosis. In addition, Aramchol™ was found to be effective in the treatment of non-hepatic fibrosis and various conditions characterized by fibrosis of environmental and/or immune etiology, hepatic fibrosis in patients with NAFLD or NASH, treatment of hepatic fibrosis in patients with NAFLD or NASH and advanced fibrosis (i.e. stage 2 or stage 3 fibrosis), treatment of hepatic fibrosis in patient with NAFLD or NASH who have cirrhosis (i.e. stage 4 fibrosis), treatment of hepatic fibrosis caused by contact with drugs, toxins or surgery, and specifically in alleviating hepatic cirrhosis, treatment of fibrosis without worsening of NAFLD or NASH, treatment of fibrosis without progression to Cirrhosis. The invention advantageously provides for the treatment of these new patient populations with enhanced efficacy and/or safety and minimized side effects. [0066] In various embodiments, the fibrosis may be pulmonary fibrosis (e.g. idiopathic pulmonary fibrosis, diffuse interstitial pulmonary fibrosis, pleural fibrosis and fibrosis associated with asthma, fibrous dysplasia, cystic fibrosis), heart fibrosis (e.g. endomyocardial fibrosis and fibrosis associated with cardiovascular disease), kidney fibrosis (e.g. associated with renal failure), dermal fibrosis (e.g. keloid), ocular fibrosis, mucosal fibrosis, fibrosis of the central nervous system, fibrosis in bone or bone marrow, fibrosis in an endocrine organ (e.g. pancreas), fibrosis in the gastro-intestinal system, mediastinal fibrosis, postfibrinous fibrosis, proliferative fibrosis, retroperitoneal fibrosis, pancreatic fibrosis, fibrosis associated with an autoimmune disease (e.g. systemic lupus erythematosus (SLE), Sjogren syndrome, or diffuse systemic sclerosis with scleroderma).
[0067] For example, endomyocardial fibrosis an idiopathic type of myocardiopathy that is endemic in various parts of Africa and rarely in other areas, characterized by cardiomegaly, marked thickening of the endocardium with dense white fibrous tissue that may extend to involve the inner myocardium, and by congestive heart failure.
[0068] Idiopathic pulmonary fibrosis (e.g. diffuse idiopathic interstitial fibrosis, diffuse interstitial pulmonary fibrosis) is achronic inflammatory progressive fibrosis of the pulmonary alveolar walls, with steadily progressive dyspnea, resulting in death from oxygen lack or right heart failure. Most cases are of unknown origin, although some are thought to result from pneumoconiosis, hypersensitivity pneumonitis, scleroderma, and other diseases.
[0069] Mediastinal fibrosis is characterized by development of hard white fibrous tissue in the upper portion of the mediastinum, sometimes obstructing the air passages and large blood vessels; called also fibrosing or fibrous mediastinitis.
[0070] Pleural fibrosis is characterized by fibrosis of the visceral pleura so that part or all of a lung becomes covered with a plaque or a thick layer of nonexpansible fibrous tissue. The more extensive form is called fibrothorax.
[0071] Postfibrinous fibrosis occurs in tissues in which fibrin has been deposited.
[0072] Proliferative fibrosis refers to a condition in which the fibrous elements continue to proliferate after the original causative factor has ceased to operate.
[0073] Retroperitoneal fibrosis (Ormond disease, periureteral fibrosis) is characterized by deposition of fibrous tissue in the retroperitoneal space, producing vague abdominal discomfort, and often causing blockage of the ureters, with resultant hydronephrosis and impaired renal function, which may result in renal failure.
[0074] In various embodiments, this invention provides a method for inhibiting the development of hepatic fibrosis in a subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero, comprising administering to the subject greater than 300 mg per day of Aramchol™, or a pharmaceutically acceptable salt thereof, thereby inhibiting the development of hepatic fibrosis in said subject. In various embodiments, the human subject being treated is afflicted with Non- Alcoholic Steatohepatitis (NASH). In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In some embodiments, each Aramchol™ dose administered to the subject is 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850 or 900 mg once, twice or three times a day; each represent a separate embodiment according to this invention. In one embodiment, the Aramchol™ is administered to the subject, twice daily, in doses of 300 mg each. In one embodiment, the Aramchol™ is administered to the subject, three times daily, in doses of 300 mg each. In one embodiment, the Aramchol™ is administered to the subject, twice daily, in doses of 400 mg each. In one embodiment, the Aramchol™ is administered to the subject, three times daily, in doses of 400 mg each.
[0075] This invention also provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to the subject greater than 300 mg per day of Aramchol™, or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject. In an embodiment the human subject being treated is afflicted with Non-Alcoholic Fatty Liver Disease (NAFLD). In an embodiment the human subject being treated is afflicted with Non-Alcoholic Fatty Liver Disease (NAFLD) In an embodiment the human subject being treated is afflicted with Non-Alcoholic Steatohepatitis (NASH). In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In some embodiments, each Aramchol™ dose administered to the subject is 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850 or 900 mg once, twice or three times a day; each represent a separate embodiment according to this invention. In one embodiment, the Aramchol™ is administered to the subject, twice daily, in doses of 300 mg each. In one embodiment, the Aramchol™ is administered to the subject, three times daily, in doses of 300 mg each. In one embodiment, the Aramchol™ is administered to the subject, twice daily, in doses of 400 mg each. In one embodiment, the Aramchol™ is administered to the subject, three times daily, in doses of 400 mg each.
[0076] In various embodiments this invention provides a method for the treatment of fibrosis in a subject.
[0077] In various embodiments, the fibrosis is other than liver fibrosis.
[0078] This invention also provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to the subject greater than 300 mg per day of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject.
[0079] The exposure and absorption of a drug is limited by its bioavailability, which is usually determined by the solubility and permeability or the drug. It is herein demonstrated based on modeling and simulations, that a two-dose regimen (BID) of Aramchol™, i.e., administration of a given amount of Aramchol™ (e.g., 600 mg) in two doses (i.e., 300 mg each), twice daily, instead of one dose (i.e., 600 mg), once daily, is expected to increase the bioavailability of the drug and accordingly its exposure. It is therefore suggested that splitting the dose of Aramchol™ will be beneficial to the subject. Therefore, according to this invention, the daily amount of Aramchol™ is administered to a subject in a split dose regimen, including but not limited to: a two doses regimen (twice daily, BID), a three doses regimen (three times daily, TID), a four-dose regimen, or a five-dose regimen.
[0080] Accordingly, this invention provides a method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to the subject a daily dose of 100, 300, 400, 600, or 900 mg of 3 b -arach i dy I a m i do - 7 a. 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In some embodiments, each Aramchol™ dose administered to the subject is 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850 or 900 mg once, twice or three (3) times a day; each represent a separate embodiment according to this invention. In yet another embodiment, a dose of 300 mg of Aramchol™ is administered twice daily. In yet another embodiment, a dose of 400 mg of Aramchol™ is administered twice a day. In yet another embodiment, a dose of 300 mg of Aramchol™ is administered three times a day. [0081] In various embodiments, the human subject being treated is afflicted with Non-Alcoholic Fatty Liver Disease (NAFLD).
[0082] In various embodiments, the human subject being treated is afflicted with Non- Alcoholic Steatohepatitis (NASH).
[0083] In various embodiments, the human subject is afflicted with NAFLD but not afflicted with Non-Alcoholic Steatohepatitis (NASH). In various embodiments, the human subject has a NAFLD Activity (NAS) Score of at least 4. In various embodiments, the human subject has a NAFLD Activity (NAS) Score of at least 5, at least 6, or at least 7. In various embodiments, the human subject has a ballooning score of at least 1, an inflammation score of at least 1, and a steatosis score of at least 1. In various embodiments, the human subject has ballooning. In other embodiments, the human subject has high levels of ALT, AST, HbAlc or any combination thereof. In other embodiments, the human subject afflicted with fibrosis has progressed to Cirrhosis.
[0084] In various embodiments, the human subject is afflicted with (NASH). In various embodiments, the human subject has a NAFLD Activity (NAS) Score of at least 4. In various embodiments, the human subject has a NAFLD Activity (NAS) Score of at least 5, at least 6, or at least 7. In various embodiments, the human subject has a ballooning score of at least 1, an inflammation score of at least 1, and a steatosis score of at least 1. In various embodiments, the human subject has ballooning. In other embodiments, the human subject has high levels of ALT, AST, HbAlc or any combination thereof. In other embodiments, the human subject afflicted with fibrosis has progressed to Cirrhosis.
[0085] In various embodiments, the human subject is afflicted with NAFLD but not afflicted with Non-Alcoholic Steatohepatitis (NASH).
[0086] In various embodiments, the human subject is afflicted with Diabetes Mellitus type II or pre-diabetes. One of the following 3 criteria is needed for pre-Diabetes: Fasting Plasma Glucose > lOOmg/dl (5.5 minol/1) or 2hPG following 75g OGTT > 140 (7.8 mmol/1) mg/dl or HbAlc > 5.7%. HbAlc can be repeated at Investigator's discretion. [0087] In various embodiments, the subject’s hepatic fibrosis is stage 2, 3, or 4 fibrosis. In various embodiments, the subject’s hepatic fibrosis is stage 1 fibrosis. In various embodiments, the subject’s hepatic fibrosis is stage la, stage lb, or stage lc fibrosis.
[0088] In various embodiments, the human subject has a diet that is high fat and high calorie. As used herein, a high fat, high calorie diet contains at least 4000 calories per day, of which approximately 50% comes from fat. In various embodiments, the human subject is resistant to lifestyle intervention. In various embodiments, the human subject is resistant to diet intervention. In various embodiments, the human subject is naive to naive to Aramchol™ treatment. In various embodiments , the subject is naive to NAFLD treatment.
[0089] The invention further relates to the treatment and reduction of fibrosis, specifically liver fibrosis. The invention provides compositions and methods useful for the treatment of hepatic cirrhosis, portal hypertension, and hepato-fibrotic conditions caused by contact with hepatotoxic chemical substances or by mechanical obstruction. Compositions and methods according to the invention employ the use of 3P-arachidylamido-7a,12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™) or a pharmaceutically acceptable salt thereof.
[0090] In one aspect, there is provided a method for the treatment of hepatic fibrosis in a subject in need thereof, the hepatic fibrosis being caused by contact with a hepatotoxic chemical substance or by mechanical obstruction, comprising administering to the subject an effective amount of Aramchol™, or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject.
[0091] In another aspect there is provided a method for the treatment of hepatic fibrosis in a subject in need thereof, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof, with the proviso that the fibrosis is associated with a disorder other than non-alcoholic liver disease and non-alcoholic steatohepatitis.
[0092] In another aspect there is provided a method for treating or inhibiting a disorder selected from the group consisting of hepatic cirrhosis and portal hypertension in a subject in need thereof, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof.
[0093] In another aspect there is provided a method of treating or inhibiting a fibrotic disease or condition in a subject in need thereof, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof, thereby treating or inhibiting the disease or condition in the subject, wherein said disease or condition is selected from the group consisting of: alcoholic liver disease, viral hepatitis, parasitic hepatitis, drug-induced hepatitis, toxin-induced hepatitis, primary biliary cirrhosis and congenital hepatic fibrosis.
[0094] Known indications suggested for FABAC treatment include those disclosed in US Patents 6,384,024, 6,395,722, 6,589,946, 7,501,403, 8,110,564 and 8,975,246, as detailed herein, and are explicitly excluded in an embodiment. In some embodiments, the subject to be treated by the methods of the invention is not afflicted with an additional medical condition.
[0095] Further, according to an aspect of the present invention, there is provided a method for the treatment of hepatic fibrosis in a subject in need thereof, the hepatic fibrosis being caused by contact with a hepatotoxic chemical substance or by mechanical obstruction, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject.
[0096] According to embodiments of the present invention, the hepatic fibrosis is caused by a factor selected from the group consisting of chronic alcoholism, malnutrition, hemochromatosis, passive congestion, exposure to poisons or toxins, exposure to drugs, immune reactions, genetically determined sensitivities to a certain substance and infections.
[0097] In other embodiments, the hepatic fibrosis is caused by a factor selected from the group consisting of viral hepatitis, syphilis and a parasitic infection. In a particular embodiment, said parasitic infection is selected from the group consisting of Schistosomiasis mansoni and S. japonica. In another particular embodiment, the viral hepatitis is associated with is chronic hepatitis C infection.
[0098] According to another embodiment of the present invention, there is provided a method for the treatment of hepatic fibrosis in a subject, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof, with the proviso that the fibrosis is associated with a disorder other than non-alcoholic liver disease and non-alcoholic steatohepatitis.
[0099] In various embodiments, the subject is not diagnosed with fatty liver. In certain embodiments, the fibrosis is associated with a disorder selected from the group consisting of autoimmune hepatitis, storage or metabolism hepatic disorders, congenital hepatic fibrosis, infection, primary biliary cirrhosis and primary sclerosing cholangitis; each possibility represents a separate embodiment of the invention. In a particular embodiment, the fibrosis is associated with congenital hepatic fibrosis, a developmental disorder of the liver, marked by formation of irregular broad bands of fibrous tissue containing multiple cysts formed by disordered terminal bile ducts, resulting in vascular constriction and portal hypertension.
[0100] Non-limiting examples for diseases associated with storage or metabolism abnormalities that are characterized by hepatic fibrosis (storage or metabolism hepatic disorders) include a 1 -Antitrypsin deficiency, copper storage diseases (e.g., Wilson disease), fructosemia, galactosemia, glycogen storage diseases (especially types III, IV, VI, IX, and X), iron-overload syndromes (hemochromatosis), lipid abnormalities (e.g., Gaucher disease), peroxisomal disorders (e.g., Zellweger syndrome), and tyrosinemia; each possibility represents a separate embodiment of the invention.
[0101] Non-limiting examples for infections characterized by liver fibrosis include bacterial infections (e.g., brucellosis), parasitic infections (e.g., echinococcosis), and viral infections (e.g., viral hepatitis including, but not limited to chronic hepatitis B or C); each possibility represents a separate embodiment of the invention. In a particular embodiment, the infection is chronic hepatitis C infection.
[0102] According to yet further embodiments, the fibrosis is associated with contact with a hepatotoxic chemical substance, including, but not limited to alcohol, amiodarone, chlorpromazine, isoniazid, methotrexate, methyldopa, oxyphenisatin, and tolbutamide; each possibility represents a separate embodiment of the invention. In a particular embodiment, said substance is alcohol.
[0103] According to further embodiments, the fibrosis is associated with mechanical obstruction, e.g. scarring due to prior liver surgery.
[0104] In another embodiment the disorder is associated with COL1A1 and/or PPAR-g dysregulation in hepatic stellate calls. In a particular embodiment, said disorder is associated with COL1A1 up-regulation and PPAR-g down-regulation in hepatic stellate calls of said subject.
[0105] In another aspect, there is provided a method for treating or inhibiting a disorder selected from the group consisting of hepatic cirrhosis and portal hypertension in a subject in need thereof, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof.
[0106] In another embodiment, said fibrosis is manifested by portal hypertension and/or hepatic cirrhosis. In a particular embodiment, said fibrosis is manifested by hepatic cirrhosis. [0107] In one embodiment, the disorder is hepatic cirrhosis. In another embodiment, said disorder is portal hypertension. According to some embodiments, the methods of the invention advantageously provide for treating an existing condition of hepatic cirrhosis and/or portal hypertension. Thus, according to some embodiments, the method comprises determining whether said subject is afflicted with of hepatic cirrhosis and/or portal hypertension, and administering said Aramchol™ or a pharmaceutically acceptable salt thereof to a subject afflicted with of hepatic cirrhosis and/or portal hypertension. According to other embodiments, said of hepatic cirrhosis and/or portal hypertension is associated with a disorder as described herein; each possibility represents a separate embodiment of the invention.
[0108] In other embodiments, the method is used for inhibiting or preventing a symptom of hepatic cirrhosis and/or portal hypertension. According to various specific embodiments, the method is used for inhibiting or preventing a symptom of portal hypertension, including, but not limited to variceal bleeding, ascites, and portosystemic encephalopathy. In other particular embodiments, the method is used for inhibiting or preventing a symptom of hepatic cirrhosis, including, but not limited to hepatic insufficiency and fatal liver failure; each possibility represents a separate embodiment of the invention.
[0109] In various embodiments, the invention provides a method of treating or inhibiting a fibrotic disease or condition in a subject in need thereof, comprising administering to the subject an effective amount of Aramchol™ or a pharmaceutically acceptable salt thereof, thereby treating or inhibiting the disease or condition in the subject, wherein said disease or condition is selected from the group consisting of: alcoholic liver disease, viral hepatitis, parasitic hepatitis, drug-induced hepatitis, toxin- induced hepatitis, primary biliary cirrhosis and congenital hepatic fibrosis. In another embodiment, said disease or condition is selected from the group consisting of: alcoholic liver disease, parasitic hepatitis, drug-induced hepatitis, and toxin-induced hepatitis; each possibility represents a separate embodiment of the invention. In another embodiment, said disease or condition is chronic. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In some embodiments, each Aramchol™ dose administered to the subject is 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850 or 900 mg once, twice or three (3) times a day; each represent a separate embodiment according to this invention.
1.1. Administration and dosage form
[0110] According to some embodiments, the compound to be administered (e.g. Aramchol™) is in the form of a composition (referred to as the composition of the invention) comprising a therapeutically effective amount of at least one of said compound. As used herein, the term "effective amount" means an amount of compound that is capable of reducing and/or attenuating a disorder or symptom as described herein. The specific dose of a compound administered according to this invention will, of course, be determined by the particular circumstances surrounding the case including, for example, the compound administered, the route of administration, the physiological state of the subject, and the severity of the condition being treated.
[0111] This invention provides a medicament comprising greater than 300 mg of 3b- arachidylamido-7a, 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™) for use in administration to a human subject, including any of the human subjects recited hereinabove.
[0112] In various embodiments, the medicament comprises greater than 350 mg of Aramchol™. In various embodiments, the medicament comprises between 350 mg and 1200 mg of Aramchol™. In various embodiments, the medicament comprises 400 mg, 500 mg, 600 mg, 700 mg, 800mg, 900mg, 1000 mg, 1100 mg, or 1200 mg of Aramchol™. In various embodiments, the medicament comprises between 400 mg and 1100 mg, or between 500 mg and 1000 mg, or between 600 mg and 900 mg of Aramchol™. In various embodiments, the medicament comprises 400 mg or 600 mg of Aramchol™.
[0113] In various embodiments, the medicament is to be administered daily. In some embodiments, the medicament daily dose is between about 400 mg and 900 mg. In some embodiments, the medicament is administered in a dose regimen of at least twice a day. In various embodiments, the medicament is to be administered once (QD), twice (BID) or three times a day (TID), in a dose of 100, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800 or 900 mg each; each represent a separate embodiment according to this invention. In various embodiments, the medicament is to be administered once a day (QD). In other embodiments, the medicament is to be administered twice a day (BID). In various embodiments, the medicament is to be administered three times a day (TID).
[0114] This invention also provides 3 b - a r ac h i dy I a m i do - 7 a. 12a-dihydroxy^-cholan-24-oic acid (Aramchol™) for use in administration to a human subject at a daily dose of greater than 300 mg. In some embodiments, the medicament daily dose is between about 400 mg and 900 mg. In some embodiments, the medicament is administered in a dose regimen of at least twice a day. In various embodiments, the medicament is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the medicament is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the medicament is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the medicament is administered twice a day (BID) in dose amount of 400 mg each dose. In various embodiments, the human subject is any of the human subjects recited hereinabove.
[0115] In various embodiments, the daily dose of Aramchol™ is greater than 350 mg. In various embodiments, the daily dose of Aramchol™ is between 350 mg and 1200 mg. In various embodiments, the daily dose of Aramchol™ is 400 mg, 500 mg, 600 mg, 700 mg, 800mg, 900mg, 1000 mg, 1100 mg, or 1200 mg; each represents a separate embodiment according to this invention. In various embodiments, the daily dose of Aramchol™ is between 400 mg and 1100 mg, or between 500 mg and 1000 mg, or between 600 mg and 900 mg. In various embodiments, the daily dose of Aramchol™ is 400 mg or 600 mg per day. In various embodiments, the daily dose of Aramchol™ is 600 mg, administered in two doses of 300 mg each. In various embodiments, the daily dose of Aramchol™ is 600 mg, administered in three doses of 200 mg each. In various embodiments, the daily dose of Aramchol™ is 900 mg, administered in three doses of 300 mg each. In various embodiments, the daily dose of Aramchol™ is 800 mg, administered in two doses of 400 mg each. In various embodiments, the daily dose of Aramchol™ is 600 mg, administered in two doses of 300 mg each, one in the morning and one in the evening.
[0116] Any suitable route may be used to administer the medicament or Aramchol™ of the invention to a subject.
[0117] According to some embodiments, suitable administration routes may be systemic routes.
According to some embodiments, administering is administering systemically. According to some embodiments, the composition is formulated for systemic administration.
[0118] According to another embodiment, administration systemically is through an enteral route.
According to another embodiment, administration through an enteral route is oral administration. According to some embodiments, the composition is formulated for oral administration.
[0119] Thus, the invention provides a method for treating the subjects recited in this application by administering Aramchol™ to the subject, wherein at least 350 mg of Aramchol™ is administered to the subject per day. In various embodiments, between 350 mg and 1200 mg of Aramchol™ is administered to the subject per day. In various embodiments, 400 mg, 500 mg, 600 mg, 700 mg, 800mg, 900mg, 1000 mg, 1100 mg, or 1200 mg of Aramchol™ is administered to the subject per day. In various embodiments, between 400 mg and 1100 mg, or between 500 mg and 1000 mg, or between 600 mg and 900 mg of Aramchol™ is administered to the subject per day. In various embodiments, 400 mg or 600 mg of Aramchol™ is administered to the subject per day. In various embodiments, the method comprises administering Aramchol™ in an amount of about 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 mg once, twice or three times per day; each represents a separated embodiment according to this invention. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the method comprises administering Aramchol™ in an amount of about 150 to about 600 mg twice a day. In various embodiments, the method comprises administering Aramchol™ in an amount of about 200 to about 450 mg twice a day. In various embodiments, the method comprises administering Aramchol™ in an amount of 300 mg twice a day. In various embodiments, 600 mg per day is administered to the subject in two doses of 300 mg each. In various embodiments, 600 mg per day is administered to the subject in three doses of 200 mg each. In various embodiments, 600 mg per day is administered to the subject in two doses of 300 mg each, one dose every 12 hrs.
[0120] In various embodiments, the medicament or Aramchol™ is administered in the morning, in the afternoon, or in the evening. In various embodiments, the medicament or Aramchol™ is administered in the morning and in the evening. In various embodiments, the medicament or Aramchol™ is administered every 12 hours. In various embodiments, the medicament or Aramchol™ is administered every 8 hours.
[0121] In various embodiments, the medicament or Aramchol™ is administered at the same time as, or within 30 minutes of a meal
[0122] In various embodiments, the meal is breakfast, lunch, or dinner. In various embodiments, the meal is breakfast and dinner. In various embodiments, the meal is breakfast lunch and dinner.
[0123] In various embodiments, the meal is a high fat meal. A high fat meal is a meal wherein approximately 500 to 600 calories are fat calories.
[0124] In various embodiments, the meal is a high calorie meal. A high calorie meal is a meal of approximately 800 to 1000 calories.
[0125] In various embodiments, the medicament or Aramchol™ is administered with water. In various embodiments, the medicament or Aramchol™ is administered with at least 100 or at least 200 mL of water. [0126] In various embodiments, the Aramchol™ is administered over the course of at least 52 weeks, at least 72 weeks, at least 96 weeks, at least 2 years, at least 3 years, or at least 4 years.
[0127] The invention further relates to use of a therapeutically effective amount of 3b- arachidylamido-7a, 12oc-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy^-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby inhibiting the development of hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the Aramchol™ is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the Aramchol™ is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 400 mg each dose.
[0128] The invention further provides 3b-hGho1i^1hih o-7a, 12oc-dihydroxy^-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3b- arachidylamido-7a, 12a^ί1^k^-5b-ϋ1io1hh-24-oίϋ acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3b-hGhϋ1i^1hihMo-7a, 12a-dihydroxy- 5b-o1io1hh-24-oΐϋ acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby inhibiting the development of hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the Aramchol™ is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the Aramchol™ is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 400 mg each dose.
[0129] The invention further provides a medicament comprising 3 b -arac h i dy I a m i do- 7o r. 12a- dihydroxy-5 — cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3 -arachidylamido-7ot, 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby for inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3 b -arac h i dy I a m i do - 7 a. 12a- di hydroxy-5 b-cho Ian -24-oic acid (Aramchol™) , or a pharmaceutically acceptable Salt thereof, thereby inhibiting the development of hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the Aramchol™ is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the Aramchol™ is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 400 mg each dose.
[0130] The invention provides a pharmaceutical package comprising a) 3 -arachidylamido-7a, 12c- -di hydroxy-5 b-cho Ian -24-oic acid (Aramchol™) , or a pharmaceutically acceptable salt thereof; and b) instructions for use in inhibiting the development of hepatic fibrosis in a human subject afflicted with Non-Alcoholic Fatty Liver Disease and having a fibrosis score of zero comprising administering to the subject greater than 300 mg per day of 3b^hLk^^ih o-7a, 12a-dihydroxy^-cholan-24-oic acid (Aramchol™) , or a pharmaceutically acceptable salt thereof, thereby inhibiting the development of hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the Aramchol™ is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the Aramchol™ is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 400 mg each dose.
[0131] According to some embodiments, oral administration is in the form of hard or soft gelatin capsules, pills, capsules, tablets, including coated tablets, dragees, elixirs, suspensions, liquids, gels, slurries or syrups and controlled release forms thereofThus, the invention provides a method of administering Aramchol™ in the form of a tablet, a capsule, or in a liquid.
[0132] Suitable carriers for oral administration are well known in the art. Compositions for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries as desired, to obtain tablets or dragee cores. Non limiting examples of suitable excipients include fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol, cellulose preparations such as, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, and sodium carbomethylcellulose, and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
[0133] If desired, disintegrating agents, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate, may be added. Capsules and cartridges of, for example, gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base, such as lactose or starch.
[0134] Solid dosage forms for oral administration include without limitation capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert pharmaceutically acceptable carrier such as sucrose, lactose, or starch. Such dosage forms can also comprise, as it normal practice, additional substances other than inert diluents, e.g., lubricating, agents. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings. The term“enteric coating”, as used herein, refers to a coating which controls the location of composition absorption within the digestive system. Non-limiting examples for materials used for enteric coating are fatty acids, waxes, plant fibers or plastics. [0135] Liquid dosage forms for oral administration may further contain adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring and perfuming agents.
[0136] According to some embodiments, concomitant treatment with fatty acids such as ethyl eicosapentanoate, eicosapentaenoic acid, and their amides, salts and phospholipids is explicitly excluded. In other embodiment, concomitant treatment with bile acids such as ursodeoxycholic acid and lithocholic acid is excluded. In other embodiments concomitant treatment with vitamin D receptor agonists, acetyl-CoA carboxylase inhibitors, dual PPAR delta/ gamma agonists, and inhibitors of myofibroblast trans-differentiation and activation is excluded. According to advantageous embodiments, Aramchol™ or the pharmaceutically acceptable salt thereof is used as a sole active ingredient.
[0137] According to some embodiments, the composition is administered in several dosages over prolonged periods until a sufficient response has been achieved.
[0138] As disclosed herein, Aramchol™ was found to be an unexpectedly potent therapeutic agent, capable of reversing established fibrosis and for inhibiting the development of fibrosis, and reducing cirrhosis and collagen synthesis in stellate calls even when used as a single therapeutic agent, in the absence of adjunct therapy. Thus, according to an advantageous embodiment of the methods of the invention, Aramchol™ or the pharmaceutically acceptable salt thereof is administered as a sole active ingredient. In another embodiment, the subject is human.
[0139] In another embodiment of the methods of the invention, Aramchol™ is administered orally.
In another embodiment of the methods of the invention, Aramchol™ is in the form of Aramchol™ free acid. In another embodiment of the methods of the invention, Aramchol™ is in the form of an amine -based salt. In certain particular embodiments, the salt is a meglumine, lysine or tromethamine Aramchol™ salt; each possibility represents a separate embodiment of the invention.
1.2. Patient outcomes
[0140] According to various embodiments, this invention provides a method of for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to the subject greater than 300 mg per day of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, thereby treating hepatic fibrosis in said subject. In some embodiments, the Aramchol™ daily dose is between about 400 mg and 900 mg. In some embodiments, the Aramchol™ is administered in a dose regimen of at least twice a day. In various embodiments, the Aramchol™ is administered once (QD), twice (BID) or three times (TID) daily in doses of 100, 150, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 mg each; each represents a separate embodiment according to this invention. In various embodiments, the daily dose is 600 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 900 mg and the Aramchol™ is administered three times a day (TID) in dose amount of 300 mg each dose. In various embodiments, the daily dose is 800 mg and the Aramchol™ is administered twice a day (BID) in dose amount of 400 mg each dose.
[0141] In various embodiments, the methods according to this invention further comprise reduction in liver fat in said subject.
[0142] In various embodiments, the methods according to this invention further comprise improvement in subject’s Steatosis.
[0143] In various embodiments, the methods according to this invention further comprise reduction in subject’s ballooning.
[0144] In various embodiments, the methods according to this invention further comprise NAFLD resolution in said subject. In other embodiments, the methods according to this invention further comprise NASH resolution in said subject.
[0145] In various embodiments, the methods according to this invention further comprise NAFLD resolution without worsening of fibrosis. In various embodiments, the methods according to this invention further comprise reduction of fibrosis without NAFLD worsening. In various embodiments, the methods according to this invention further comprise NASH resolution without worsening of fibrosis. In various embodiments, the methods according to this invention further comprise reduction of fibrosis without NASH worsening.
[0146] In various embodiments, the methods according to this invention further comprise reduction of ALT levels in said subject.
[0147] In various embodiments, the methods according to this invention further comprise reduction of AST levels in said subject.
[0148] In various embodiments, the methods according to this invention further comprise reduction of HbAlc levels in said subject. [0149] In various embodiments, the methods according to this invention further comprise lack of subject’s progression to Cirrhosis.
[0150] In various embodiments, the methods according to this invention further comprise inhibiting progression of Non-Alcoholic Fatty Liver Disease (NAFLD) and/or Non-Alcoholic Steatohepatitis (NASH).
[0151] In various embodiments, the methods according to this invention further comprise any combination of hereinbove mentioned indications and/or symptoms.
[0152] In various embodiments, treating the subject comprises lack of worsening of the subject’s NAFLD Activity (NAS) score.
[0153] In an embodiment treating the subject comprises lack of worsening of the subject’s Steatosis, Activity and Fibrosis (SAF) Activity score.
[0154] In various embodiments, treating the subject comprises lack of worsening of the subject’s fibrosis score.
[0155] In various embodiments, the lack of worsening is lack of worsening at 52, 65, 72 or 96 weeks from the commencement of administration of Aramchol™.
[0156] In various embodiments, the lack of worsening is lack of worsening at 2, 3, or 4 years from the commencement of administration of Aramchol™.
[0157] In various embodiments, treating the subject treating comprises an improvement of the subject’s NAFLD Activity (NAS) score.
[0158] In various embodiments, the subject’s NAS score is at least 4 at the commencement of administration of Aramchol™ and the improvement of the subject’s NAS score is an improvement of at least 2 points.
[0159] In various embodiments, treating the subject comprises an improvement of the subject’s Steatosis, Activity and Fibrosis (SAF) Activity Score.
[0160] In various embodiments, the subject’s SAF Activity score is at least 4 at the commencement of administration of Aramchol™ and improvement of the subject’s SAF Activity score is an improvement of at least 2 points. [0161] In various embodiments, treating the subject comprises an improvement of the subject’s fibrosis score.
[0162] In various embodiments, the improvement of the subject’s fibrosis score is an improvement of 1 grade, or greater than 1 grade. In various embodiments, improvement is improvement at 52, 65, 72, or 96 weeks from the commencement of administration of Aramchol™.
[0164] In various embodiments, improvement is improvement at 2, 3, or 4 years from the commencement of administration of Aramchol™.
[0165] In various embodiments, treating the fibrosis comprises inhibiting progression of Non- Alcoholic Fatty Liver Disease (NAFLD). In various embodiments, inhibiting the fibrosis comprises inhibiting progression of Non-Alcoholic Fatty Liver Disease (NAFLD).
[0166] In various embodiments, inhibiting progression of NAFLD comprises prevention of progression, or reduced progression relative to a patient not treated with Aramchol™.
[0167] In various embodiments, the human subject is afflicted with Non-Alcoholic Steatohepatitis (NASH) and treating comprises inhibiting progression of NASH.
[0168] In various embodiments, inhibiting progression of NASH comprises prevention of progression, or reduced progression relative to a patient not treated with Aramchol™.
[0169] In various embodiments, treating comprises preventing progression from Non-Alcoholic Fatty Liver Disease (NAFLD) to NASH.
[0170] In various embodiments, improvement progression is progression at 2, 4, 8, 24, 40, 52, 65, 72, or 96 weeks from the commencement of administration of Aramchol™.
[0171] In various embodiments, improvement progression is progression at 2, 3, or 4 years from the commencement of administration of Aramchol™.
[0172] In various embodiments, the human subject is afflicted with Non-Alcoholic Steatohepatitis (NASH) and the treating comprises NASH resolution in the subject.
[0173] In various embodiments, NASH resolution comprises the human subject having a ballooning score of 0 and an inflammation score of 0 or 1. [0174] In various embodiments, treating comprises NASH resolution in the subject at 52, 72, or 96 weeks from the commencement of administration of Aramchol™.
[0175] In various embodiments, treating comprises NASH resolution in the subject at 2, 3, or 4 years from the commencement of administration of Aramchol™.
[0176] In various embodiments , treating comprises a reduction in the level of liver triglycerides in the subject relative to the level at the commencement of administration of Aramchol™.
[0177] In various embodiments, treating comprises a reduction in the ratio of liver triglycerides to water in the subject relative to the ratio at the commencement of administration of Aramchol™.
[0178] In various embodiments, there is a greater than 10% reduction in ratio of liver triglycerides to water.
[0179] In various embodiments, there is a 10% to 40% reduction in ratio of liver triglycerides to water.
[0180] In various embodiments, there is a 15% to 35% reduction in ratio of liver triglycerides to water.
[0181] In various embodiments, there is a 20% to 30% reduction in ratio of liver triglycerides to water.
[0182] In various embodiments, the methods according to this invention comprise: a. a reduction in the level of Hemoglobin A1C or HOMA-IR; b. a reduction in the level of Fibrinogen, CK-18, C-reactive protein (CRP), TNFa, IL 6 and fibrosis Tests (NFS; c. a reduction in the ratio of leptin to adinopectin; or d. an increase in the level of adinopectin; in the subject relative to the level or ratio at the commencement of administration of Aramchol™.
[0183] In various embodiments, the methods according to this invention further comprise: a. a reduction in the human subject’s body weight relative to the human subject’ s body weight at the commencement of administration of Aramchol™; b. a reduction in the human subject’s waist circumference relative to the human subject’s waist circumference at the commencement of administration of Aramchol™; or c. a reduction in the human subject’s Fatty Liver Index relative to the human subject’s Fatty Liver Index at the commencement of administration of Aramchol™.
[0184] In various embodiments, the reduction or increase is a reduction or increase at 2, 4, 8, 24, 40, 52, 65, 72, or 96 weeks from the commencement of administration of Aramchol™.
[0185] In various embodiments, the reduction or increase is a reduction or increase at 2, 3, or 4 years from the commencement of administration of Aramchol™.
[0186] In another embodiment, administration of Aramchol™ or a pharmaceutically acceptable salt thereof according to the methods of the invention inhibits collagen synthesis (e.g. COL1A1 expression) in hepatic stellate calls. In another embodiment, administration of Aramchol™ or a pharmaceutically acceptable salt thereof according to the methods of the invention enhances PPAR-g expression in hepatic stellate calls.
[0187] The present invention also provides a medicament or Aramchol™ of the invention wherein the medicament or Aramchol™ is effective to prevent worsening of NASH in the subject.
[0188] In various embodiments, the medicament or Aramchol™ is effective to prevent worsening of fibrosis in the subject.
[0189] The present invention also provides a medicament or Aramchol™ of the invention wherein the human subject being administered the medicament or Aramchol™ is afflicted with fibrosis and wherein the medicament or Aramchol™ is effective to improve the human subject’s fibrosis score.
[0190] In various embodiments, the medicament or Aramchol™ is effective to improve the human subject’s NAFLD Activity (NAS) score.
[0191] In various embodiments, the medicament or Aramchol™ is effective to improve the human subject’s NAFLD Activity (NAS) score by at least 2 points. [0192] In various embodiments, the medicament or Aramchol™ is effective to improve the human subject’s Steatosis, Activity and Fibrosis (SAF) Activity score.
[0193] In various embodiments, the medicament or Aramchol™ is effective to improve the human subject’s Steatosis, Activity and Fibrosis (SAF) Activity score by at least 2 points.
[0194] In various embodiments, the medicament or Aramchol™ is effective to resolve NASH in the human subject.
[0195] In various embodiments, resolving NASH comprises reducing ballooning to a score of 0 and reducing inflammation to a score of 0 or 1.
[0196] In various embodiments, the medicament or Aramchol™ is effective at 2, 4, 8, 24, 40, 52, 65, 72, or 96 weeks, or 2, 3, or 4 years from the commencement of administration.
[0197] In various embodiments, the medicament or Aramchol™ is effective to reduce the level of liver triglycerides in the subject relative to the level at the commencement of administration.
[0198] In various embodiments, the medicament or Aramchol™ is effective to reduce the ratio of liver triglycerides to water in the subject relative to the ratio at the commencement of administration.
[0199] In various embodiments, the medicament or Aramchol™ is effective to reduce the ratio of liver triglycerides to water in the subject by at least 10% relative to the ratio at the commencement of administration.
[0200] In various embodiments, the medicament or Aramchol™ is effective to reduce the ratio of liver triglycerides to water in the subject by between 10% and 40% relative to the ratio at the commencement of administration.
[0201] In various embodiments, the medicament or Aramchol™ is effective to reduce the ratio of liver triglycerides to water in the subject by between 15% and 35% relative to the ratio at the commencement of administration.
[0202] In various embodiments, the medicament or Aramchol™ is effective to reduce the ratio of liver triglycerides to water in the subject by between 20% and 30% relative to the ratio at the commencement of administration.
[0203] In various embodiments, the medicament or Aramchol™ is effective to: a. reduce the level of Hemoglobin A1C or HOMA-IR; b. reduce the level of Fibrinogen, CK-18, C-reactive protein (CRP), TNFa, IL 6 and fibrosis Tests (NFS); c. reduce the ratio of leptin to adinopectin; or d. increase the level of adinopectin; in the subject relative to the level or ratio at the commencement of administration.
[0204] In various embodiments, the medicament or Aramchol™ is effective to: a. reduce the human subject’s body weight relative to the human subject’s body weight at the commencement of administration; b. reduce the human subject’s waist circumference relative to the human subject’s waist circumference at the commencement of administration; or c. reduce the human subject’s Fatty Liver Index relative to the human subject’s Fatty Liver Index at the commencement of administration.
[0205] In various embodiments, the medicament or Aramchol™ is effective at 2, 4, 8, 24, 40, 52, 65, 72, or 96 weeks, or 2, 3, or 4 years from the commencement of administration.
[0206] The invention relates to use of a therapeutically effective amount of 3 -arachidylamido-7a, 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for the treatment and inhibition of fibrotic disorders, hepato-fibrotic conditions associated with Non- Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH).
[0207] The invention provides 3 b - a r ac h i dy I a m i do - 7 a. 12a-dihydroxy-5 -cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for the treatment and inhibition of fibrotic disorders, hepato-fibrotic conditions associated with Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH).
[0208] The invention provides a medicament comprising 3 b - a r ac h i dy I a m i do - 7 a . 12a-dihydroxy^- cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, for the treatment and inhibition of fibrotic disorders, hepato-fibrotic conditions associated with Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH).
[0209] The invention provides a pharmaceutical package comprising a) 3 b - ar ac h i dy I a m i do- 7a. 12a- di hydroxy-5 b-cho Ian -24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof; and b) instructions for use of the Aramchol™ in the treatment and inhibition of fibrotic disorders, hepato- fibrotic conditions associated with Non-Alcoholic Fatty Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH).
Combination Therapy
[0210] The C-C motif chemokine receptor CCR5 is involved in the process by which HIV, the virus that causes AIDS, enters cells. CCR5 receptor antagonists are a class of small molecules that antagonize the CCR5 receptor. Hence, antagonists of this receptor are entry inhibitors and have potential therapeutic applications in the treatment of HIV infections. The C-C chemokine receptor type 2 (CCR2) is a protein that in humans is encoded by the CCR2 gene. This gene encodes two isoforms of a receptor for monocyte chemoattractant protein-1 (CCL2), a chemokine which specifically mediates chemotaxis of cells such as monocytes and macrophages. Cenicriviroc ((S,E)-8- (4-(2-Butoxyethoxy)phenyl)- 1 -isobutyl -N -(4-((( 1 -propyl- lH-imidazol -5 -yl)methyl)sulfinyl)phenyl)- l,2,3,4-tetrahydrobenzo[b]azocine-5-carboxamide, CAS No. 497223-25-3) is an inhibitor of both CCR2 and CCR5 receptors.
[0211] In various embodiments, of the invention, the methods further comprise administering a therapeutically effective amount of a pharmaceutical composition comprising a C-C chemokine receptor type 2 (CCR2) antagonist, a C-C chemokine receptor type 5 (CCR5) antagonist, a dual CCR2/CCR5 antagonist, or a combination or pharmaceutically acceptable salt thereof.
[0212] In various embodiments,, the CCR2 antagonist is selected from the group consisting of: a double-stranded RNA, a compound antagonizing the binding of CCR2 to its ligand, a neutralizing antibody to CCR2, a ligand corresponding to a neutralizing antibody to CCR2, an isolated peptide derived from the sequences of CCR2 or analogs thereof capable of inhibiting CCR2, an antisense nucleic acid, an antagonist microRNA, and an enzymatic RNA molecule.
[0213] In various embodiments, the methods further comprise administering to the subject a CCR5 antagonist and a CCR2 antagonist, or a dual CCR2/CCR5 antagonist. [0214] In various embodiments, the CCR2/CCR5 dual antagonist is (S,E)-8-(4-(2- Butoxyethoxy)phenyl)- 1 -isobutyl-N -(4-((( 1 -propyl- lH-imidazol -5 -yl)methyl)sulfinyl)phenyl)- l,2,3,4-tetrahydrobenzo[b]azocine-5-carboxamide (Cenicriviroc) or Cenicriviroc mesylate.
[0215] In various embodiments, the methods comprise administering a daily dose of 50 to 500 mg of Cenicriviroc. In certain embodiments, the method described above comprises administering a daily dose of 10 to 200 mg of Cenicriviroc. In certain embodiments, the method comprises administering a dailydose of 50 to 500 mg, 50 to 400 mg, 50 to 300 mg, 50 to 200 mg, or 100 to 200 mg of Cenicriviroc. In certain embodiments, the method described above comprises administering a daily dose of 100-200 mg of Cenicriviroc. In various embodiments, the method comprises administering a daily dose of 100 or 200 mg of Cenicriviroc.
[0216] In some embodiments, the methods further comprise administering a therapeutically effect amount of a pharmaceutical composition comprising at least one compound selected from the group consisting of ethyl eicosapentanoate (EPA-E), eicosapentaenoic acid (EPA) and its pharmaceutically acceptable amides, salts, esters and phospholipids.
[0217] In some embodiments, the EPA-E or EPA may be at least 40% by weight in total of the fatty acids and their derivatives.
[0218] In some embodiments, the pharmaceutical composition comprises Epadel® (Machida Pharmaceutical Co., Ltd., Tokyo Japan), Lovaza™ (GlaxoSmithKline, FL USA), Omacor™ (Pronova Biopharma ASA, Oslo Norway), Lotriga™(Takeda Pharmaceutical Co., Ltd., Osaka Japan), Vascepa™ (Amarin Pharma Inc., NJ USA), Epanova™ (Astra Zeneca Pharmaceuticals LP, Wilmington, Germany) or Omtryg™ (Trygg Pharma Inc., VA USA).
[0219] Lovaza™, omega-3-acid ethyl esters, predominantly a combination of ethyl esters of eicosapentaenoic acid (EPA - approximately 465 mg) and docosahexaenoic acid (DHA - approximately 375 mg), is indicated as an adjunct to diet to reduce triglyceride (TG) levels in adult patients with severe (>500 mg/dL) hypertriglyceridemia (Lovaza, Food and Drug Administration Approved Labeling (Reference ID:3371921) [online], GlaxoSmithKline, 2013).
[0220] The recommended dose and schedule for Lovaza™ is 4g per day. The daily dose may be taken as a single 27 4-gram dose (4 capsules) or as two 2-gram doses (2 capsules given twice daily).
[0221] In some embodiments, the administration of Lovaza™ comprises 4.0g per day, 3.5g per day, 3.0g per day, 2.5g per day, 2.0g per day, 1.5g per day, l.Og per day or less of Lovaza™. [0222] Omacor™, omega-3-acid ethyl esters, predominantly a combination of ethyl esters of eicosapentaenoic acid (EPA - approximately 465 mg) and docosahexaenoic acid (DHA - approximately 375 mg), is a a lipid-regulating agent. (Reference ID:”FPL for approved NDA 21-654”) [online], Abbott Laboratories).
[0223] The recommended dose and schedule for Omacor™ is 4 g per day. The daily dose may be taken as a single 4-g dose (4 capsules) or as two 2-g doses (2 capsules given twice daily).
[0224] In some embodiments, the administration of Omacor™ comprises 4.0g per day, 3.5g per day, 3.0g per day, 2.5g per day, 2.0g per day, 1.5g per day, l.Og per day or less of Omacor™.
[0225] Vascepa™, containing 1 gram of icosapent ethyl, an ethyl ester of the omega-3 fatty acid eicosapentaenoic acid (EPA), is a a lipid-regulating agent indicated as an adjunct to diet to reduce triglyceride (TG) levels in adult patients with severe (> 500 mg/dL) hypertriglyceridemia (Vascepa™, Food and Drug Administration Approved Labeling (Reference ID:3783357) [online], Amarin Pharmaceuticals, 2012).
[0226] The recommended dose and schedule for Vascepa™ is 4 grams per day taken as 2 capsules twice daily with food.
[0227] In some embodiments, the administration of Vascepa™ comprises 4.0g per day, 3.5g per day, 3.0g per day, 2.5g per day, 2.0g per day, 1.5g per day, l.Og per day or less of Vascepa™.
[0228] Epanova™, containing 1 gram of fish oil-derived free fatty acids, designated“omega-3 - carboxylic acids,” with at least 850 mg of polyunsaturated fatty acids, including multiple omega-3 fatty acids (eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA] being the most abundant), is a lipid-regulating agent indicated as an adjunct to diet to reduce triglyceride (TG) levels in adult patients with severe (>500 mg/dL) hypertriglyceridemia (Epanova™, Food and Drug Administration Approved Labeling (Reference ID:3501113) [online], AstraZeneca Pharmaceuticals, 2014).
[0229] The recommended dose and schedule for Epanova™ is 4 grams per day taken as 2 grams (2 capsules) twice daily, or 4 grams (4 capsules) once daily.
[0230] In some embodiments, the administration of Epanova™ comprises 4.0g per day, 3.5g per day, 3.0g per day, 2.5g per day, 2.0g per day, 1.5g per day, l.Og per day or less of Epanova™. [0231] Omtryg™, a combination of ethyl esters of omega-3 fatty acids predominantly a combination of ethyl esters of eicosapentaenoic acid (EPA - approximately 465 mg) and docosahexaenoic acid (DHA - approximately 375 mg), is a lipid-regulating agent, indicated as an adjunct to diet to reduce triglyceride (TG) levels in adult patients with severe (>500 mg/dL) hypertriglyceridemia (Omtryg™, Food and Drug Administration Approved Labeling (Reference ID:3494935) [online], Trygg Pharma Inc., 2014).
[0232] The recommended dose and schedule for Omtryg™ is 4 grams per day taken as 2 grams (2 capsules) twice daily, or 4 grams (4 capsules) once daily.
[0233] In some embodiments, the administration of Omtryg™ comprises 4.0g per day, 3.5g per day, 3.0g per day, 2.5g per day, 2.0g per day, 1.5g per day, l.Og per day or less of Omtryg™.
[0234] The compositions recited hereinabove are described in U.S. Patent Application Publication No. 2016/0213639, the entire contents of which is incorporated by reference.
[0235] In some embodiments, the method further comprises administering a therapeutically effect amount of an inhibitor of Acetyl-CoA carboxylase (ACC) alone, or in combination with one or more additional therapeutic agents.
[0236] As used herein generally, "ACC inhibitor" means any therapeutic agent that reduces the activity of an acetyl CoA carboxylase enzyme.
[0237] Suitable ACC inhibitors include those described in W02013/071169A1, W02014/182943A1, W02014/182945 Al, W02014/182950A1, and W02014/182951A1, the entirety of each of which is hereby incorporated by reference.
[0238] In some embodiments, the ACC inhibitor is soraphen A.
[0239] In some embodiments, additional therapeutic agents are independently selected from the group consisting of angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacylglycerol 0-acyltransferase 1 (DGAT1) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, farnesoid X receptor (FXR) agonists, FXR/TGR5 dual agonists, galectin-3 inhibitors, glucagon-like peptide (GLPI) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, I I — hydroxysteroid dehydrogenase (I I— HSD I) inhibitors, IL-I- antagonists, IL-6 antagonists, IL-I 0 agonists, IL-I 7 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5- lipoxygenase inhibitors, LPL gene stimulators, lysyl oxidase homolog 2 (LOXL2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PLC) inhibitors, PPARa agonists, PPARy agonists, PPAR8 agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturasel inhibitors, thyroid hormone receptor- agonists, tumor necrosis factor a (TNFa) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, PTPib inhibitors, and ASKI inhibitors.
[0240] The compositions recited hereinabove are described in PCT International Application Publication No. WO 2016/112305, the entire contents of which is incorporated by reference.
[0241] In some embodiments, the method further comprises administering a therapeutically effect amount of Pioglitazone hydrochloride (Actos®) or an enantiopure deuterium-enriched pioglitazone.
[0242] As used herein, the deuterated pioglitazone contains deuterium enrichment at the chiral center of pioglitazone and optionally in other locations in the compound. Further, the deuterium- enriched pioglitazone is provided in enantiomerically pure form.
[0243] In some embodiments, the deuterium-enriched compound having an optical purity of at least 75% enantiomeric excess.
[0244] Pioglitazone hydrochloride, the active ingredient of Actos®, is a thiazolidinedione and an agonist for peroxisome proliferatoractivated receptor (PPAR) gamma indicated as an adjunct to diet and exercise to improve glycemic control in adults with type 2 diabetes mellitus in multiple clinical settings (Actos®, Food and Drug Administration Approved Fabeling (Reference ID:2983732) [online], Takeda Pharmaceuticals, 2009-2011).
[0245] The recommended dose and schedule for Actos® is 15 mg or 30 mg once daily starting dose.
If there is inadequate glycemic control, the dose can be increased in 15 mg increments up to a maximum of 45 mg once daily.
[0246] In some embodiments, the administration of Actos® comprises 15mg, 16mg, 17mg, 18mg, 19mg, 20mg, 25mg, 30mg, 35mg, 40mg, or 45mg or less of Actos®.
[0247] The compositions recited hereinabove are described in PCT International Application Publication No. WO 2016/153948, the entire contents of which is incorporated by reference. [0248] In some embodiments, the method further comprises administering a therapeutically effect amount of a peroxisome proliferator activated receptor (PPAR) delta and gamma dual agonist.
[0249] In some embodiments, the delta activity is greater than gamma activity, and gamma activity is greater than alpha activity.
[0250] In some embodiments, the method further comprises administering a therapeutically effect amount of an indane acetic acids and their derivatives, which are dual PPAR delta and gamma agonists.
[0251] Exemplary additional therapeutic agents may include, but are not limited to combination with: farnesoid X receptor agonists such as obeticholic acid and Px-104, GR-MD-02, cysteamine bitartrate, simtuzumab, emricasan, GFT-505, CER-002, KD3010, KD3020, MBX8025, LUM002, RP- 103, galectin-3 blockers such as LIPC-1010 and GR-MD-02, cenicriviroc, vascular adhesion protein- 1 inhibitors such as PXS4728A, metformin, PPAR gamma agonists such as rosiglitazone and pioglitazone, metformin, pentoxyfylline, vitamin E, selenium, omega-3 fatty acids and betaine.
[0252] The compositions recited hereinabove are described in PCT International Application Publication No. WO 2016/154258, the entire contents of which is incorporated by reference.
[0253] The embodiments referred to above refer to several drugs being substantially effective in the body at a same time. Several drugs can be administered substantially at the same time, or can be administered at different times but have effect on the body at the same time. For example, this includes administering Aramchol™ before or subsequently, while functioning of Aramchol™ in the body is substantially extant.
[0254] Thus in some embodiments, the method further comprises administering a therapeutically effect amount of a pharmaceutical composition comprising at least one compound selected from the group consisting of: ethyl eicosapentanoate (EPA-E), eicosapentaenoic acid (EPA) and its pharmaceutically acceptable amides, salts, esters and phospholipids; an inhibitor of Acetyl-CoA carboxylase (ACC) alone, or in combination with one or more additional therapeutic agents; pioglitazone hydrochloride or an enantiopure deuterium-enriched pioglitazone; and a peroxisome proliferator activated receptor (PPAR) delta and gamma dual agonist. [0255] In some embodiments, additional therapeutic agents are independently selected from the group consisting of angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacylglycerol O-acyltransferase 1 (DGAT1) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, farnesoid X receptor (FXR) agonists such as obeticholic acid and Px-104, FXR/TGR5 dual agonists, galectin-3 inhibitors such as LIPC- 1010 and GR-MD-02, glucagon-like peptide (GFPI) agonists, glutathione precursors, hepatitis C virus NS 3 protease inhibitors, HMG Co A reductase inhibitors, 1 1— hydroxy steroid dehydrogenase (I I— HSD I) inhibitors, IL-I- antagonists, IF-6 antagonists, IF-10 agonists, IF-1 7 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, FPF gene stimulators, lysyl oxidase homolog 2 (FOXF2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PEC) inhibitors, PPARa agonists, PPAR gamma agonists such as rosiglitazone and pioglitazone, metformin, pentoxyfylline, vitamin E, selenium, omega-3 fatty acids and betaine, PPAR8 agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturasel inhibitors, thyroid hormone receptor- agonists, tumor necrosis factor a (TNFa) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, PTPib inhibitors, ASKI inhibitors, and vascular adhesion protein-1 inhibitors such as PXS4728A, metformin, GR-MD-02, cysteamine bitartrate, simtuzumab, emricasan, GFT-505, CER-002, KD3010, KD3020, MBX8025, LUM002, RP-103, and cenicriviroc.
[0256] The administration of two drugs to treat a given condition, such as non-alcoholic fatty liver disease (NAFLD), raises a number of potential problems. In vivo interactions between two drugs are complex. The effects of any single drug are related to its absorption, distribution, and elimination. When two drugs are introduced into the body, each drug can affect the absorption, distribution, and elimination of the other and hence, alter the effects of the other. For instance, one drug may inhibit, activate or induce the production of enzymes involved in a metabolic route of elimination of the other drug. (Guidance for Industry, 1999) Thus, when two drugs are administered to treat the same condition, it is unpredictable whether each will complement, have no effect on, or interfere with the therapeutic activity of the other in a human subject.
[0257] Not only may the interaction between two drugs affect the intended therapeutic activity of each drug, but the interaction may increase the levels of toxic metabolites (Guidance for Industry, 1999). The interaction may also heighten or lessen the side effects of each drug. Hence, upon administration of two drugs to treat a disease, it is unpredictable what change will occur in the negative side effect profile of each drug. [0258] Additionally, it is difficult to accurately predict when the effects of the interaction between the two drugs will become manifest. For example, metabolic interactions between drugs may become apparent upon the initial administration of the second drug, after the two have reached a steady-state concentration or upon discontinuation of one of the drugs. (Guidance for Industry, 1999)
[0259] It is understood that where a parameter range is provided, all integers within that range, and tenths thereof, are also provided by the invention. For example,“0.2-5 mg/kg/day” is a disclosure of 0.2 mg/kg/day, 0.3 mg/kg/day, 0.4 mg/kg/day, 0.5 mg/kg/day, 0.6 mg/kg/day etc. up to 5.0 mg/kg/day.
[0260] Each embodiment disclosed herein is contemplated as being applicable to each of the other disclosed embodiments. Thus, all combinations of the various elements described herein are within the scope of the invention.
[0261] The following examples are presented in order to more fully illustrate some embodiments of the invention. They should, in no way be construed, however, as limiting the broad scope of the invention.
EXAMPLES
[0262] Examples are provided below to facilitate a more complete understanding of the invention.
The following examples illustrate the exemplary modes of making and practicing the invention. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only.
Example 1 - Thioacetamide (TAA) -induced fibrosis - model for hepatic cirrhosis
[0263] Liver fibrosis was induced in Wistar rats by intraperitoneal injections of TAA (20mg/100 gr body weight) twice per week during 10 weeks. I.p. application of TAA results in hepatic centrolobular necrosis, elevated transaminase activity and robust liver fibrosis. Treatment groups further included co-administration of Aramchol™ (1 or 5 mg/kg orally) or obaticholic acid (OCA, 5 mg/kg). A control group of saline-treated rats (in the absence of TAA administration) was further included. Rats were then sacrificed, and livers were observed macroscopically for signs of cirrhosis and necrotic lesions, and microscopically, following Masson Goldner staining. The fibrosis score, calculated at a scale of 0-4, was determined for each sample, wherein 0 indicates no fibrosis and 4 indicates advanced fibrosis and cirrhosis.
[0264] As can be seen in Figures 1-2, treatment with Aramchol™ (5 mg/kg) significantly prevented TAA induced fibrosis. The treatment reduced significantly the development of necrosis and cirrhosis (Figure 1), as well as the fibrotic score and collagen distribution in the tissue (Figure 2), in a dose- dependent manner. In contradistinction, OCA did not induce statistically significant reduction in these parameters.
[0265] Thus, Aramchol™ was surprisingly found to be a potent anti-fibrotic and anti -cirrhotic agent.
Aramchol™ was also found to be unexpectedly superior to OCA and provide for improved, effective treatment for liver fibrosis.
[0266] Cirrhosis and portal hypertension from TAA intoxication may eventually lead to the development of acute liver failure and associated conditions such as hepatic encephalopathy, and the TAA model is also used in evaluating these phenomena. Accordingly, as disclosed herein, Aramchol™ may also be used in some embodiments for preventing acute or fatal liver failure and/or hepatic or portosystemic encephalopathy, for example toxin-induced liver failure and/or hepatic encephalopathy.
Example 2 - Inhibition of collagen synthesis in stellate cells
[0267] LX2 cells (150.000 cells per well) were plated in DMEM media containing antibiotics, glutamine and bovine fetal serum. After 24 hours incubation, media was changed to 0% serum and incubated for an additional period of 16 hours. Then, Aramchol™ (10 mM) was added and 24 hours later RNA was extracted with Trizol.
[0268] Surprisingly, as can be seen in Figures 3 and 4, COL1A1 expression in LX -2 human hepatic stellate cells was reduced by Aramchol™ via PPARy up-regulation.
[0269] Consistently, Aramchol™ significantly down regulates collagen production in LX -2 human hepatic stellate cells relative to a DMSO control (Ligure 5). Again, Aramchol™ was surprisingly found to be effective in reducing the production of collagen specifically in stellate cells.
Example 3 - Aramchol™ reduces established fibrosis in a MCD diet animal model
[0270] The study described below investigates the mechanism of action of Aramchol™ and its potential effect on fibrosis using the 0.1% methionine- and choline-deficient (0.1MCD) diet mouse model of NASH.
[0271] C57B1/6 were fed the Methionine and Choline Deficient (MCD) and control diet and were sacrificed after 4 weeks. The MCD diet induces aminotransferase elevation and changes in hepatic histological features, characterized by steatosis, local inflammation, hepatocyte necrosis and fibrosis. These changes occur rapidly and are morphologically similar to those observed in human NASH. In this study the MCD diet contained 0.1% methionine to minimize and stabilize weight loss. At the end of the second week, after verification of established NASH, O.lMCD-fed mice were treated orally by gavage with Aramchol™ (5 mg/Kg/day) or vehicle (n=10, each condition). Control diet-fed mice were also treated with vehicle for same duration (n=10). At the end of the experiment, blood and liver samples were obtained. A diagram of the experimental design is shown below:
0.1 MCD diet 0.1 MCD diet + Aramchol TM
5 mg/kg
14 days 14 days
28 days
[0272] Results from the study showed: 1) treatment with Aramchol™ significantly down regulates steatosis in the liver (Figure 6); 2) treatment with Aramchol™ significantly down regulates / normalizes infiltration and activation status of macrophages in the liver (Figure 7); 3) treatment with Aramchol™ significantly down regulates / normalizes fibrosis in the liver (Figure 8); 4) Aramchol™ significantly down regulates collagen in the liver (Figure 9); and 5) Aramchol™ significantly up regulates glutathione and elevates GSH/GSSG ratio in 0.1 % MCD mice (Figure 10).
[0273] Additionally, Aramchol™ treatment further reduced SCD1 activity, which was evidenced by a marked decrease in SCD1 expression, in the FA(16:1)/FA(16:0) ratio and in the total content of monounsaturated FA (MUFA), which led to a reduction in the hepatic content of diglycerides (DG) and TG. Aramchol™ treatment improved oxidative stress, as shown by the normalization of the GSH/GSSG ratio, a biomarker of the cellular redox potential, and a marked reduction in the content of total oxFA including oxLA, which has been associated with liver injury in human NASH.
[0274] The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying current knowledge, readily modify and/or adapt for various applications such specific embodiments without undue experimentation and without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. The means, materials, and steps for carrying out various disclosed functions may take a variety of alternative forms without departing from the invention.
Example 4
(a) Brief Summary
[0275] This is a multicenter, Phase lib, randomized, double blind, placebo-controlled study designed to evaluate the efficacy and safety of two Aramchol™ doses in subjects that are 18 to 75 years of age, with Non-Alcoholic Steatohepatitis (NASH) confirmed by liver biopsy performed in a period of 6 months before entering the study, with overweight or obesity and who are pre diabetic or type II diabetic.
(b) Intervention
[0276] Drug: Aramchol™
[0277] Subjects will be administered Aramchol™ as follows: a. One tablet of Aramchol™ 400 mg and one tablet of matching placebo for Aramchol™. b.One tablet of Aramchol™ 400 mg and one tablet of Aramchol™ 200 mg. c. Two tablet of Aramchol™ matching placebo. The tablets should be taken orally in the morning within 30 min after breakfast with a glass of water (250 ml).
[0278] Subjects are allowed to omit study drugs up to 3 consecutive days during the study.
[0279] Other Name: Placebo
(c) Study Arms
[0280] Experimental: Aramchol™ 600mg a. One tablet of Aramchol™ 400 mg and one tablet of Aramchol™ 200 mg. b. Intervention: Drug: Aramchol™
[0281] Experimental: Aramchol™ 400mg a. One tablet of Aramchol™ 400 mg and one tablet of matching placebo for Aramchol™. b. Intervention: Drug: Aramchol™
[0282] Placebo Comparator: Placebo a. Two tablets of Aramchol™ matching placebo. b. Intervention: Drug: Aramchol™
(d) Estimated Enrollment
[0283] 240
(e) Inclusion Criteria
[0284] Male or female age 18 to 75 years.
[0285] BMI between 25kg/m2 to 40kg/m2 or waist circumference between 88 cm to 200 cm for women, and between 102 cm to 200 cm for men. If there is deviation above the upper limit, please consult the MRI center, to ensure that the machine is suitable for the patient.
[0286] Known type II Diabetes Mellitus or pre-Diabetes according to American Diabetes Association. One of the following 3 criteria is needed for pre-Diabetes: Fasting Plasma Glucose > lOOmg/dl (5.5 mmol/1) or 2hPG following 75g OGTT > 140 (7.8 mmol/1) mg/dl or HbAlc > 5.7%. HbAlc can be repeated at Investigator's discretion.
[0287] Histologically proven Steatohepatitis on a diagnostic liver biopsy performed either during screening or within 6 months before screening visit, confirmed by central laboratory reading of the slides. (Steatosis >1 + inflammation >1 + ballooning >1). Total activity NAS score of 4 or more.
[0288] Liver fat concentration in the liver of 5.5% or more as measured by NMRS.
[0289] Biopsies with an activity NAS score of 4 or more.
[0290] Normal synthetic liver function (serum albumin >3.2g/dl, INR 0.8-1.2, conjugated bilirubin < 35 pmol/L).
[0291] Understanding the nature of the study and signature of the written informed consent. [0292] Negative pregnancy test at study entry for females of child bearing potential.
[0293] Females of child bearing potential practicing reliable contraception throughout the study period (including oral contraceptives) as well as negative pregnancy test at study entry.
[0294] Hypertensive patients must be well controlled by stable dose of anti-hypertensive medication for at least 2 months prior to screening.
[0295] Patients previously treated with vitamin E (>400IU/day), Polyunsaturated fatty acid (>2g/day) or Ursodeoxycholic acid or fish oil can be included if stopped or at least maintained on stable dose at least 3 months prior to diagnostic liver biopsy (and are not started during the trial). These treatments-dosages are allowed if they were stable for at least 12 months prior to biopsy and can remain stable throughout the study. (Dosages less than the amounts stated above are allowed without washout- or stable-period restrictions).
[0296] For patients with type II Diabetes, glycaemia must be controlled (Glycosylated Hemoglobin Ale <9%) while any HbAlc change should not exceed 1.5% during 6 months prior to enrolment). Treatments with anti-diabetic medications (except for those mentioned in Exclusion 16) are permitted if glycaemia is self-monitored by the patient. HbAlc can be repeated at Investigator's discretion.
Exclusion Criteria
[0297] Exclusion Criteria:
[0298] Patients with other active (acute or chronic) liver disease other than NASH (e.g. viral hepatitis, unless eradicated at least 3 years prior to screening; genetic hemochromatosis; Wilson disease; alpha 1 antitripsin deficiency; alcohol liver disease; drug-induced liver disease) at the time of randomization.
[0299] Patients with clinically or histologically documented liver cirrhosis
[0300] Known alcohol and/or any other drug abuse or dependence in the last five years.
[0301] Known history or presence of clinically significant cardiovascular, gastrointestinal, metabolic other than Diabetes Mellitus, neurologic, pulmonary, endocrine, psychiatric, neoplastic disorder or nephrotic syndrome, that in the opinion of the Investigator warrant exclusion from the study. [0302] Patients with familial (i.e., genetic) hypertriglyceridemia and familial (i.e., genetic) hypercholesterolemia.
[0303] History or presence of any disease or condition known to interfere with the absorption distribution, metabolism or excretion of drugs including bile salt metabolites (e.g. inflammatory bowel disease (IBD)), previous intestinal (ileal or colonic) operation, chronic pancreatitis, celiac disease or previous vagotomy. Ongoing Chronic constipation
[0304] Patients with heart or brain pacemaker (i.e., implantable neurological devices).
[0305] Surgery during the last three month before screening which involved stent implantation of metal devices (e.g. knee, hip etc.)
[0306] Weight loss of more than 5% within 6 months prior to randomization.
[0307] History of bariatric surgery within 5 years of liver biopsy.
[0308] Uncontrolled arterial hypertension.
[0309] Women who are pregnant and breast feeding.
[0310] Diabetes Mellitus other than type II (type I, endocrinopathy, genetic syndromes etc.).
[0311] Patients with HIV infection.
[0312] Daily alcohol intake >20 g/day for women and >30 g/day for men (on average per day) as per medical history.
[0313] Treatment with other anti-diabetic medications: GLP-1 receptor agonists and Thiazolidinediones (TZDs), unless started at least 12 months prior to biopsy and on stable dose for 6 months. In case of GLP-1 receptor agonists stopped, it should be at least 6 months before biopsy as per medical history.
[0314] SGLT-2 Inhibitors, Metformin, fibrates, statins, insulin, DPP-4 inhibitors and sulfonylurea unless prescribed dose has been stable for the last 6 months prior to the biopsy.
[0315] Treatment with Valproic acid, Tamoxifen, Methotrexate, Amiodarone or chronic treatment with anti-cholinergic agents, corticosteroids, high dose estrogen and tetracycline within 12 months prior to the screening visit. [0316] Chronic treatment with antibiotics (e.g. Rifaximin).
[0317] Homeopathic and/or alternative treatments. Any treatment should be stopped during the screening period at least 48 hours before randomization.
[0318] Uncontrolled hypothyroidism defined as Thyroid Stimulating hormone >2X the upper limit of normal (ULN). Thyroid dysfunction controlled for at least 6 months prior to screening is permitted.
[0319] Patients with renal dysfunction eGFR< 40.
[0320] Unexplained serum creatine phosphokinase (CPK) >3X the upper limit of normal (UNL).
Patients with a reason for CPK elevation may have the measurement repeated prior to randomization; a CPK retest > 3X ULN leads to exclusion.
[0321] Patients with condition(s) that makes them unsuitable to perform the NMRS (as determined by the PI or the MRI facility).
[0322] Hypersensitivity to Aramchol™ or to any of the excipients in the tablets
[0323] Hypersensitivity to cholic acid or bile acid sequestrants
Detailed Description
[0324] This is a multicenter, Phase ITb, randomized, double blind, placebo-controlled study designed to evaluate the efficacy and safety of two Aramchol™ doses in subjects that are 18 to 75 years of age, with Non-Alcoholic Steatohepatitis (NASH) confirmed by liver biopsy performed in a period of 6 months before entering the study, with overweight or obesity and who are pre diabetic or type II diabetic.
[0325] Eligible subjects will be enrolled into three treatments arms: Aramchol™ 400 and 600 mg tablets and placebo tablets in ratio 2:2:1.
[0326] The subjects will be evaluated at study sites for 11 scheduled visits: at screening (visit l(weeks -4 - 0)), baseline (visit 2 (day 0)), visit 3 (week 2), visit 4 week 4), visit 5 (week 8), visit 6 (week 12), visit 7 (week 24), visit 8 (week 32), visit 9 (week 40) and visit 10 (week 52 - (End of Treatment/early termination visit)). After completion of the study treatment period, the subjects will be followed for an additional period of 13 weeks without study medication (until visit 11 (week 65)). [0327] During the screening period, the severity of the disease will be evaluated with blood tests, liver biopsy and NMRS.
[0328] During the study the following assessments will be performed: a. Vital signs will be measured at each study visit. b.A physical examination will be performed at the screening visit, 24 weeks, End of
Treatment/early termination and week 65 visit.
[0329] The following blood tests will be performed: complete blood count (CBC), serum chemistry (including electrolytes, liver enzymes, direct and total bilirubin, glucose, lipid profile which include triglyceride, cholesterol, HDL, LDL and VLDL, CPK, creatinine, urea, albumin, alkaline phosphatase), ESR and urinalysis during the screening visit, baseline, week 2, 4, 8, 24, 40, 52 and 65 (end of follow up) visits. Serology (HBV, HCV and HIV) will be performed during the screening visit. Coagulation (fibrinogen, PT/INR, aPTT) will be measured in screening and baseline, week 24, End of Treatment/early termination and week 65 visits. Insulin (HOMA) will be measured in the screening, week 24 and End of Treatment/early termination visits. HbAlC will be measured in the screening, week 8, 24, 40 and End of Treatment/early termination visits. C reactive protein, Leptin, Adiponectin, CK-18 (M30 and M65), Ferritin, PAI-1, IL-6, TNF-alpha, FGF-19, C4 (7-alpha-hydroxy-4-cholesten- 3-one), pool serum Bile Acids, B-hydroxybutyrate and Free Fatty Acids will be measured in baseline visit and end of treatment period. The blood samples taken at these visits, will be tested for possible biomarkers, including, but not limited to, Fetuine A and GDF15. TSH, T3 and T4 will be measured during the screening visit. beta-hCG in women of childbearing potential will be performed during the screening visit. A serum sample will be collected and kept frozen until study end in case special investigation needs to be performed. This sample will be collected during the screening and visit 10/Early Termination.
[0330] Body weight and waist circumference will be measured in screening, baseline, week 24, end of treatment and week 65 visits. Height will be measured during the screening visit.
[0331] ECG will be performed during the screening visit, visit 7 (week 24) and end of treatment visits.
[0332] All subjects will undergo two NMRS scans, at screening and end of treatment visits.
[0333] FibroMax test will be performed only if the investigator thinks it is necessary [0334] Liver biopsy will be conducted during the screening and end of treatment visit. The biopsy in the screening visit will be performed only if it was not done within the 6 months prior to this visit.
[0335] Metabolomics blood test will be performed at the screening, visit 7 and the End-of- Treatment/Early Termination visits. From some consenting patients (about 15) a sample from the liver biopsy will be taken for analysis.
[0336] Endothelial Function will be conducted in selected sites. The test will be conducted during the baseline visit before the study treatment will be given and End of Treatment/early termination visit.
[0337] Blood sample for Aramchol™ trough level will be collected (pre-dose) from patients in Israel at baseline (visit 2) week 4 (visit 4), week 12 (visit 6), week 24 (visit 7), week 40 (visit 9), end of treatment (visit 10) and follow up (visit 11). At selected sites in Mexico, USA and Hong Kong one blood sample will be collected (pre-dose) on visit 4 (up to 10 subjects per country) to test for trough Aramchol™ blood level differences between populations (e.g., African American, Asian, Hispanic).
[0338] Blood sample for gene analysis will be taken from all consenting patients during the baseline visit, will be kept frozen and analyzed only at the study end.
[0339] Life style questionnaire will be completed in all visits.
[0340] Adverse events will be monitored throughout the study.
[0341] Concomitant Medications will be monitored throughout the study.
[0342] Telephone contacts will be performed on week 16, 20, 28, 36, 44 and 48. An interim safety analysis will be conducted as soon as 120 subjects will completed the follow up period of 24 weeks under study treatment. An independent DSMB will analyze the safety data and recommend a continued course of action. All patients will continue to be treated under the study protocol until conclusion of the analysis will be known.
[0343] Safety assessment will include frequency and severity of treatment-emergent AEs, clinically significant laboratory abnormalities, ECG changes and physical examination findings.
Results
(i) Primary and Secondary Outcome Measures (400 mg arm) [0344] Treatment with 400 mg of Aramchol™ significantly reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS).
[0345] Treatment with 400 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 10-40%.
[0346] Treatment with 400 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 15%-35%.
[0347] Treatment with 400 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 20% -30%.
[0348] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo.
[0349] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo. The improvement ratio is at least 2 when compared to subjects treated with a placebo.
[0350] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects treated with a placebo.
[0351] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects treated with a placebo.
[0352] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects treated with a placebo.
(ii) Exploratory Outcome Measures (400 mg arm)
[0353] Treatment with 400 mg of Aramchol™ inhibits worsening of the subject’s fibrosis score significantly more than what would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides. [0354] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis improves the subject’s fibrosis score significantly more than what would be expected based on Aramchol™’s effect on the subject’s liver triglycerides.
[0355] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage la hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0356] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage lb hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0357] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage lc hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0358] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage 2 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0359] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage 3 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0360] Treatment with 400 mg of Aramchol™ to subjects afflicted with stage 4 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0361] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis improves the subject’s SAF score more than what would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0362] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo. [0363] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point), compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0364] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0365] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo. The improvement ratio is at least 2 when compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0366] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0367] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0368] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0369] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0370] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0371] Treatment with 400 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0372] Treatment with 400 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0373] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0374] Treatment with 400 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0375] Treatment with 400 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0376] Treatment with 400 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0377] Treatment with 400 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0378] Treatment with 400 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
(iii) Primary and Secondary Outcome Measures (600 mg arm)
[0379] Treatment with 600 mg of Aramchol™ significantly reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS). [0380] Treatment with 600 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 10-40%.
[0381] Treatment with 600 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 15%-35%.
[0382] Treatment with 600 mg of Aramchol™ reduces liver triglycerides ratio as measured by Magnetic Resonance Spectroscopy (MRS) by 20%-30%.
[0383] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo.
[0384] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo. The improvement ratio is at least 2 when compared to subjects treated with a placebo.
[0385] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects treated with a placebo.
[0386] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects treated with a placebo.
[0387] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects treated with a placebo.
(iv) Exploratory Outcome Measures (600 mg arm)
[0388] Treatment with 600 mg of Aramchol™ inhibits worsening of the subject’s fibrosis score significantly more than what would be expected based on Aramchol™’s effect on the subject’s liver triglycerides. [0389] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis improves the subject’s fibrosis score significantly more than what would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0390] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage la hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0391] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage lb hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0392] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage lc hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0393] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage 2 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0394] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage 3 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0395] Treatment with 600 mg of Aramchol™ to subjects afflicted with stage 4 hepatic fibrosis improves the subject’s fibrosis score significantly more than the effect that would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0396] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis improves the subject’s SAF score more than what would be expected based on Aramchol™’ s effect on the subject’s liver triglycerides.
[0397] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo. [0398] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point), compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0399] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0400] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects having fibrosis improvement (i.e. decrease > or = to 1 point) without a worsening of NASH, compared to subjects treated with a placebo. The improvement ratio is at least 2 when compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0401] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0402] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0403] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0404] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0405] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0406] Treatment with 600 mg of Aramchol™ to subjects afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects afflicted with hepatic fibrosis treated with a placebo.
[0407] Treatment with 600 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0408] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0409] Treatment with 600 mg of Aramchol™ results in a significantly higher proportion of subjects with NAS Score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0410] Treatment with 600 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0411] Treatment with 600 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with SAF Activity score improvement (i.e. improvement of at least 2 points) without worsening of fibrosis score, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0412] Treatment with 600 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
[0413] Treatment with 600 mg of Aramchol™ to subjects not afflicted with hepatic fibrosis results in a significantly higher proportion of subjects with NASH resolution (ballooning of 0, inflammation of 0 or 1) without worsening of fibrosis, compared to subjects not afflicted with hepatic fibrosis treated with a placebo.
Discussion
[0414] Based on studies described herein, Aramchol™ is surprisingly found to be a potent anti- fibrotic and anti-cirrhotic agent. Aramchol™ is also found to be unexpectedly superior to OCA and provides improved, effective treatment for liver fibrosis. Accordingly, Aramchol™ may be used to prevent acute or fatal liver failure and/or hepatic or portosystemic encephalopathy, for example toxin- induced liver failure and/or hepatic encephalopathy.
[0415] Furthermore, Aramchol™ is also surprisingly found to be effective in reversing established fibrosis. Aramchol™ treatment improves liver histology as determined by a reduction of lipid accumulation (Sudan red staining), fibrosis (Sirius red and SMA staining) and inflammation (F4/80 and CD64 staining). Indeed, Aramchol™ has an effect on fibrosis in addition to main pathologies of NASH, namely steatosis and inflammation.
[0416] Results presented herein show that Aramchol™ down-regulates collagen production from human stellate cells, the effects of Aramchol™ are mediated through down regulation of SCD 1 and up regulation of glutathione production, and the effect of Aramchol™ on fibrosis is mediated via down regulation of steatosis and inflammation as well as directly via down regulation of collagen production from stellate cells. Taken together, information herein supports the effects of Aramchol™ in human patients as set forth in the claims.
[0417] Results analogous to those of Example 4 for 400 mg or 600 mg doses are expected for higher doses of Aramchol™ that are recited herein.
Example 5: In vivo studies with Aramchol™, a stearoyl Co A desaturase inhibitor, in human subjects with NASH. One-year results of the Global Phase 2b randomized placebo-controlled ARREST Trial.
[0418] In a 3 -month, phase 2a human NAFLD study, Aramchol™ reduced hepatic fat. It also had antifibrotic effects in preclinical models.
[0419] The final results from the global phase 2b ARREST study evaluating a 1-year treatment with Aramchol™ in patients (pts) with NASH are reported herein.
Methods
[0420] Overweigh t/obese pts with pre-diabetes or diabetes and biopsy-proven NASH (with NAS>4;
F<4) were included. Liver biopsy and liver fat measurement by MR spectroscopy (LF-MRS) were performed at baseline and week 52. Both read centrally. The primary endpoint was the absolute change from baseline in LF-MRS (600mg vs. placebo, PLB; mixed model repeated measures). Key secondary endpoints included: NASH resolution without fibrosis worsening, > 1 stage fibrosis reduction without NASH worsening and ALT reduction.
Results
[0421] 247 pts from 11 countries (US, Europe, Latin America) were randomized 2:2:1 to
Aramchol™ 400mg QD (N=101), 600mg QD (N=98) or PLB (N=48). At baseline: 65% females, mean age 54.4yrs, BMI 32.7kg/m2, HbAlc 6.6%±1.0% (mean±SD), LF-MRS 28.5%±11.7% (mean+SD); 60% fibrosis stage 2-3 and NAS 5.12+1.00 (mean+SD). 89% of pts completed the study. The results of the study are summarized in Figures 16-20.
[0422] LF-MRS was significantly reduced vs PLB by 400mg (p=0.045) and 600mg (p<0.066). A statistically significant >5% absolute reduction in liver fat was seen in 47% of pts on 600mg vs 24% on PLB (p<0.028) and 37% on 400mg suggesting a dose response (Figure 16B). Pts in the 600mg arm had NASH resolution without fibrosis worsening more often than PLB: 16.7% vs. 5.0% respectively, (OR=4.74, 95% Cl: 0.99-22.66, p=0.051)(Figure 18A). A > 1 stage fibrosis reduction without NASH worsening occurred in 29.5% of the 600mg arm vs 17.5% of the PLB arm (p=0.21)(Figure 19A). One (1.3%) pt progressed to cirrhosis in the 600mg arm vs 6 (7.5%) in the 400mg arm and 3 (7.5%) in the PLB arm (Figure 19B). Both doses significantly reduced ALT (p<0.001)(Figure 20A), AST (p=0.002)(Figure 20B) and HbAlc (p<0.007) vs PLB in a dose response manner. Serum lipid parameters and body weight did not change. Discontinuation due to adverse events (AE) was < 5% and serious AE occurred in 10% of pts without a difference between arms.
Conclusion
[0423] In a one year, PLB -controlled, phase 2b trial in pts with NASH, Aramchol™ significantly reduced liver fat, improved histology, hepatic biochemistry and glycemic control with excellent safety and tolerability. The therapeutic potency of Aramchol™ for NASH should be tested in a larger, phase 3 trial.
Example 6: In vivo studies with Aramchol™, in human subjects with NASH. Modelling and simulation of 300mg BID and 600mg QD doses of ARAMCHOL™ in patients.
[0424] Table 1 summarizes the reported plasma exposure from three studies - single dose Phase I in healthy volunteers (30-900mg, AUC = AUCmf); Phase II ARAMCHOL™ 003 repeat dose study (AUC on D84 from 100 & 300mg QD), Phase lib Cmin on D84 (400 and 600mg QD). Table 1. Summary of exposure data in healthy volunteers and patients.
[0425] Exposure in healthy volunteers is sub-proportional with an approximate four-fold decrease in AUC/Dose across the dose range 30-900mg (397-97 ng/mL.h/(mg/kg)). Biovailability relative to 30mg/kg has been calculated as (AUC/Dose)/(AUC/30mg) and is plotted for this group in Figure 11.
[0426] Exposure in patients is also sub-proportional from 100 to 300mg. It is noted that the average steady-state AUC in this study at 300mg (32992ng/mL.h) is approximately 2-fold lower than the AUCmf from a single dose in healthy volunteers (64285ng/mL.h). This may be due to the use of a tablet formulation in the patient study versus the powder/suspension in healthy volunteers or may simply reflect a high degree of inter-subject variability (typically exposure CV is >50% within a study group). [0427] The steady state (day 84) Cmm in patients at 400mg and 600mg has been measured. While the AUC in this study was not calculated, as the half-life is relatively long (>24h), Cmm, Cmax & Cavg is expected to be similar and so AUC can be approximated by Cmm*24 to allow comparison with other studies (the true value is expected to be slightly higher). This indicates that exposure at 400mg (68616ng/mL.h) is super-proportional relative to 300mg in patients (32992ng/mL.h) which is contrary to the general trend that AUC/Dose decreases with dose. Again, this may reflect inter-subject variability or a difference in the excipients used in the tablets.
[0428] Plots of AUC v Dose and AUC/Dose v Dose are shown in Figures 12A and 12B.
Modelling and simulation
[0429] Given the sub-proportional exposure generally seen, splitting the same total daily dose into two BID doses is likely to increase plasma AUC. The aim of this analysis was to model this behavior.
[0430] The 300mg data on day 1 and day 84 in patients (Phase 2 ARAMCHOL™ 003) has been modelled in Phoenix 64® assuming single compartment pharmacokinetics (parameterized by Cl and V) and first order absorption at a rate Ka with a delay, Tlag. The fitted model parameters all have a CV < 25% and the predicted concentration data reasonably capture the observed data as shown in
Figure 13A-C.
[0431] These model parameters were used to simulate exposure at 300mg BID as shown in Figure 14. The predicted steady state Cmm using this model is 2630ng/mL with an AUC of 65500 ng/mL.h.
[0432] To predict exposure at 600mg, a correction for sub-proportional PK (lower bioavailability at higher dose) has been made assuming the same non-linearity at observed in healthy volunteers. Based on, Figure 13A-C, relative bioavailability will be approximately 0.75 fold lower at 600mg (ca. 0.4) compared to 300mg (0.53) and the model parameters V/F and Cl/F from the 300mg data have been adjusted accordingly.
[0433] The simulated exposure from a 600mg QD dose is shown in Figure 15 with a predicted Cmm of 1820ng/mL and an AUC of 49100ng/mL.h.
[0434] Given the variability in exposure between subjects and between studies, the absolute values of AUC and Cmm are likely to differ from those predicted but a 300mg BID dose is predicted, on average, to give a Cmm approximately 1.4-fold higher than 600mg QD and a 1.3-fold higher AUC. In summary:
[0435] Exposure of drug is expressed by area under the curve (AUC, the integrated concentration time profile) and is often linear with dose. Under these circumstances, one dose of 900mg Aramchol™ is expected to give three times the exposure (AUC) of 300mg dose. However, it was found that higher doses result in a lower than linear increase in exposure. Accordingly, a single 900mg dose gave only 1.35 fold higher exposure (AUC = 86943ng/mL.h) than a single 300mg dose (AUC = 64285ng/mL.h) in healthy volunteers, despite the 3 fold higher dose. This is likely due to incomplete absorption of Armachol at the higher dose level. Based on this relationship, two 300mg doses, for example, with a 12h dose interval are expected to give higher exposure than a single daily 600mg dose by a factor of approximately 1.4.
Example 7: Mechanistic studies with Aramchol™: Aramchol™ Downregulates SCD1 and Induces PPARD in Hepatic Stellate Cells to Attenuate Cellular Activation and Fibrogenesis.
Background:
[0436] Activation of hepatic stellate cells (HSCs) drives hepatic fibrosis through a process that is inhibited by PPARD signaling. Aramchol™ (arachidyl amido cholanoic acid) is a fatty acid-bile acid conjugate that reduces liver fat content in nonalcoholic fatty liver disease (NAFLD) and improved nonalcoholic steatohepatitis (NASH) without worsening fibrosis in a Phase 2b study. Aramchol™ attenuates fibrosis in two distinct models (MCD diet and thioacetamide). In mice, Aramchol™ reduces liver fat by downregulating the fatty acid synthetic enzyme stearoyl Co-A desaturase 1 (SCD-1) in hepatocytes. The mechanism for Aramchol™’ s antifibrotic effect is not known; moreover, although HSCs also store lipids as retinyl esters. The role of SCD-1 in HSCs and the impact of Aramchol™ on SCD-1 activity are unknown. .
Aim:
[0437] To investigate the direct anti-fibrotic effect of Aramchol™ on HSCs using LX -2, a human hepatic stellate cell line, and to define the mechanism of Aramchol™’ s effects on HSCs.
Methods:
[0438] Serum-starved LX2 cells were treated with Aramchol™ (10 mM) for 24 or 48 hours.
Fibrogenic gene expression and SCD-1 protein expression were measured by qPCR and Western, respectively. RNAseq was performed at 24 and 48 h in duplicate. Differential gene expression was assessed with DSeq2, while gene set enrichment analysis (GSEA) and gene ontology (GO) analyses assessed functionality of gene expression changes. Primary hepatocytes from C57BL/6 mice were harvested by perfusion and treated with Aramchol™ (10 mM) for 48 hours in culture. PPAR y mRNA expression was measured by qPCR.
Results:
[0439] Results are shown in Figures 21-25.
[0440] Figures 21A and 21B depict how Aramchol™ downregulates SCD-1 mRNA and fibrogenic genes and upregulates PPARy mRNA in HSCs. Based on RT-PCR (N = 3) *p<0.05, **p<0.01,
***p<0.001.
[0441] Figures 22A and 22B depict how Aramchol™ downregulates SCD-1 in HSCs. Figure 22A and Figure 22B depict the Western Blot and the densitometry respectively of downregulated SCD-1 protein in HSCs treated with Aramchol™ for 24 or 48 hrs. (N=3) *p<0.05, **p<0.01, ***p<0.001
[0442] Figure 23 depicts the confirmation by RNASeq for downregulation of fibrogenic genes and
SCD-1 mRNA, and upregulation of PPARy mRNA after 48 hrs of treatment with Aramchol™. The cholesterol efflux regulatory protein, ABCA1, is also upregulated.
[0443] Figure 24 depicts that Aramchol™ does not affect PPARy mRNA expression in primary mouse hepatocytes after 48 hours of treatment. (N=3) *p<0.05, **p<0.01, ***p<0.001
[0444] Figures 25A and 25B depict how Aramchol™ downregulates cholesterol biosynthesis and collagen formation in HSCs. Gene Set Enrichment Analysis of LX -2 cells after 24 (Figure 25A) or 48 (Figure 25B) hours of treatment.
Conclusions:
[0445] Aramchol™ elevates P ARy mRNA and down regulates SCD-1 mRNA and protein and in hepatic stellate cells. By RNAseq and pathway analysis, Aramchol™ downregulates fibrogenic genes that are part of a clinically validated HSC activation signature, including COL1A1 and aSMA, as well as pathways involved in cholesterol biosynthesis & homeostasis, and collagen formation. Aramchol™ upregulates PPARy mRNA selectively in HSCs and not in hepatocytes.
Plasma concentrations of Aramchol after oral dosing to steady state with regimens of 300 mg Q12h and 600 mg Q24h [0446] This was a single center, open-label, 2-period, randomized, crossover study, in which subjects received oral doses of Aramchol 300 mg twice daily (Q12 h) for 9 days in one period, with a single dose on the morning of Day 10, and 600 mg once daily (Q24 h) for 10 days in the other period. A pharmacokinetic (PK) profile was obtained over the dosing interval at steady state on Day 10 of each period. Subjects ate a light meal within 30 minutes before each dose of Aramchol.
[0447] The subjects were randomized 1 : 1 so that 8 received each regimen in the first period and the alternate regimen in the second period as shown below:
[0448] Study Population
[0449] Sixteen healthy male or female subjects aged 18-45 years. Main Inclusion criteria: aged 18- 45 years at time of consent; body mass index in the range 18.0-30.9 kg/m2 inclusive; ability to understand the nature of the trial and comply with its requirements; deemed healthy on the basis of a clinical history, physical examination, ECG, vital signs, and clinical laboratory tests; able and willing to give fully informed written consent; agree to use reliable contraception; agree not to donate blood or blood products during the study and for 3 months after; willing to give written consent to have data entered into The Over-volunteering Prevention System (TOPS).
[0450] Study Medication
[0451] The study medication, Aramchol, was packaged and labelled by the sponsor and HMR Pharmacy, in accordance with The Rules Governing Medicinal Products in the European Union, Volume 4: Good Manufacturing Practice (GMP), and with HMR’s Manufacturing Authorization for Investigational Medicinal Products (IMPs). The IMP labels included all the information required by Annex 13 of Volume 4: GMP. The IMP was stored and accounted for according to GMP and HMR Standard Operating Procedures. IMP accountability records were maintained at the site from IMP receipt. IMP supplies were kept in a secure, limited-access, temperature-controlled storage area between 15 and 25 °C. Records of temperature readings for the IMP storage area were kept throughout the duration of the study. At the end of the trial, all unused IMP supplies were returned to the sponsor. Only authorized personnel had access to the IMP.
[0452] Schedule and Procedures [0453] Schedule of Clinical Procedures: Subjects were screened within 21 days before their first dose of study medicine. In each period they remained resident on the ward from Day -1 until the morning of Day 11. An interval of at least 7 days between the last dose in Period 1 and the first dose in Period 2 was specified in the protocol. The actual interval was 30 days. Subjects attended a follow up visit 14 days (+2 days) after the end of the second period. The end of the study was defined as the final follow-up visit by the last subject. A baseline sample was taken before the morning dose on Day 1, in Period 1 only. In both treatment periods, blood samples for assay of plasma Aramchol were taken before the morning doses on Days 7, 8, 9 and 10 and at 1, 2, 4, 6, 8, 10 and 12 h post-dose for the twice-daily dosing regimen on Day 10 with additional samples for the once daily regimen at 16, 20 and 24 h post-dose. Safety and tolerability including vital signs, 12-lead safety ECGs, physical examination, laboratory safety tests, and adverse events were assessed frequently throughout the study until the subject’s last visit.
[0454] Bioanalysis and Pharmacokinetic Analyses: Aramchol in human plasma collected in tubes containing the anticoagulant K2EDTA were assayed after protein precipitation followed by liquid- liquid extraction using LC/MS/MS detection. The lower limit of quantification was 50 ng/mL. The following plasma PK parameters were calculated for each subject on Day 10 of Periods 1 and 2: Area under the concentration-time curve after final dose (AUCtau) during a dosing interval (tau) from zero time (pre-dose) to time tau calculated using the (specified) trapezoidal method with doubling of the value of (AUC12h) for the 300 mg Q12h regimen to obtain the value for 24 h; Average plasma concentration over the dosing interval Cavg calculated by AUCtau; Concentration at the end of the dosing interval Ctau obtained from the observed concentration.
[0455] Statistics: For the primary endpoint the hypotheses to be tested were: Null hypothesis: There is no significant difference in AUCtau between the two dosing intervals (Q12h and Q24h) Alternative hypothesis: There is a significant difference in AUCtau between the two dosing intervals (Q12h and Q24h). The PK data were subjected to a linear mixed effects analysis with period, treatment, and sequence as fixed effects and subject within sequence as a random effect. The geometric mean ratios and associated 90% Confidence Intervals were calculated.
[0456] Results
[0457] Population: Sixteen subjects were enrolled, 3 women and 13 men. Twelve subjects were Caucasian (White), two were Black, one was of mixed Caucasian and Black African heritage and one was Asian (East Asian excluded). Their mean age was 34.3 (range 22 to 43) years. [0458] Safety and Tolerability: Aramchol was safe and well tolerated. There were no deaths or severe AEs and no AE led to early termination. One SAE was reported in Period 2 in which the subject was taking Aramchol 600 mg Q24h. The subject reported diplopia on Dosing Day 9 of Period 2, stating that the symptom had started 2 days earlier. Dosing was continued and the subject completed the study the next day as per protocol. A Left 6th cranial nerve palsy of unknown etiology was diagnosed. There were no other abnormal neurological findings. No abnormality was demonstrated on head CT and MRI. The event was still ongoing one month after the last study dose. Of the 22 AEs reported, 12 (54.5%) were rated mild and, except for the SAE and one other AE of Subject 1006, all resolved rapidly. A similar number of subjects reported AEs during the Aramchol 600 mg Q24h regimen (n=12) and 300 mg Q12h regimen (n=10). The most frequent AEs reported were headache (reported by 4 subjects). All other AEs were single events. No clinically significant abnormalities were documented in vital signs or electrocardiogram (ECG) morphology or intervals during the study. There were no clinically significant abnormalities of laboratory safety parameters, which included complete blood counts, plasma biochemistry and urinalysis.
[0459] Pharmacokinetic Results
[0460] The AUCtau and Cavg calculated by AUCtau/t for each subject with the two dosing regimens and the ratios of AUC0-24h 300 mg Q12h / 600 mg Q24h are shown in Table 1.
[0461] Table 1. Individual AUCtau and Cavg values and Ratios of AUCO-24 values
[0462] * calculated as 2 x AUC0- 12 [0463] Trough concentrations (prior to morning dose) on Days 7, 8, 9 show that all subjects were at steady state before Day 10. Mean + SD values on Days 7 and 10 on the 300 mg Q12h regimen were 4633.6 + 1033.3 and 4526.7 + 1180.4 ng/mL respectively and corresponding values for 600 mg Q24h were 2948.6 +1008.6 and 2837.1 + 942.8 ng/mL respectively. The statistical analyses of geometric mean ratios for AUCO-24 and Cavg over the dosing interval with 90% CIs and Coefficients of Variation (CV%) are shown in Table 4 and Table 5 respectively. The ratio of geometric mean AUCO- 24 values with the 300 mg Q12h regimen relative to the 600 mg Q24h regimen was 1.53 (90% CIs 1.38, 1.69). The mean plasma concentration at the end of the dosing interval Ctau was 4,981 ng/mL after Aramchol 300 mg Q12h and 3031 ng/mL after Aramchol 600 mg Q24h. It is apparent that average exposure with the twice daily dosing regimen was about 50% higher than with the once daily regimen. As expected, the data also show less variability with the Q12h dosing regimen than with the Q24h regimen.
Table 2. AUCtau ng*h/mL Overdosing Interval After Final Dose
Table 3. Cavg ng/mL Overdosing Interval After Final Dose
[0464] Dosing error: On the morning of Day 9, the penultimate day of dosing of Period 2, Subject 1007 should have received Aramchol 300 mg but instead was administered a dose of Aramchol 600 mg in error. The subject had received the correct doses of 300 mg twice daily for the first 8 days. The trial physician and sponsor agreed not to dose the subject that evening so that his treatment did not exceed the scheduled total daily dose. In view of this error, the statistical analysis was repeated with omission of this subject. The analyses of ratios for AUCO-24 and Cavg with 90% CIs and CVs are presented in Table 6 and Table 7 respectively. The mean ratio of geometric mean values for AUCO- 24 with the 300 mg Q12h relative to the 600 mg Q24h regimen was 1.55 (90% CIs 1.39, 1.72). The average plasma concentration over the dosing interval was 5011 ng/mL after Aramchol 300 mg Q12h and 2998 ng/mL after the Aramchol 600 mg Q24h.
Table 4. AUCtau ng.h/mL Overdosing Interval After Final Dose - Subject 1007 Excluded
Table 5. Cavg ng/mL Overdosing Interval After Final Dose With Subject 1007 Excluded
[0465] Discussion and Conclusions: Administration of Aramchol in a dose of 300 mg 12-hourly resulted in 24-hour plasma concentrations significantly greater than those observed with the administration of Aramchol 600 mg once every 24h. Average concentrations were approximately 50% higher and exposure was greater in all 16 subjects with the 12-hourly dosing regimen. As shown in Table 3, the smallest increase was a ratio of 1.07 in a subject who had relatively high exposure on Aramchol 600 mg Q24h and the greatest was 2.69, this occurring in a subject with the lowest exposure in Period 1 when receiving Aramchol 600 mg Q24h. Exclusion of one subject who received a dose of Aramchol 600 mg on the morning of Day 9 instead of the scheduled dose of Aramchol 300 mg made little difference to the results. It is concluded that the twice-daily regimen reliably results in substantially higher exposure. This may translate to greater effectiveness of Aramchol in the treatment of NASH. The higher exposure in this study was not associated with any detriment to safety. It was well tolerated and all subjects completed the 2 x 10 day dosing periods. With the exception of headache, all AE’s were single events and there were no severe AEs. However, there was one SAE, a case of diplopia due to 6th cranial nerve palsy, the cause of which is unknown. The subject had no other neurological findings on examination and CT and MRI showed no abnormalities. The subject reported a common cold during the screening period but to date there is no information to support a viral etiology. The SAE was reported on Day 9 of Period 2 (600mg Q24h) and was still ongoing one month after the last study dose. It was considered unlikely to be related to administration of the investigational product. In conclusion, in contrast to increasing the single daily dose above 600 mg, it is considered that greater exposure to Aramchol is achievable with a 300 mg twice daily regimen. This regimen has been well tolerated by healthy subjects dosed for 10 days. There has been no safety concern in any of the studies in healthy volunteers to date or in the Phase 2 studies in which subjects were treated for up to 1 year.
[0466] While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims (38)

What is claimed is:
1. A method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein the average AUC0-12 is between about 40,000 to about 90,000ng/ml.
2. The method of claim 1, wherein the average AUC0-12 is about 55,000 ng/ml.
3. A method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein the average AUC0-24 is between about 80,000 to about 180,000ng/ml.
4. The method of claim 3, wherein the average AUC0-24 is about 110,000ng/ml.
5. A method for treating hepatic fibrosis in a human subject afflicted with hepatic fibrosis comprising administering to a subject a daily dose of 3 b -arac h i dy I a m i do- 7 a. 12a-dihydroxy-5P-cholan-24-oic acid (Aramchol™), or a pharmaceutically acceptable salt thereof, so as to provide a systemic exposure of said Aramchol™ wherein Cavg per 24h is between about 3000 to about 7000 ng/ml.
6. The method of claim 5, wherein Cavg per 24h is about 4500 ng/ml.
7. The method of any one of the preceding claims, wherein said Aramchol™ is administered in a daily dose of between 400 mg and 900 mg at least twice a day.
8. The method of claim 7, wherein the daily dose amount is 600 mg or 800 mg.
9. The method of claim 7, wherein each administered dose is 300 mg or 400 mg.
10. The method of claim 7, wherein the Aramchol™ is administered twice a day.
1 1. The method of claim 7, wherein the Aramchol™ daily dose is 600 mg and it is administered twice a day wherein each administered dose is 300 mg.
12. The method of any one of the preceding claims, wherein said subject is afflicted with Non- Alcoholic Fatty Liver Disease (NAFLD); the human subject has a ballooning score of at least 1, an inflammation score of at least 1, and a steatosis score of at least 1; the human subject is afflicted with Diabetes Mellitus type II or pre-diabetes; or any combination thereof.
1 3. The method of any one of the preceding claims, wherein the human subject is afflicted with Non- Alcoholic Steatohepatitis (NASH).
14. The method of any one of the preceding claims, wherein the human subject has a NAFLD Activity (NAS) Score of at least 4, at least 5, at least 6, or at least 7.
15. The method any one of the preceding claims, wherein the human subject is not afflicted with Non- Alcoholic Steatohepatitis (NASH).
16. The method of any one of the preceding claims, wherein the hepatic fibrosis is stage 1, 2, 3, or 4 fibrosis.
17. The method of any one of the preceding claims, wherein 300 mg of Aramchol™ is administered to the subject every 12 hours.
18. The method of any one of the preceding claims, wherein the Aramchol™ is administered orally.
19. The method of any one of the preceding claims, wherein the Aramchol™ is administered in the morning, and in the evening.
20. The method of any one of the preceding claims, wherein the Aramchol™ is administered with water, or at the same time as, or within 30 minutes of a meal, preferably wherein the meal is breakfast or dinner, more preferably wherein the meal is a high fat meal or a high calorie meal.
21. The method of any one of the preceding claims, wherein the Aramchol™ is administered over the course of at least 52 weeks, at least 72 weeks, at least 96 weeks, at least 2 years, at least 3 years, or at least 4 years.
22. The method of any one of the preceding claims, wherein said method further comprises lack of worsening of the subject’s NAFLD as defined by Activity (NAS) score, lack of worsening of the subject’s Steatosis, Activity and Fibrosis (SAF) Activity score, reduction of liver fat in said subject, improvement in subject’s Steatosis, improvement in subject’s ballooning, NAFLD resolution, NAFLD resolution without worsening of fibrosis, reduction of fibrosis without NAFLD worsening, reduction of ALT levels in said subject, reduction of AST levels in said subject, reduction of HbAlc levels in said subject, lack of subject’s progression to Cirrhosis, inhibiting progression of Non- Alcoholic Fatty Liver Disease (NAFLD) and/or Non-Alcoholic Steatohepatitis (NASH), or any combination thereof.
23. The method of claim 22, further comprising an improvement of the subject’s NAFLD Activity (NAS) score, or of the subject’s Steatosis, Activity and Fibrosis (SAF) Activity score, an improvement of the subject’ s fibrosis score, or any combination thereof.
24. The method of claim 23 wherein the subject’s NAS score is at least 4 at the commencement of administration of Aramchol™ and the improvement of the subject’s NAS score is an improvement of at least 2 points, contributed by more than one parameter; b) the subject’s SAF Activity score is at least 4 at the commencement of administration of Aramchol™ and improvement of the subject’s SAF Activity score is an improvement of at least 2 points.
25. The method of any one of the preceding claims, wherein said method further reduces liver fat, improves histology, hepatic biochemistry and glycemic control in said subject.
26. The method of any one of the preceding claims, wherein said method has excellent safety and tolerability.
27. The method of claim 22, wherein inhibiting progression of NAFLD or NASH comprises prevention of progression, or reduced progression relative to a patient not treated with Aramchol™.
28. The method of any one of the preceding claims, wherein the human subject is afflicted with Non- Alcoholic Steatohepatitis (NASH) and the method further comprises inhibiting progression of NASH, NASH resolution in the subject or combination thereof.
29. The method of claim 22 wherein the improvement, lack of worsening, or progression is improvement, lack of worsening, or progression at 2, 4, 8, 24, 40, 52, 65, 72, or 96 weeks; 2, 3, or 4 years from the commencement of administration of Aramchol™.
30. The method of claim 28 wherein NASH resolution comprises the human subject having a ballooning score of 0 and an inflammation score of 0 or 1.
31 . The method of any one of the preceding claims, wherein the method further comprises a reduction in the ratio of liver triglycerides to water in the subject relative to the ratio at the commencement of administration of Aramchol™ as measured by MRS.
32. The method of claim 31 wherein there is a 10% to 40% reduction in ratio of liver triglycerides to water.
33. The method of any one of the preceding claims, wherein the method further comprises: a) a reduction in the level of Hemoglobin A1C or HOMA-IR; b) a reduction in the level of Fibrinogen, CK-18, C-reactive protein (CRP), TNFa, IL 6 and fibrosis Tests (NFS; c) a reduction in the ratio of leptin to adinopectin; or d) an increase in the level of adinopectin; in the subject relative to the level or ratio at the commencement of administration of Aramchol™, or e) a reduction in the human subject’s body weight relative to the human subject’s body weight at the commencement of administration of Aramchol™; f) a reduction in the human subject’s waist circumference relative to the human subject’s waist circumference at the commencement of administration of Aramchol™; or g) a reduction in the human subject’s Fatty Liver Index relative to the human subject’s Fatty Liver Index at the commencement of administration of Aramchol™.
34, The method of any one of the preceding claims, wherein the human subject has a diet that is high fat and high calorie; and/or is resistant to lifestyle intervention or is resistant to diet intervention.
35. The method of any one of the preceding claims, further comprising administering a therapeutically effect amount of a pharmaceutical composition comprising at least one compound selected from the group consisting of: a) ethyl eicosapentanoate (EPA-E), eicosapentaenoic acid (EPA) and its pharmaceutically acceptable amides, salts, esters and phospholipids; b) an inhibitor of Acetyl-CoA carboxylase (ACC) alone, or in combination with one or more additional therapeutic agents; c) pioglitazone hydrochloride or an enantiopure deuterium-enriched pioglitazone; d) a peroxisome proliferator activated receptor (PPAR) delta and gamma dual agonists; and e) angiotensin II receptor antagonists, angiotensin converting enzyme (ACE) inhibitors, caspase inhibitors, cathepsin B inhibitors, CCR2 chemokine antagonists, CCR5 chemokine antagonists, chloride channel stimulators, cholesterol solubilizers, diacylglycerol O-acyltransferase 1 (DGAT1) inhibitors, dipeptidyl peptidase IV (DPPIV) inhibitors, farnesoid X receptor (FXR) agonists such as obeticholic acid and Px-104, FXR/TGR5 dual agonists, galectin-3 inhibitors such as FIPC-1010 and GR-MD-02, glucagon-like peptide (GFPI) agonists, glutathione precursors, hepatitis C virus NS3 protease inhibitors, HMG CoA reductase inhibitors, 1 1— hydroxysteroid dehydrogenase (I I— HSD I) inhibitors, IL-I- antagonists, IF-6 antagonists, IF-I 0 agonists, IF-I 7 antagonists, ileal sodium bile acid cotransporter inhibitors, leptin analogs, 5 -lipoxygenase inhibitors, FPF gene stimulators, lysyl oxidase homolog 2 (FOXF2) inhibitors, PDE3 inhibitors, PDE4 inhibitors, phospholipase C (PEC) inhibitors, PPARa agonists, PPAR gamma agonists such as rosiglitazone and pioglitazone, metformin, pentoxyfylline, vitamin E, selenium, omega-3 fatty acids and betaine, PPAR8 agonists, Rho associated protein kinase 2 (ROCK2) inhibitors, sodium glucose transporter-2 (SGLT2) inhibitors, stearoyl CoA desaturasel inhibitors, thyroid hormone receptor- agonists, tumor necrosis factor a (TNFa) ligand inhibitors, transglutaminase inhibitors, transglutaminase inhibitor precursors, PTPib inhibitors, ASKI inhibitors, and vascular adhesion protein-1 inhibitors such as PXS4728A, metformin, GR-MD-02, cysteamine bitartrate, simtuzumab, emricasan, GFT-505, CER-002, KD3010, KD3020, MBX8025, LUM002, RP-103, and cenicriviroc.
36. The method of any one of the preceding claims,, wherein the hepatic fibrosis is caused by contact with a hepatotoxic chemical substance or by mechanical obstruction, malnutrition, hemochromatosis, passive congestion, exposure to poisons or toxins, exposure to drugs, immune reactions, genetically determined sensitivities to a certain substance, infections, bacterial infections, viral infections, parasitic infections, viral hepatitis, syphilis, autoimmune hepatitis, toxin-induced hepatitis, storage or metabolism hepatic disorders, congenital hepatic fibrosis, primary biliary cirrhosis, drug-induced hepatitis, parasitic hepatitis, primary sclerosing cholangitis, Budd-Chiari syndrome, hepatic veno-occlusive disease, portal vein thrombosis, or scarring due to prior liver surgery, preferably said fibrosis is manifested by portal hypertension and/or hepatic cirrhosis.
37. The method of any one of the preceding claims,, wherein the subject is naive to Aramchol™ treatment, or naive to NAFLD treatment.
38. A method of any one of the preceding claims, wherein said fibrosis is being caused by a factor selected from the group consisting of pulmonary fibrosis, heart fibrosis, kidney fibrosis, dermal fibrosis, ocular fibrosis, mucosal fibrosis, fibrosis of the central nervous system, fibrosis in bone or bone marrow, fibrosis in an endocrine organ, fibrosis in the gastro-intestinal system, mediastinal fibrosis, postfibrinous fibrosis, proliferative fibrosis, retroperitoneal fibrosis, pancreatic fibrosis, or fibrosis associated with an autoimmune disease.
AU2019376907A 2018-11-07 2019-10-16 Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients Pending AU2019376907A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16/182,767 2018-11-07
US16/182,767 US20190175619A1 (en) 2016-11-10 2018-11-07 Treatment of fibrosis, and inhibition of fibrosis in non-alcoholic fatty liver disease patients
PCT/IL2019/051121 WO2020095293A1 (en) 2018-11-07 2019-10-16 Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients

Publications (1)

Publication Number Publication Date
AU2019376907A1 true AU2019376907A1 (en) 2021-06-24

Family

ID=70611747

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2019376907A Pending AU2019376907A1 (en) 2018-11-07 2019-10-16 Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients

Country Status (8)

Country Link
EP (1) EP3876945A4 (en)
CN (1) CN113301902A (en)
AU (1) AU2019376907A1 (en)
BR (1) BR112021008896A2 (en)
CA (1) CA3117958A1 (en)
IL (1) IL282854B (en)
MX (1) MX2021005420A (en)
WO (1) WO2020095293A1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3043284C (en) * 2016-11-10 2022-06-28 Galmed Research And Development Ltd. Treatment for fibrosis
US11197870B2 (en) * 2016-11-10 2021-12-14 Galmed Research And Development Ltd Treatment for hepatic fibrosis
US20190175619A1 (en) * 2016-11-10 2019-06-13 Galmed Research And Development Ltd. Treatment of fibrosis, and inhibition of fibrosis in non-alcoholic fatty liver disease patients

Also Published As

Publication number Publication date
EP3876945A1 (en) 2021-09-15
MX2021005420A (en) 2021-09-10
CA3117958A1 (en) 2020-05-14
BR112021008896A2 (en) 2021-08-10
WO2020095293A1 (en) 2020-05-14
IL282854A (en) 2021-06-30
CN113301902A (en) 2021-08-24
EP3876945A4 (en) 2022-08-24
IL282854B (en) 2021-12-01

Similar Documents

Publication Publication Date Title
US11197870B2 (en) Treatment for hepatic fibrosis
AU2017357867B2 (en) Treatment for fibrosis
US20190175619A1 (en) Treatment of fibrosis, and inhibition of fibrosis in non-alcoholic fatty liver disease patients
JP6941109B2 (en) Methods for using FXR agonists
AU2018345817B2 (en) Combination therapy comprising an ACC inhibitor
CN113908158A (en) Methods of using FXR agonists
KR20190044666A (en) Combinations of FXR agonists
KR20190044667A (en) New Therapy of FXR Agents
JP2022541307A (en) Treatments including FXR agonists
US20220362263A1 (en) Treatment for fibrosis and inhibition of fibrosis
AU2019376907A1 (en) Treatment and inhibition of fibrosis in non-alcoholic fatty liver disease patients
US20190275061A1 (en) Inhibition of fibrosis in non alcohol fatty liver disease patients
US20240074998A1 (en) Methods of treatment of cholestatic diseases