WO2023210716A1 - Liver fibrosis therapeutic regimen for cbp/catenin inhibitor - Google Patents

Liver fibrosis therapeutic regimen for cbp/catenin inhibitor Download PDF

Info

Publication number
WO2023210716A1
WO2023210716A1 PCT/JP2023/016556 JP2023016556W WO2023210716A1 WO 2023210716 A1 WO2023210716 A1 WO 2023210716A1 JP 2023016556 W JP2023016556 W JP 2023016556W WO 2023210716 A1 WO2023210716 A1 WO 2023210716A1
Authority
WO
WIPO (PCT)
Prior art keywords
liver
pharmaceutical composition
composition according
drug administration
methyl
Prior art date
Application number
PCT/JP2023/016556
Other languages
French (fr)
Japanese (ja)
Inventor
公則 木村
Original Assignee
地方独立行政法人東京都立病院機構
大原薬品工業株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 地方独立行政法人東京都立病院機構, 大原薬品工業株式会社 filed Critical 地方独立行政法人東京都立病院機構
Publication of WO2023210716A1 publication Critical patent/WO2023210716A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics

Definitions

  • the present invention relates to a pharmaceutical composition for the prevention and treatment of liver diseases, particularly liver diseases such as liver fibrosis and cirrhosis.
  • liver cirrhosis causes liver dysfunction, esophageal varices, and hepatocellular carcinoma, and is responsible for more than 1 million deaths annually worldwide.
  • causes of liver cirrhosis include hepatic steatosis due to excessive drinking and overeating, and hepatitis due to viral infection, and each is treated with alcohol abstinence, diet therapy, and antiviral drugs.
  • chronic cholestasis and subsequent liver damage and cirrhosis can be caused by certain drugs, primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), familial intrahepatic cholangitis, Alagille syndrome, It may be caused by certain diseases such as gestational intrahepatic cholangitis.
  • PBC and PSC are chronic progressive biliary liver diseases caused by autoimmune diseases of unknown cause, and currently there is no effective treatment other than liver transplantation.
  • the prevalence of PBC is 140 cases/million people, and the prevalence of PSC is 0-317 cases/million people.
  • Bile acidic liver damage is the result of destruction of hepatic parenchymal cells due to retention of hydrophobic bile acids (BA).
  • BA hydrophobic bile acids
  • Initial hepatocyte damage induces a subsequent inflammatory response, which exacerbates hepatocyte and intralobar bile duct damage, leading to fibrosis and ultimately liver failure due to cirrhosis.
  • ursodeoxycholic acid and bezafibrate are recognized as therapeutic drugs for PBC, antifibrotic drugs for liver cirrhosis associated with PBC and PSC have not yet been put into practical use (Non-patent Documents 1 and 2).
  • BA receptors such as FXR, its transporter system, and fibroblast growth factor 19 are attracting attention as therapeutic targets for cholestatic liver disease and liver fibrosis (Non-patent Documents 3 and 4).
  • Non-patent Documents 3 and 4 Non-patent Documents 3 and 4
  • Non-Patent Documents 5 to 7 Studies have previously reported that abnormalities in Wnt/ ⁇ -catenin signaling are involved in fibrosis. It has been reported that ⁇ -catenin deletion increased cholestatic liver damage and fibrosis in Mdr2-KO mice, but suppressed cholestatic liver damage and fibrosis in bile duct ligation mice ( Non-Patent Documents 8, 9).
  • Patent No. 6303112 WO2020/122022
  • the present invention aims to provide a pharmaceutical composition useful for preventing or treating liver diseases.
  • CSCs cancer stem cells
  • the present inventor conducted an investigator-initiated domestic trial of foscenvivint in patients with advanced PBC.
  • hemophilia HIV/HCV co-infection hemophilia-associated human immunodeficiency virus and hepatitis C virus
  • liver disease is accompanied by liver fibrosis or cirrhosis.
  • liver fibrosis or cirrhosis is a disease caused by hepatitis B or C, or primary biliary cholangitis (PBC).
  • PBC primary biliary cholangitis
  • liver fibrosis or cirrhosis is a disease caused by hemophilia, HIV/HCV co-infection, or non-alcoholic steatohepatitis.
  • liver disease is classified as A, B or C according to the Child-Pugh score.
  • [7] The pharmaceutical composition according to any one of [1] to [6] above, which is for maintaining or improving liver tissue hardness.
  • [8] The pharmaceutical composition according to [7] above, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the start of drug administration.
  • [12] The pharmaceutical composition according to any one of [1] to [6] above, which is for maintaining or improving a liver reserve index.
  • [13] The pharmaceutical composition according to [12] above, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the start of drug administration.
  • [14] The pharmaceutical composition according to [12] above, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
  • the hepatic reserve index is represented by serum albumin concentration, serum bilirubin concentration, prothrombin activity value, or ALBI score.
  • 280 mg/m 2 is administered intravenously once or twice a week (e.g., twice a week) over 2 to 5 hours (e.g., 4 hours) each time, and the administration is continued for 2 to 6 months ( For example, when continued for 12 weeks), it shows an excellent therapeutic effect on liver cirrhosis (liver fibrosis).
  • FIG. 1 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in Mdr2-KO mice.
  • Mdr2-KO mice MDR2KO, male, 8-10 weeks old
  • FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls.
  • A shows the scheme of the treatment protocol.
  • B shows the results of Sirius red staining (scale bar, 500 ⁇ m, 1 mm, and 500 ⁇ m from the upper figure, respectively).
  • FIG. 2 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in BDL mice.
  • FIG. 3 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in DDC mice.
  • Wild type C57BL/6 mice male, 8 weeks old were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS.
  • the results of hematoxylin and eosin staining and Sirius red staining are shown.
  • FIG. 4 shows that compounds of the present disclosure reduce bile acid (BA) synthesis in Mdr2-KO mice.
  • Mdr2-KO mice (8-10 weeks old) and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls.
  • FIG. 6 shows that compounds of the present disclosure reduce bile acid (BA) synthesis in DDC mice.
  • Wild type C57BL/6 (male, 8 weeks old) mice were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS.
  • FIG. 7 is a diagram showing that the compound of the present disclosure improves liver dysfunction in Mdr2-KO mice.
  • Male Mdr2-KO mice and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks.
  • FIG. 10 is a diagram showing that the compounds of the present disclosure improve liver fibrosis in Mdr2-KO mice.
  • FIG. 11 is a diagram showing that the compound of the present disclosure improves liver fibrosis in DDC mice. Wild type C57BL/6 (male, 8 weeks old) mice were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS.
  • Figure 2 shows changes in serum TBA concentration before (baseline), during (after 4 weeks, after 8 weeks) and after 12 weeks (end of administration) of a compound of the present disclosure.
  • Data represent mean ⁇ SD.
  • FIG. 13 is based on the results of a clinical Phase I/IIa study of compounds of the present disclosure in patients with liver cirrhosis.
  • FIG. 14 is based on the results of a clinical Phase IIa study of compounds of the present disclosure in patients with liver cirrhosis.
  • Figure 2 shows changes in serum albumin concentration before (baseline), during (3 weeks, 6 weeks, 9 weeks after) and 12 weeks (end of administration) administration of a compound of the present disclosure (A). Changes in serum albumin concentration before compound administration (baseline) and 9 months after the start of medication are shown (B).
  • FIG. 15 is based on the results of a clinical trial conducted on patients with liver cirrhosis using the dosing schedule of Protocol A.
  • FIG. 17-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function.
  • FIG. 17-2 shows a case with Child-Pugh (CP) classification B.
  • FIG. 18-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function.
  • Figure 18-2 shows a case with Child-Pugh (CP) classification B.
  • FIG. 19-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function.
  • Figure 19-2 shows a case with Child-Pugh (CP) classification B.
  • Figure 20 is based on the results of a Phase I clinical trial of compounds of the present disclosure in hemophilic HIV/HCV coinfected cirrhotic patients.
  • FIG. 2 is a diagram showing the effect of improving liver fibrosis.
  • FIG. 21 is a diagram showing the effect of the compound of the present disclosure on improving liver fibrosis.
  • the present disclosure provides 4-( ⁇ (6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinoline-8- yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl ⁇ methyl)phenyl dihydrogen phosphate (foscenvivint), a compound of the present disclosure or PRI- 724 or OP-724) or a pharmaceutically acceptable salt thereof as an active ingredient, for the prevention and/or treatment of liver diseases (hereinafter also referred to as "the pharmaceutical composition of the present disclosure")
  • the pharmaceutical composition of the present disclosure provides foscenvivint at a dose of 100 to 400 mg/m 2 (e.g., 280 mg/m 2 ) once or twice a week (e.g.
  • liver cirrhosis Shows therapeutic effect on liver fibrosis. This will be explained in detail below.
  • liver disease refers to any disease in the liver.
  • the liver disease targeted by the present invention may be any liver disease, but may be a disease accompanied by liver fibrosis or even more advanced symptoms of liver cirrhosis in some cases.
  • Liver fibrosis and cirrhosis occur during the repair process of liver cells that have become necrotic due to various liver disorders such as viral hepatitis, alcoholic liver injury, autoimmune liver injury, and metabolic liver injury. This is a state in which the production of fibrous tissues called extracellular matrix is promoted, and these fibrous tissues gradually accumulate as inflammation progresses.
  • liver fibrosis and cirrhosis are not particularly limited, but include viral hepatitis (e.g., hepatitis B, hepatitis C), alcoholic liver damage, autoimmune liver damage (e.g., primary biliary cholangitis), Liver disorders include drug-induced liver injury, metabolic liver injury (eg, fatty liver, non-alcoholic steatohepatitis, hemochromatosis).
  • liver diseases to be prevented or treated with the pharmaceutical composition of the present disclosure include those accompanied by liver fibrosis or cirrhosis.
  • hepatitis B or C with primary biliary cholangitis, hemophilia, HIV/HCV co-infection, or liver fibrosis or cirrhosis due to non-alcoholic steatohepatitis Can be mentioned.
  • prevention or treatment of liver disease means improvement and maintenance of liver function in some embodiments.
  • liver function means all functions possessed by the liver, and is not particularly limited.
  • the functions of the liver include blood storage (e.g., regulation of circulating volume); processing of hemoglobin (e.g., processing and excretion of hemoglobin); production of bile and bile pigments and enterohepatic circulation; Functions in the blood and circulation such as synthesis of phase proteins, albumin, blood clotting factors, steroid binding proteins, other hormone binding proteins, etc.
  • Liver synthesis capacity nutrients and vitamins (glucose and/or other sugars, amino acids, lipids) and/or metabolic functions of nutrients, such as the metabolism of fatty acids, cholesterol, lipoproteins, fat-soluble vitamins, and water-soluble vitamins; various substances (toxins, steroids such as estrogen and androsterone, and other Detoxification or decomposition functions such as inactivation of hormones, etc.); and immune functions.
  • Liver mitochondria also contribute to energy production in the liver through their electron transport chain [electron transport chain function] (Sherlock's Diseases of the Liver & Biliary System, 13th ed. S.Dooley, A.S.F.Lok, G.Garcia-Tsao Other authors, published June 2018, WILEY-BLACKWELL, Inc.).
  • improved/maintenance of liver function refers to improving each of the liver functions described above and preventing or suppressing (maintaining) decline in liver function due to liver dysfunction due to aging and disease.
  • the term also includes therapeutic effects on liver fibrosis and cirrhosis, which cause liver dysfunction.
  • the effect of "improving/maintaining" liver function can also be translated into a therapeutic effect on liver diseases.
  • "improvement/maintenance" of liver function includes, but is not particularly limited to, improvement/maintenance of liver reserve capacity (hepatic synthetic ability), improvement/maintenance of bile production and enterohepatic circulation. .
  • liver reserve capacity is evaluated by a method that measures the amount and function of proteins synthesized in the liver. In some embodiments, it can be assessed by measuring serum albumin concentration as an indicator of hepatic reserve. In some embodiments, serum albumin concentration can be measured by spectrophotometry (eg, Lowry method), electrophoresis-dye densitometry, high performance liquid chromatography-ultraviolet absorption detection, and the like. In some embodiments, it can also be evaluated by measuring the amount of serum alanine aminotransferase (ALT), alkaline phosphatase (ALP), cholinesterase, or cholesterol.
  • ALT serum alanine aminotransferase
  • ALP alkaline phosphatase
  • cholinesterase or cholesterol.
  • other liver-synthesized proteins include prothrombin
  • methods of assessing its function include measuring prothrombin time.
  • prothrombin time When liver function is impaired, bile secretion decreases and absorption of fat-soluble vitamin K is inhibited.
  • coagulation factors II, VII, IX and X are vitamin K dependent and decrease faster than other factors synthesized in the liver.
  • factor VII has the shortest half-life, so when the liver synthesis ability decreases, the prothrombin time in which factor VII is involved is acutely prolonged.
  • factor VII is an extrinsic coagulation factor, over time, the activated partial thromboplastin time involving endogenous coagulation factors also increases.
  • improvement and maintenance of bile and bile pigment production and enterohepatic circulation can be evaluated by measuring total bile acid concentration in serum using cholestasis as an indicator.
  • Bile is produced by liver cells and excreted into the duodenum through the biliary system, but a condition in which the flow of bile is obstructed somewhere along this route is called cholestasis. Bile components remain in the liver and bile ducts and even leak into the blood.
  • Bilirubin is a yellow pigment produced when hemoglobin in old red blood cells breaks down. Bilirubin is carried through the bloodstream to the liver, where it is processed and excreted into the bile. Bilirubin before it is processed by the liver is called “indirect bilirubin", and bilirubin that is processed and enters the bile is called “direct bilirubin”, and the two together are called “total bilirubin”.
  • the Child-Pugh score can be used to assess liver reserve.
  • the Child-Pugh score is a functional evaluation method for liver cirrhosis based on five items: (1) hepatic encephalopathy, (2) ascites, (3) serum bilirubin level, (4) serum albumin level, and (5) prothrombin activity. Yes (Table 1). Based on the total score of each item, the evaluation is divided into three levels: class A (5 to 6 points), class B (7 to 9 points), and class C (10 to 15 points).
  • the liver disease is classified as A, B, or C according to the Child-Pugh score.
  • the liver disease is at least 5, or at least 6, or at least 7, or at least 8, or at least 9, or at least 10, or at least 11, or at least 12, or at least 13, or at least 14, Or classified according to the total score according to the Child-Pugh classification of 15.
  • hepatic reserve can be assessed by improvement in Child-Pugh classification grade (A, B, C).
  • hepatic reserve can also be evaluated by the amount of change in the Child-Pugh score (5 to 15 points). It can also be evaluated by the amount of change in the scores (1 to 3 points) of each evaluation item (hepatic encephalopathy, ascites, serum bilirubin level, serum albumin level, prothrombin activity) that make up the Child-Pugh score.
  • the ALBI grade can be used to assess liver reserve.
  • the (MELD) score can be used to assess liver reserve.
  • the Model for end-stage liver disease (MELD) score is used to predict the short-term prognosis of patients with decompensated liver cirrhosis and to determine suitability for liver transplantation.
  • MELD score is calculated from three items: (1) serum bilirubin, (2) prothrobin time-international normalized ratio (PT-INR), and (3) serum creatinine value.
  • serum bilirubin [Reference value] 0.3 to 1.2 (mg/dL)
  • Unconjugated bilirubin binds to albumin, is transported to the liver, and is taken up by hepatocytes from the sinusoidal side.
  • the taken up bilirubin binds to ligandin, is transported to the endoplasmic reticulum, undergoes glucuronidation by bilirubin UDP-glucuronosyl transfer (BUGT), is transported to the bile canaliculi side, and is removed from the bile canaliculi by multidrug resistance protein 2 (MRP2). via bile duct It is excreted into the duodenum.
  • MRP2 multidrug resistance protein 2
  • Hyperbilirubinemia occurs due to abnormalities in this bilirubin metabolism and excretion pathway. Table 2 shows diseases in which bilirubin increases.
  • Prothrobin time [Reference value] PT: 80-130 (%) Prothrobin time (PT) reflects the overall coagulation activity of extrinsic coagulation factors VII, X, V, and II factors (prothrobin) and fibrinogen. (PT) is the actual measured value (seconds). % activity relative to normal plasma. PT is a more sensitive indicator of liver reserve capacity (protein synthesis ability) than albumin, and its value is lower than that of severe liver failure, vitamin K deficiency, and oral warfarin administration.
  • the therapeutic effect on liver fibrosis and liver cirrhosis can be evaluated by observing the state of fibrosis and defibrosis in the liver parenchymal region.
  • tissue staining using histopathological techniques (eg, Sirius Red staining). Sirius red staining is a staining for evaluating fibrosis, and stains collagen fibers red.
  • the liver tissue stiffness can also be evaluated by measuring the stiffness of the liver tissue, and in some embodiments, the method includes Improvement and maintenance of hardness can be evaluated.
  • a fibroscan test is a test that measures the hardness of the liver and the amount of fat in the liver tissue by applying a special "probe" to the surface of the body and measuring the vibrations and ultrasound transmitted by the probe.
  • the basic principle of MR elastography (MRE) is to convert the vibrational phase of shear elastic waves generated in the liver into the rotational phase of protons using a heating device that generates extracorporeal vibrations, and detect this phase difference using an MRI phase image. , the shear modulus is obtained.
  • the therapeutic effect can also be confirmed by evaluating liver fibrosis by measuring the expression level of collagen genes.
  • the FIB-4 index can be measured to evaluate the degree of progression of liver fibrosis.
  • the FIB-4 index is a scoring system that combines blood test data to evaluate the degree of progression of liver fibrosis. This test method calculates the degree of progression of fibrosis by combining four blood test items: AST value, ALT value, platelet count, and age. This is a score that predicts liver fibrosis in hepatitis C. Recently, it has been used for other liver diseases such as NASH, and it is important to use FIB-4 index as an indicator in fatty liver disease to detect advanced cases of liver fibrosis early in the follow-up of NAFLD patients. It is believed that.
  • APRI can be measured to evaluate the degree of progression of liver fibrosis.
  • APRI aspartate aminotransferase to platelet ratio index
  • the effect of "improving/maintaining" liver function can be evaluated based on each of the above-mentioned indicators.
  • each of the indicators described above can be used to assist in evaluating the effectiveness.
  • the optimal dosage regimen for treatment can be determined.
  • the evaluation indicators include liver tissue stiffness (e.g., fibroscan measurements), liver reserve indicators (e.g., serum albumin levels), and cholestatic indicators (e.g., serum total bile acid levels). At least one, two, or all three can be used.
  • the above evaluation indicators are usually measured before starting drug administration, during the drug administration period, and at the end of drug administration, and are used to evaluate the effect of the drug on improving and maintaining liver function (therapeutic effect on liver disease). I can do it. Furthermore, by measuring after a certain period of time (e.g. 6 to 12 months) after the end of drug administration, it is possible to determine the durability of the drug's further improvement in liver function and maintenance effect (therapeutic effect on liver disease). can be evaluated.
  • the pharmaceutical compositions of the present disclosure contain 4-( ⁇ (6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[( quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl ⁇ methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof .
  • the pharmaceutical compositions of the present disclosure include 4-( ⁇ (6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo- 8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl ⁇ methyl)phenyl dihydrogen phosphate, or pharmaceutically acceptable thereof Contains salt obtained.
  • pharmaceutically acceptable means useful in preparing pharmaceutical compositions that are generally safe, non-toxic, and not biologically or otherwise undesirable. , and is acceptable for human as well as veterinary uses.
  • the term "pharmaceutically acceptable salt” refers to a salt of the compound of the present invention that is pharmaceutically acceptable and has the desired pharmacological activity, as defined above.
  • such salts include inorganic acids, such as, for example, hydrochloric, hydrobromic, sulfuric, nitric, and phosphoric acids; or, for example, acetic, propionic, hexanoic, heptanoic, cyclopentane.
  • pharmaceutically acceptable salts also include base addition salts that may be formed when acidic protons present are capable of reacting with an inorganic or organic base.
  • acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide, and calcium hydroxide.
  • acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • the pharmaceutical compositions of the present disclosure may be in a dosage form that is used orally or parenterally. These dosage forms can be formulated by those skilled in the art by incorporating appropriate combinations of pharmaceutically acceptable carriers and excipients into unit dosage forms as required by generally accepted pharmaceutical practice. can.
  • the pharmaceutical compositions of the present disclosure can be manufactured according to known methods, such as those described in the Japanese Pharmacopoeia or the United States Pharmacopeia (USP).
  • methods of administering the pharmaceutical compositions of the present disclosure include intravenous administration, continuous intravenous administration, and the like.
  • the pharmaceutical composition of the present disclosure is administered to humans as a pharmaceutical preparation (eg, injection, drip) prepared by a conventional method.
  • the administration is at a dose of 100 to 400 mg/m 2 of active ingredient.
  • the administration is at a dose of 200 to 300 mg/m 2 of active ingredient.
  • the administration is in an amount of active ingredient of 200 mg/m 2 , 210 mg/m 2 , 220 mg/m 2 , 230 mg/m 2 , 240 mg/m 2 , 250 mg/m 2 , 260 mg / m2 , 270mg/ m2 , 280mg/ m2 , 290mg/ m2 , 300mg/ m2 .
  • the administration is at a dose of 280 mg/m 2 of active ingredient.
  • the administration is administered intravenously over one or more hours each time at least once a week.
  • the administration is administered intravenously once or twice a week over a period of 2 to 5 hours each time. In some embodiments, the administration is administered intravenously twice a week over a 4 hour period each time.
  • the administration is continued for one month.
  • the administration is continued for 2 to 6 months. In some embodiments, the administration is used to continue for 12 weeks.
  • the pharmaceutical composition of the present disclosure is administered to humans once or twice a week for 2 to 5 hours each time as a conventionally formulated pharmaceutical preparation (e.g., injection, infusion).
  • the drug is administered intravenously over a period of time, and the administration is continued over a period of 2 to 6 months.
  • the pharmaceutical composition of the present disclosure is administered intravenously to humans twice a week for 4 hours each time as a pharmaceutical formulation (e.g., injection, infusion) formulated in a conventional manner. It is used such that the administration is continued for 12 weeks.
  • ALP alkaline phosphatase ALT: alanine aminotransferase
  • BA bile acid
  • BDL bile duct ligation
  • CA cholic acid
  • CDCA chenodeoxycholic acid
  • CK19 cytokeratin-19 (cytokeratin 19)
  • FXR farnesoid X receptor
  • MDR2 multidrug resistance-associated protein 2
  • PBC primary biliary cholangitis
  • PSC primary sclerosing cholangitis
  • TBA total bile acid
  • Mdr2-KO Mouse Model Mdr2-KO mice were purchased from Jackson Laboratory (Bar Harbor, ME, USA) and wild-type littermates on FVB/NJ background were used as controls. . 8-10 week old male Mdr2-KO mice and littermate controls were treated weekly for 10 weeks with 20 mg/kg of a compound of the present disclosure (foscenvivint; Prism BioLab, Tokyo, Japan) dissolved in PBS or PBS as a control. Three intraperitoneal injections were given.
  • mice Female wild-type mice aged 8 to 10 weeks were obtained from Japan SLC (Shizuoka, Japan) and used as previously reported (Osawa Y, et al. Inhibition of Cyclic Adenosine Monophosphate (cAMP)-response Element. -binding Protein (CREB)-binding Protein (CBP)/beta-Catenin Reduces Liver Fibrosis in Mice. eBioMedicine 2, 1751-1758 (2015).) The common bile duct above the pancreas was incised and BDL was performed. The mice received 20 mg/kg of a compound of the present disclosure dissolved in PBS intraperitoneally three times a week.
  • cAMP Cyclic Adenosine Monophosphate
  • CBP -binding Protein
  • Bilirubin Serum bilirubin was measured using the QuantiChrom Bilirubin Assay Kit (BioAssay Systems, Hayward, CA, USA).
  • BA analysis Serum and liver TBA, CA, and CDCA concentrations were determined using the Total Bile Acid Assay Kit, Cholic Acid ELISA Kit, and Chenodeoxycholic Acid ELISA Kit (Cell Biolabs, San Diego, CA, USA), respectively, according to the manufacturer's instructions. Analyzed.
  • RNA extraction DNA removal, and reverse transcription from liver tissue and cultured cells
  • RNeasy and DNase Kits Qiagen, Valencia, CA, USA
  • High-Capacity cDNA Reverse Transcription Kit Applied Biosystems, Foster City, CA, USA
  • qPCR was performed in triplicate on a LightCycler 480 (Roche Applied Science, Mannheim, Germany) using probe and primer sets purchased from Thermo Fisher Scientific and TaqPath qPCR Master Mix, CG (Applied Biosystems).
  • the expression level of the target gene was normalized to the expression level of GAPDH in each sample. (mouse primer set)
  • HE staining is a staining method that allows the cell nucleus and cytoplasm to be dyed separately to observe the morphology of tissues.
  • HE staining makes it possible to grasp the overall picture of cells and tissue structures. Sections are cut from paraffin blocks of liver tissue prefixed with Bouin solution and stained with Lillie- Mayer's Hematoxylin (Muto Chemical Co., Ltd., Japan) and eosin solution (Fujifilm Wako Pure Chemical Industries, Ltd., Japan). (Sirius red staining) To visualize collagen deposition, Bouin-fixed liver sections are stained using picrosirius red solution (Waldeck GmbH & Co., Germany). For quantitative analysis, a digital camera (DFC280, Leica, Germany) was used to capture Sirius Red-stained sections near the central vein at 200x magnification, and positive areas were identified in 5 fields per section using ImageJ. Measure using software (ImageJ, National Institute of Health, USA).
  • Fib-4 Index Measurement The Fib-4 Index is a score that predicts liver fibrosis, and is calculated by combining four items: AST, ALT, platelet count, and age. It is possible to evaluate the degree of progress.
  • Fibroscan test As a non-invasive testing method, Fibroscan is a non-invasive testing method based on the principle of transient elastography, which measures the propagation velocity of pulsed vibration waves in tissues in terms of elasticity (kPa) using ultrasonic image analysis. FibroScan manufactured by ECHOSENS is used.
  • Serum albumin is a protein produced in the liver, and its amount is known to decrease as liver function declines. Serum albumin can be measured according to a conventional method. For example, the amount of albumin in a sample is determined by using a reagent that generates a pigment by binding with albumin and measuring the pigment spectroscopically. Examples of such dyes include bromocresol green, which reacts with albumin at around pH 4.0 to produce a blue albumin-binding dye. A commercially available measurement reagent (Shikaliquid ALB, Kanto Kagaku Co., Ltd.) can also be used. Mix the reagent with the sample and measure the absorbance of the bound dye produced.
  • Example 1 Preclinical test (evaluation in PBC model mice) 1 (hepatic fibrosis improving effect)
  • PBC primary biliary cholangitis
  • BDL mice bile duct ligation model mice
  • MDR2 knockout mice MDR2 knockout mice
  • bile duct injury model mice The hepatic fibrosis-improving effect of the compound of the present disclosure was investigated using DDC mice).
  • MDR2KO mice and BDL mice were treated with compounds of the present disclosure according to the protocols in Figures 1A and 2A, respectively.
  • DDC mice were wild-type C57BL/6 male 8-week-old mice fed with 0.1% DCC feed for 18 days. During that time, compounds of the present disclosure (20 mg/kg three times per week) or PBS were administered intraperitoneally. After the treatment, the liver was collected and the anti-fibrotic effect of the compound of the present disclosure was examined by Sirius red staining of the liver tissue. Sirius red positive areas indicate collagen deposition. For BDL mice and DDC mice, hematoxylin and eosin staining was also performed at the same time. A significant decrease in the Sirius red positive area, ie, a decrease in the fibrotic area, was observed in all mouse model mice (FIGS. 1 to 3), confirming the anti-fibrotic effect of the compound of the present disclosure.
  • Example 2 Preclinical test (evaluation in PBC model mice) 2 (cholestasis improving effect) As in Example 1, the cholestasis-improving effect of the compound of the present disclosure was investigated using BDL mice, Mdr2-KO mice, and DDC mice as PBC model mice. Since bile acid (BA) metabolism is involved in liver fibrosis due to cholestasis, the amount of BA (total bile acids (TBA), cholic acid (CA), and chenodeoxycholic acid (CDCA)) was measured. The results are shown in FIG. 4 (Mdr2-KO mouse), FIG. 5 (BDL mouse), and FIG. 6 (DDC mouse).
  • BA total bile acids
  • CA cholic acid
  • DDC mouse chenodeoxycholic acid
  • Example 3 Preclinical test (evaluation in PBC model mice) 3 (hepatic function improvement effect) As in Example 1, the liver function improving effect of the compound of the present disclosure was investigated using BDL mice, Mdr2-KO mice, and DDC mice as PBC model mice.
  • Serum alanine aminotransferase (ALT) is an enzyme produced by liver cells, and when the liver cells are destroyed, it is released into the blood, resulting in increased levels.
  • Alkaline phosphatase (ALP) is an enzyme that breaks down phosphoric acid compounds. It is produced in the liver, kidneys, intestinal mucosa, bones, etc., and is processed in the liver and released into bile.
  • Bilirubin includes indirect bilirubin before being processed by the liver, and direct bilirubin that is processed into bile (total bilirubin), and direct bilirubin is excreted from the biliary tract.
  • Example 4 Preclinical test (evaluation in PBC model mice) 4 (improving effect on collagen gene expression level) Using Mdr2-KO mice and DDC mice as PBC model mice, the effect of the compound of the present disclosure on improving collagen gene expression was investigated.
  • Collagen particularly type IV collagen
  • Type IV collagen constitutes the basement membrane. Although there is no basement membrane in normal liver sinusoids, proliferation of the basement membrane occurs as liver fibrosis progresses, and the expression level of type IV collagen increases.
  • the expression level of collagen gene in hepatocytes was measured for each PBC model mouse. The results are shown in FIG. 10 (Mdr2-KO mouse) and FIG. 11 (DDC mouse). In all of the PBC model mice, reduction in liver fibrosis was confirmed, as indicated by a decrease in collagen gene expression.
  • Example 5 Clinical trial (evaluation in PBC patients) (cholestasis improving effect) This example is based on the results of a Phase I study designed to examine the safety and tolerability of the disclosed compounds and to determine recommended doses in PBC patients.
  • the subject was a patient who was diagnosed with primary biliary cholangitis and who was diagnosed with advanced fibrosis (Scheuer stage III or higher) as a result of liver histological examination.
  • compounds of the present disclosure were administered intravenously twice a week (4 hours) for 12 weeks.
  • the dose scheduled for the first cycle was administered once 7 days before the first cycle administration by continuous intravenous administration for 4 hours, and safety and pharmacokinetics were evaluated from the day of administration to the day after administration. .
  • TBA concentrations Serum total bile acid concentrations
  • Example 6 (evaluation in hepatitis patients) (improving effect on liver tissue hardness)
  • This example describes a phase I study aimed at evaluating the safety and pharmacokinetics of administering a compound of the present disclosure to patients with HCV or HBV cirrhosis and determining recommended doses of the compound of the present disclosure; Based on the results of a Phase II study aimed at evaluating the efficacy and safety of recommended doses of compounds of the present disclosure administered to patients with HCV or HBV cirrhosis.
  • the dosing schedule in the phase I study, the drug was administered at three doses: (Level 1) 140 mg/m 2 /4 hours, (Level 2) 280 mg/m 2 /4 hours, and (Level 3) 380 mg/m 2 /4 hours.
  • the drug was administered intravenously for 4 consecutive hours twice a week (acceptable range of administration time: ⁇ 15 minutes). This was done with a single dose and 12 cycles (total 12 weeks).
  • the dose scheduled for the first cycle is administered intravenously for 4 consecutive hours (tolerable range of administration time: ⁇ 15 minutes) once 7 days before the first cycle administration, and from the day of administration to the day after administration.
  • the safety and pharmacokinetics were evaluated.
  • Phase II (IIa) study continuous intravenous administration was performed for 4 hours twice a week at the recommended dose determined in Phase I (Level 2). This was done for 12 cycles (total 12 weeks) (Protocol A).
  • liver tissue stiffness was measured using Fibroscan after the completion of a 12-week dosing schedule of compounds of the present disclosure. The results are shown in FIG. A clear decrease in liver tissue stiffness was observed with the drug treatment, confirming the liver tissue stiffness improving effect of the compound of the present disclosure. Particularly good results were obtained with the level 2 dosing schedule.
  • serum albumin concentrations were measured using the Protocol A dosing schedule. The results are shown in Figure 14A. An improving effect on serum albumin concentration was confirmed by administering the compound of the present disclosure. Serum albumin concentrations were determined following the dosing schedule of compounds of the present disclosure in Protocol A. The results are shown in Figure 14B.
  • FIG. 18-1 shows a case with Child-Pugh (CP) classification B.
  • KOM-009 A case in which an albumin preparation was used during administration of the compound of the present disclosure
  • KOM-021 a case in which ascites puncture was performed 6 months after the completion of administration of the compound of the present disclosure
  • KOM-033 A case in which an albumin preparation was used up to 28 days after the end of administration of the compound of the present disclosure.
  • CP class B excluding 3 cases where the evaluation of liver function was affected and 2 cases where results were not obtained 6 months after the end of administration of the compound of the present disclosure (KOM-012, KOM-018).
  • Two of the four patients showed an improvement in their CP score (serum albumin level rating) from 2 points (2.8 to 3.5 g/dL) to 1 point (over 3.5 g/dL) 6 months after the end of treatment.
  • Ta Six months after the end of administration of the compound of the present disclosure, improvement in liver function was suggested due to improvement in serum albumin levels.
  • prothrombin time activity values were determined 6 months after the end of the Protocol A dosing schedule. The results are shown in Figure 19-1.
  • Figure 19-2 shows a case with Child-Pugh (CP) classification B.
  • CP Child-Pugh
  • a case in which an albumin preparation was used during administration of the compound of the present disclosure KOM-009
  • KOM-021 a case in which ascites puncture was performed 6 months after the completion of administration of the compound of the present disclosure
  • KOM-021 a case in which ascites puncture was performed 6 months after the completion of administration of the compound of the present disclosure
  • KOM-033 a case with moderate or higher amount of ascites.
  • KOM-033 in which an albumin preparation was used up to 28 days after the end of administration of the compound of the present disclosure.
  • CP class B excluding 3 cases where the evaluation of liver function was affected and 2 cases where results were not obtained 6 months after the end of administration of the compound of the present disclosure (KOM-012, KOM-018).
  • prothrombin time activity score Three of the four patients showed an improvement in their CP score (prothrombin time activity score) from 2 points (40-70%) to 1 point (over 70%) 6 months after the end of treatment. Six months after the end of administration of the compound of the present disclosure, improvement in the prothrombin time activity value suggested improvement in liver function.
  • Example 7 Evaluation in patients with hemophilia and cirrhosis caused by HIV/HCV co-infection
  • This example is a phase I study aimed at examining the safety and tolerability of administering the compound of the present disclosure to patients with hemophilia and liver cirrhosis caused by HIV/HCV co-infection. Based on results.
  • the dosing schedule was two doses: (Level 1) 140 mg/m2/4 hours and (Level 2) 280 mg/m2/4 hours, twice a week, for 4 consecutive hours (administration time tolerance range: ⁇ 15 minutes). This was done with a single dose and 12 cycles (total 12 weeks).
  • the dose scheduled for the first cycle is administered intravenously for 4 consecutive hours (tolerable range of administration time: ⁇ 15 minutes) once 14 days before the first cycle administration, and from the day of administration to the day after administration.
  • the safety and pharmacokinetics were evaluated.
  • liver tissue stiffness was measured using Fibroscan and FIB-4 index and APRI were calculated. The results are shown in FIG. 20, FIG. 21A, and FIG. 21B, respectively.
  • the liver tissue stiffness measured by Fibroscan showed a decrease from the baseline in both Level 1 and Level 2 of the FIB-4 index and APRI. and APRI showed a greater decline at level 2.
  • a decrease in liver stiffness as determined by Fibroscan, and a decreasing trend in FIB-4 index and APRI score was confirmed.
  • Example 8 Evaluation in patients with decompensated liver cirrhosis This example demonstrates the efficacy of compounds of the present disclosure when administered to patients with decompensated cirrhosis due to HCV or HBV infection, and patients with decompensated cirrhosis due to (suspected) non-alcoholic steatohepatitis; It is based on the design of a Phase II study aimed at evaluating safety and pharmacokinetics.
  • the severity of decompensated cirrhosis is Child-Pugh classification B, and patients with decompensated cirrhosis due to HCV or HBV infection are classified into Cohort A, and patients with decompensated cirrhosis due to non-alcoholic steatohepatitis (including suspected).
  • Cohort B Patients will be divided into Cohort B and will receive the compound or placebo in a double-blind manner.
  • the administration schedule is 280 mg/m2, administered intravenously for 3 to 4 consecutive hours each time (acceptable range of administration time: ⁇ 15 minutes).
  • Cohort A has 3 groups (the compound is administered twice a week, the compound and placebo are administered once a week, and the placebo is administered twice a week), and
  • Cohort B has 2 groups (the compound is administered twice a week). Patients were randomly assigned to a group receiving twice a week administration of a placebo and a group receiving a placebo twice a week, and were administered for 24 weeks, with observation being made until 52 weeks after the start of administration.
  • Efficacy evaluation items include liver reserve index (Child-Pugh score, ALBI score, MELD score, Child-Pugh classification, mALBI grade, serum albumin level, serum bilirubin level, prothrombin activity value), liver fibrosis index (fibroscan Evaluate liver tissue stiffness, MRE liver tissue stiffness, serum fibrosis marker, FIB-4 index).
  • liver reserve index Choild-Pugh score, ALBI score, MELD score, Child-Pugh classification, mALBI grade, serum albumin level, serum bilirubin level, prothrombin activity value
  • liver fibrosis index fibroscan Evaluate liver tissue stiffness, MRE liver tissue stiffness, serum fibrosis marker, FIB-4 index.
  • albumin preparations used to treat ascites events associated with decompensated cirrhosis (esophageal/gastric variceal bleeding, ascites, etc.), and the severity of ascites and hepatic encephalopathy will also be evaluated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

[Problem] To provide a pharmaceutical composition useful for the prevention or treatment of liver disease, in particular, liver fibrosis or liver cirrhosis. [Solution] Provided is a pharmaceutical composition that is for the prevention and/or treatment of liver disease and that contains 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinoline-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazine-6-yl}methyl)phenyl dihydrogen phosphate or a pharmaceutically acceptable salt thereof. The pharmaceutical composition is characterized in that 100 to 400 mg/m2 of the 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinoline-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazine-6-yl}methyl)phenyl dihydrogen phosphate or a pharmaceutically acceptable salt thereof is administered intravenously to a human once or twice a week for 2 to 5 hours each time, and the administration is continued for 2 to 6 months.

Description

CBP/カテニン阻害剤の肝線維症治療レジメンCBP/catenin inhibitor liver fibrosis treatment regimen
 本発明は肝疾患、特に肝線維症や肝硬変等の肝疾患の予防、治療用の医薬組成物に関する。 The present invention relates to a pharmaceutical composition for the prevention and treatment of liver diseases, particularly liver diseases such as liver fibrosis and cirrhosis.
 慢性的な臓器損傷は、肝臓、心臓、腎臓、肺などの患部組織の線維化を引き起こす。肝臓では、線維化は肝機能障害、食道静脈瘤、肝細胞癌を引き起こす肝硬変の発症への第一歩であり、世界中で年間100万人以上の死亡の原因となっている。肝硬変の原因としては、過度の飲酒、過食による肝脂肪症、ウイルス感染による肝炎などがあり、それぞれ禁酒、食事療法、抗ウイルス剤などで治療される。また、慢性胆汁うっ滞とその後の肝障害や肝硬変は、特定の薬剤や原発性胆汁性胆管炎(PBC)、原発性硬化性胆管炎(PSC)、家族性肝内胆道炎、Alagille症候群、妊娠性肝内胆道炎などの特定の疾患によって引き起こされることがある。PBCとPSCは、原因不明の自己免疫疾患による慢性進行性胆汁性肝疾患であり、現在、肝移植以外に有効な治療法はない。PBCの有病率は140症例/百万人、PSCの有病率は0~317症例/百万人である。胆汁酸性肝障害は、疎水性の胆汁酸(BA)が滞留するために肝実質細胞が破壊される結果である。最初の肝細胞の損傷は、その後の炎症反応を誘発し、肝細胞および葉内胆管損傷を悪化させ、線維化を引き起こし、最終的には肝硬変による肝不全となる。PBCの治療薬としてウルソデオキシコール酸やベザフィブラートが認知されているが、PBCやPSCに伴う肝硬変の抗線維化薬はまだ実用化されていない(非特許文献1、2)。胆汁うっ滞性肝疾患や肝線維症の治療標的として、FXRやそのトランスポーター系、線維芽細胞増殖因子19などのBA受容体が注目されている(非特許文献3、4)。しかし、BAの量そのものを低下させる治療薬は現在のところ存在しない。 Chronic organ damage causes fibrosis of affected tissues such as the liver, heart, kidneys, and lungs. In the liver, fibrosis is the first step toward the development of cirrhosis, which causes liver dysfunction, esophageal varices, and hepatocellular carcinoma, and is responsible for more than 1 million deaths annually worldwide. Causes of liver cirrhosis include hepatic steatosis due to excessive drinking and overeating, and hepatitis due to viral infection, and each is treated with alcohol abstinence, diet therapy, and antiviral drugs. In addition, chronic cholestasis and subsequent liver damage and cirrhosis can be caused by certain drugs, primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), familial intrahepatic cholangitis, Alagille syndrome, It may be caused by certain diseases such as gestational intrahepatic cholangitis. PBC and PSC are chronic progressive biliary liver diseases caused by autoimmune diseases of unknown cause, and currently there is no effective treatment other than liver transplantation. The prevalence of PBC is 140 cases/million people, and the prevalence of PSC is 0-317 cases/million people. Bile acidic liver damage is the result of destruction of hepatic parenchymal cells due to retention of hydrophobic bile acids (BA). Initial hepatocyte damage induces a subsequent inflammatory response, which exacerbates hepatocyte and intralobar bile duct damage, leading to fibrosis and ultimately liver failure due to cirrhosis. Although ursodeoxycholic acid and bezafibrate are recognized as therapeutic drugs for PBC, antifibrotic drugs for liver cirrhosis associated with PBC and PSC have not yet been put into practical use (Non-patent Documents 1 and 2). BA receptors such as FXR, its transporter system, and fibroblast growth factor 19 are attracting attention as therapeutic targets for cholestatic liver disease and liver fibrosis (Non-patent Documents 3 and 4). However, there are currently no therapeutic agents that reduce the amount of BA itself.
 Wnt/β-カテニンシグナル伝達の異常が線維化に関与していることは、これまでに研究により報告されている(非特許文献5~7)。β-カテニン欠失は、Mdr2-KOマウスでは胆汁うっ滞による肝損傷と線維化を増加させたが、胆管結紮マウスでは胆汁うっ滞による肝損傷と線維化を抑制することが報告されている(非特許文献8、9)。 Studies have previously reported that abnormalities in Wnt/β-catenin signaling are involved in fibrosis (Non-Patent Documents 5 to 7). It has been reported that β-catenin deletion increased cholestatic liver damage and fibrosis in Mdr2-KO mice, but suppressed cholestatic liver damage and fibrosis in bile duct ligation mice ( Non-Patent Documents 8, 9).
 Wnt経路における細胞内のタンパク質間相互作用の阻害に関連する低分子治療薬として4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート(foscenvivint)が知られており、当該化合物は、肝線維症の予防・治療薬として有用であることが報告されている(特許文献1)。また。当該化合物は、肝機能、特に肝臓の糖代謝機能、電子伝達系機能及び肝合成能における機能低下を改善する作用を有し肝機能改善剤として有効である(特許文献2)。 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo -8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate (foscenvivint) is known. It has been reported that this compound is useful as a prophylactic/therapeutic agent for liver fibrosis (Patent Document 1). Also. The compound has the effect of improving liver function, particularly liver glucose metabolism function, electron transport system function, and liver synthesis ability, and is effective as a liver function improving agent (Patent Document 2).
特許第6303112号公報Patent No. 6303112 WO2020/122022WO2020/122022
 本発明は、肝疾患の予防又は治療に有用な医薬組成物の提供を目的とする。 The present invention aims to provide a pharmaceutical composition useful for preventing or treating liver diseases.
 下記式: The following formula:
で表される化合物、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート(foscenvivint、(別名:ホスセンビビント、PRI-724、OP―724))以下「本開示の化合物」とも称する)は、がん幹細胞(CSC)の制御に不可欠なWnt経路における細胞内のタンパク質間相互作用の阻害に関連する低分子治療薬である。
 本発明者は、foscenvivintの、日本人のB型肝炎ウイルスあるいはC型肝炎ウイルスに起因する肝硬変の患者を対象とした医師主導国内治験に続き、進行性PBC患者を対象とした医師主導国内治験及び血友病合併ヒト免疫不全ウイルスとC型肝炎ウイルスの重複感染(以下、血友病HIV/HCV重複感染)に起因する肝硬変の患者を対象とした医師主導国内治験を開始し、その過程で本開示の化合物の用法用量及び評価指標を鋭意検討し、最適化することに成功し本発明を完成するに至った。
 本開示は、以下の特徴を包含する。
The compound represented by 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro -2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate (foscenvivint, (also known as foscenvivint, PRI-724, OP-724)) hereinafter referred to as "the present disclosure" (also referred to as "compound") is a small molecule therapeutic agent associated with the inhibition of intracellular protein-protein interactions in the Wnt pathway, which is essential for the regulation of cancer stem cells (CSCs).
Following an investigator-initiated domestic trial of foscenvivint in Japanese patients with liver cirrhosis caused by hepatitis B virus or hepatitis C virus, the present inventor conducted an investigator-initiated domestic trial of foscenvivint in patients with advanced PBC. We have started an investigator-initiated domestic clinical trial targeting patients with liver cirrhosis caused by co-infection with hemophilia-associated human immunodeficiency virus and hepatitis C virus (hereinafter referred to as hemophilia HIV/HCV co-infection), and in the process, we have completed this clinical trial. The inventors have diligently studied and optimized the dosage and evaluation index of the disclosed compound, and have completed the present invention.
The present disclosure includes the following features.
[1]100乃至400mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられることを特徴とする、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩を含有する、肝疾患の予防及び/又は治療用医薬組成物。
[2]280mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週2回、毎回4時間かけて静脈内に投与され、前記投与が12週間にわたり継続されるように用いられることを特徴とする、上記[1]記載の医薬組成物。
[3]肝疾患が、肝線維化又は肝硬変を伴う、上記[1]又は[2]記載の医薬組成物。
[4]肝線維化又は肝硬変がB型若しくはC型肝炎、原発性胆汁性胆管炎(PBC)、に起因する疾患であることを特徴とする上記[3]記載の医薬組成物。
[5]肝線維化又は肝硬変が血友病HIV/HCV重複感染、又は非アルコール性脂肪性肝炎に起因する疾患であることを特徴とする[3]記載の医薬組成物。
[6]肝疾患が、Child-Pughスコアによる分類A、BまたはCのものである、[3]記載の医薬組成物。
[7]肝組織硬度の維持又は改善用である、上記[1]~[6]のいずれかに記載の医薬組成物。
[8]維持又は改善が、薬剤投与中または薬剤投与終了時点において、薬剤投与開始前と比較して評価されるものである、上記[7]記載の医薬組成物。
[9]維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、上記[7]記載の医薬組成物。
[10]肝組織硬度がフィブロスキャン測定値で示される、上記[7]記載の医薬組成物。
[11]肝組織硬度がMRエラストグラフィ測定値で示される、[7]記載の医薬組成物。
[12]肝予備能指標の維持又は改善用である、上記[1]~[6]のいずれかに記載の医薬組成物。
[13]維持又は改善が、薬剤投与中または薬剤投与終了時点において、薬剤投与開始前と比較して評価されるものである、上記[12]記載の医薬組成物。
[14]維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、上記[12]記載の医薬組成物。
[15]肝予備能指標が、血清アルブミン濃度、血清ビリルビン濃度、プロトロンビン活性値、又はALBIスコアで示される、上記[12]記載の医薬組成物。
[16]肝予備能指標が、血清ビリルビン濃度、プロトロンビン活性値、又はALBIスコアで示される、[12]記載の医薬組成物。
[17]肝予備能指標が、Child Pugh分類(A,B,C)の変化またはスコア(点数)の変化量で示される、上記[12]記載の医薬組成物。
[1] 100 to 400 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl) ) methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered once a week to humans. 4-({(6S,9S,9aS )-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2, 4] A pharmaceutical composition for the prevention and/or treatment of liver diseases containing triazin-6-yl}methyl)phenyl dihydrogen phosphate or a pharmaceutically acceptable salt thereof.
[2] 280 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl ] Octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered twice weekly to humans. The pharmaceutical composition according to [1] above, which is administered intravenously over 4 hours each time, and is used so that the administration is continued for 12 weeks.
[3] The pharmaceutical composition according to [1] or [2] above, wherein the liver disease is accompanied by liver fibrosis or cirrhosis.
[4] The pharmaceutical composition according to [3] above, wherein the liver fibrosis or cirrhosis is a disease caused by hepatitis B or C, or primary biliary cholangitis (PBC).
[5] The pharmaceutical composition according to [3], wherein the liver fibrosis or cirrhosis is a disease caused by hemophilia, HIV/HCV co-infection, or non-alcoholic steatohepatitis.
[6] The pharmaceutical composition according to [3], wherein the liver disease is classified as A, B or C according to the Child-Pugh score.
[7] The pharmaceutical composition according to any one of [1] to [6] above, which is for maintaining or improving liver tissue hardness.
[8] The pharmaceutical composition according to [7] above, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the start of drug administration.
[9] The pharmaceutical composition according to [7] above, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration, compared to the time point at which drug administration ended.
[10] The pharmaceutical composition according to [7] above, wherein the liver tissue hardness is indicated by a Fibroscan measurement value.
[11] The pharmaceutical composition according to [7], wherein the liver tissue hardness is indicated by an MR elastography measurement value.
[12] The pharmaceutical composition according to any one of [1] to [6] above, which is for maintaining or improving a liver reserve index.
[13] The pharmaceutical composition according to [12] above, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the start of drug administration.
[14] The pharmaceutical composition according to [12] above, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
[15] The pharmaceutical composition according to [12] above, wherein the hepatic reserve index is represented by serum albumin concentration, serum bilirubin concentration, prothrombin activity value, or ALBI score.
[16] The pharmaceutical composition according to [12], wherein the hepatic reserve index is indicated by serum bilirubin concentration, prothrombin activity value, or ALBI score.
[17] The pharmaceutical composition according to [12] above, wherein the hepatic reserve index is indicated by a change in Child Pugh classification (A, B, C) or a change in score.
[18]胆汁うっ滞指標の維持又は改善用である、上記[1]~[6]のいずれかに記載の医薬組成物。
[19]維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、上記[18]記載の医薬組成物。
[20]胆汁うっ滞指標が血清中総胆汁酸濃度で示される、上記[18]記載の医薬組成物。
[21]4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩の肝疾患に対する治療効果の評価を補助する方法であって、肝組織硬度、肝予備能指標及び胆汁うっ滞指標から選択される少なくとも1つを測定することを特徴とする方法。
[22]該測定が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して為されるものである、上記[21]記載の方法。
[23]肝組織硬度がフィブロスキャン測定値で示される、上記[21]又は[22]記載の方法。
[24]肝予備能指標が血清アルブミン濃度で示される、上記[21]又は[22]記載の方法。
[25]胆汁うっ滞指標が血清中総胆汁酸濃度で示される、上記[21]又は[22]記載の方法。
[26]100乃至400mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられることを特徴とする、肝疾患の予防及び/又は治療方法。
[27]280mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週2回、毎回4時間かけて静脈内に投与され、前記投与が12週間にわたり継続されるように用いられることを特徴とする、上記[26]記載の肝疾患の予防及び/又は治療方法。
[28]肝疾患の予防及び/又は治療用医薬組成物の製造における、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩の使用であって、
100乃至400mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられることを特徴とする、使用。
[29]280mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週2回、毎回4時間かけて静脈内に投与され、前記投与が12週間にわたり継続されるように用いられることを特徴とする、上記[28]記載の使用。
[18] The pharmaceutical composition according to any one of [1] to [6] above, which is for maintaining or improving a cholestasis index.
[19] The pharmaceutical composition according to [18] above, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
[20] The pharmaceutical composition according to [18] above, wherein the cholestasis index is indicated by serum total bile acid concentration.
[21] 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H- A method for assisting in evaluating the therapeutic effect of pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, on liver diseases, the method comprising: , liver tissue hardness, liver reserve index, and cholestasis index.
[22] The method according to [21] above, wherein the measurement is performed 6 to 12 months after the end of drug administration, in comparison with the time point at which drug administration ended.
[23] The method according to [21] or [22] above, wherein the liver tissue hardness is indicated by a fibroscan measurement value.
[24] The method according to [21] or [22] above, wherein the hepatic reserve index is indicated by serum albumin concentration.
[25] The method according to [21] or [22] above, wherein the cholestasis index is indicated by serum total bile acid concentration.
[26] 100 to 400 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl) ) methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered once a week to humans. A method for preventing and/or treating liver diseases, which is administered intravenously once or twice over 2 to 5 hours each time, and the administration is continued for 2 to 6 months.
[27] 280 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl ] Octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered twice weekly to humans. The method for preventing and/or treating liver diseases according to [26] above, wherein the method is administered intravenously over 4 hours each time, and the administration is continued for 12 weeks.
[28] 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo- 8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable thereof The use of salt,
100 to 400 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl] Octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered to humans once or twice a week. Use characterized in that it is administered intravenously over a period of 2 to 5 hours each time, and the administration is continued for 2 to 6 months.
[29] 280 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl ] Octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered twice weekly to humans. The use according to [28] above, wherein the use is administered intravenously over 4 hours each time, and the administration is continued for 12 weeks.
 4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート又はその医薬上許容され得る塩は、それを必要とする患者に対して100乃至400mg/m(例、280mg/m)を、週1回又は2回(例、週2回)、毎回2乃至5時間(例、4時間)かけて静脈内に投与され、前記投与が2乃至6か月(例、12週間)にわたり継続されることによって、優れた肝硬変(肝線維化)治療効果を示す。 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2 ,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate or a pharmaceutically acceptable salt thereof is administered at a dosage of 100 to 400 mg/m 2 (e.g. , 280 mg/m 2 ) is administered intravenously once or twice a week (e.g., twice a week) over 2 to 5 hours (e.g., 4 hours) each time, and the administration is continued for 2 to 6 months ( For example, when continued for 12 weeks), it shows an excellent therapeutic effect on liver cirrhosis (liver fibrosis).
図1は本開示の化合物がMdr2-KOマウスの肝線維化を抑制する効果を有することを示す図である。Mdr2-KOマウス(MDR2KO、雄性、8~10週齢)およびFVBバックグラウンドコントロールマウスに、本開示の化合物(20mg/kg、週3回)を10週間投与した。対照としては投与しないマウスを用いた。Aは処置プロトコルのスキームを示す。Bはシリウスレッド染色の結果を示す(スケールバー、上図からそれぞれ500μm、1mm、500μm)。FIG. 1 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in Mdr2-KO mice. Mdr2-KO mice (MDR2KO, male, 8-10 weeks old) and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls. A shows the scheme of the treatment protocol. B shows the results of Sirius red staining (scale bar, 500 μm, 1 mm, and 500 μm from the upper figure, respectively). 図2は本開示の化合物がBDLマウスの肝線維化を抑制する効果を有することを示す図である。野生型C57BL/6マウス(雄性、8~10週齢)にBDLまたはsham手術を施し、本開示の化合物(20mg/kg、週3回)またはPBSで処置した。手術後14日目に動物を安楽死させた。Aは処置プロトコルのスキームを示す。Bはヘマトキシリン・エオシン染色、シリウスレッド染色、およびCK19の免疫組織化学染色(スケールバー、上図からそれぞれ100μm、500μm、および250μm)の結果を示す。FIG. 2 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in BDL mice. Wild type C57BL/6 mice (male, 8-10 weeks old) underwent BDL or sham surgery and were treated with compounds of the present disclosure (20 mg/kg, three times a week) or PBS. Animals were euthanized 14 days after surgery. A shows the scheme of the treatment protocol. B shows the results of hematoxylin and eosin staining, Sirius red staining, and CK19 immunohistochemical staining (scale bars, 100 μm, 500 μm, and 250 μm, respectively, from the upper figure). 図3は本開示の化合物がDDCマウスの肝線維化を抑制する効果を有することを示す図である。野生型C57BL/6マウス(雄性、8週齢)に0.1%DDC飼料を18日間与え、本開示の化合物(20mg/kg、週3回)またはPBSを投与した。ヘマトキシリン・エオシン染色及びシリウスレッド染色(スケールバー、上図からそれぞれ250μmと500μm)の結果を示す。FIG. 3 is a diagram showing that the compound of the present disclosure has the effect of suppressing liver fibrosis in DDC mice. Wild type C57BL/6 mice (male, 8 weeks old) were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS. The results of hematoxylin and eosin staining and Sirius red staining (scale bar, 250 μm and 500 μm from the above figure, respectively) are shown. 図4は、本開示の化合物がMdr2-KOマウスの胆汁酸(BA)合成を減少させることを示した図である。Mdr2-KOマウス(8~10週齢)およびFVBバックグラウンドコントロールマウスに本開示の化合物(20mg/kg、週3回)を10週間投与した。対照としては投与しないマウスを用いた。肝臓および血清のTBA、CA、CDCAレベル(各群n=10)を示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。**p<0.01;****p<0.0001、Student’s t-test。FIG. 4 shows that compounds of the present disclosure reduce bile acid (BA) synthesis in Mdr2-KO mice. Mdr2-KO mice (8-10 weeks old) and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls. Liver and serum TBA, CA, and CDCA levels (n=10 for each group) are shown. Results shown represent the mean±SD of at least three independent experiments. **p<0.01; ***p<0.0001, Student's t-test. 図5は、本開示の化合物が、BDLマウスの胆汁酸(BA)合成を減少させることを示した図である。野生型C57BL/6(雄性、8~10週齢)マウスにBDLまたはsham手術を施し、本開示の化合物(20mg/kg、週3回)またはPBSで処置した。肝臓のTBA、CA、およびCDCAレベル(1群あたりn=6~9)を示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。*p<0.05;**p<0.01;****p<0.0001、one-way ANOVA。FIG. 5 shows that compounds of the present disclosure reduce bile acid (BA) synthesis in BDL mice. Wild type C57BL/6 (male, 8-10 weeks old) mice underwent BDL or sham surgery and were treated with compounds of the present disclosure (20 mg/kg, 3 times a week) or PBS. Liver TBA, CA, and CDCA levels (n=6-9 per group) are shown. Results shown represent the mean±SD of at least three independent experiments. *p<0.05; **p<0.01; ***p<0.0001, one-way ANOVA. 図6は、本開示の化合物が、DDCマウスの胆汁酸(BA)合成を減少させることを示した図である。野生型C57BL/6(雄性、8週齢)マウスに0.1%DDC食を18日間与え、本開示の化合物(20mg/kg、週3回)またはPBSを投与した。肝臓および血清のTBA、CA、CDCAレベル(各群n=6)を示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。**p<0.01、unpaired Student’s t-test。FIG. 6 shows that compounds of the present disclosure reduce bile acid (BA) synthesis in DDC mice. Wild type C57BL/6 (male, 8 weeks old) mice were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS. Liver and serum TBA, CA, and CDCA levels (n=6 for each group) are shown. Results shown represent the mean±SD of at least three independent experiments. **p<0.01, unpaired Student's t-test. 図7は、本開示の化合物が、Mdr2-KOマウスの肝機能障害を改善することを示した図である。雄性Mdr2-KOマウスおよびFVBバックグラウンドコントロールマウスに、本開示の化合物(20mg/kg、週3回)を10週間投与した。対照としては投与しないマウスを用いた。血清ALT、ALP、およびT.Bilレベル(各群n=5~7)を示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。**p<0.01;***p<0.005;****p<0.0001、一元配置分散分析。FIG. 7 is a diagram showing that the compound of the present disclosure improves liver dysfunction in Mdr2-KO mice. Male Mdr2-KO mice and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls. Serum ALT, ALP, and T. Bil levels (n=5 to 7 for each group) are shown. Results shown represent the mean±SD of at least three independent experiments. **p<0.01; ***p<0.005; ***p<0.0001, one-way ANOVA. 図8は、本開示の化合物がBDLマウスの肝機能障害を改善することを示した図である。野生型C57BL/6(雄性、8~10週齢)マウスにBDLまたはsham手術を施し、本開示の化合物(20mg/kg、週3回)またはPBSで処置した。血清ALT、ALP、およびT.Bilレベル(各群n=6~9)を示す。示された結果は、少なくとも3つの独立した実験の代表である。データは平均値±SDを表す。**p<0.01;***p<0.005、one-way ANOVA。FIG. 8 is a diagram showing that the compound of the present disclosure improves liver dysfunction in BDL mice. Wild type C57BL/6 (male, 8-10 weeks old) mice underwent BDL or sham surgery and were treated with compounds of the present disclosure (20 mg/kg, 3 times a week) or PBS. Serum ALT, ALP, and T. Bil level (n=6 to 9 for each group) is shown. Results shown are representative of at least three independent experiments. Data represent mean ± SD. **p<0.01; ***p<0.005, one-way ANOVA. 図9は、本開示の化合物がDDCマウスの肝機能障害を改善することを示した図である。野生型C57BL/6(雄性、8週齢)マウスに0.1%DDC食を18日間与え、本開示の化合物(20mg/kg、週3回)またはPBSを投与した。血清ALT、ALP、およびT.Bilレベル(各群n=6)を示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。**p<0.01、unpaired Student’s t-test。FIG. 9 is a diagram showing that the compound of the present disclosure improves liver dysfunction in DDC mice. Wild type C57BL/6 (male, 8 weeks old) mice were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS. Serum ALT, ALP, and T. Bil level (n=6 for each group) is shown. Results shown represent the mean±SD of at least three independent experiments. **p<0.01, unpaired Student's t-test. 図10は、本開示の化合物が、Mdr2-KOマウスの肝線維化を改善することを示した図である。雄性Mdr2-KOマウスおよびFVBバックグラウンドコントロールマウスに、本開示の化合物(20mg/kg、週3回)を10週間投与した。対照としては投与しないマウスを用いた。RT-qPCRによって決定された肝臓における所定の遺伝子(Col1a1、Col1a2)のmRNA発現レベルを示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。*p<0.05;**p<0.01、一元配置分散分析。FIG. 10 is a diagram showing that the compounds of the present disclosure improve liver fibrosis in Mdr2-KO mice. Male Mdr2-KO mice and FVB background control mice were administered compounds of the present disclosure (20 mg/kg, three times a week) for 10 weeks. Mice that were not administered were used as controls. Shows the mRNA expression levels of certain genes (Col1a1, Col1a2) in the liver determined by RT-qPCR. Results shown represent the mean±SD of at least three independent experiments. *p<0.05; **p<0.01, one-way ANOVA. 図11は、本開示の化合物が、DDCマウスの肝線維化を改善することを示した図である。野生型C57BL/6(雄性、8週齢)マウスに0.1%DDC食を18日間与え、本開示の化合物(20mg/kg、週3回)またはPBSを投与した。RT-qPCRによって決定された肝臓における所定の遺伝子(Col1a1、Col1a2、Col3a1)のmRNA発現レベルを示す。示された結果は、少なくとも3つの独立した実験の平均値±SDを表す。**p<0.01、unpaired Student’s t-test。FIG. 11 is a diagram showing that the compound of the present disclosure improves liver fibrosis in DDC mice. Wild type C57BL/6 (male, 8 weeks old) mice were fed a 0.1% DDC diet for 18 days and administered a compound of the present disclosure (20 mg/kg, three times a week) or PBS. Shows the mRNA expression levels of certain genes (Col1a1, Col1a2, Col3a1) in the liver determined by RT-qPCR. Results shown represent the mean±SD of at least three independent experiments. **p<0.01, unpaired Student's t-test. 図12は、進行したPBC患者(n=5)を対象とした本開示の化合物の臨床第I相試験の結果に基づく。本開示の化合物投与前(baseline)、投与中(4週間後、8週間後)及び12週間後(投与終了)の血清TBA濃度の変化を示す。データは平均値±SDを表す。paired Student’s t-test。FIG. 12 is based on the results of a Phase I clinical trial of compounds of the present disclosure in patients with advanced PBC (n=5). Figure 2 shows changes in serum TBA concentration before (baseline), during (after 4 weeks, after 8 weeks) and after 12 weeks (end of administration) of a compound of the present disclosure. Data represent mean ± SD. paired student's t-test. 図13は、肝硬変患者を対象とした本開示の化合物の臨床第I/IIa相試験の結果に基づく。本開示の化合物投与前(baseline)、および12週間後時(投与終了)の肝組織硬度の変化を示す。paired t-test。肝組織硬度の測定にはフィブロスキャンを用いた。FIG. 13 is based on the results of a clinical Phase I/IIa study of compounds of the present disclosure in patients with liver cirrhosis. Figure 2 shows changes in liver tissue stiffness before administration of a compound of the present disclosure (baseline) and after 12 weeks (end of administration). paired t-test. Fibroscan was used to measure liver tissue hardness. 図14は、肝硬変患者を対象とした本開示の化合物の臨床第IIa相試験の結果に基づく。本開示の化合物投与前(baseline)、投与中(3週間後、6週間後、9週間後)及び12週間後(投与終了)の血清アルブミン濃度の変化を示す(A)。化合物投与前(baseline)、および投薬開始から9か月後の血清アルブミン濃度の変化を示す(B)。FIG. 14 is based on the results of a clinical Phase IIa study of compounds of the present disclosure in patients with liver cirrhosis. Figure 2 shows changes in serum albumin concentration before (baseline), during (3 weeks, 6 weeks, 9 weeks after) and 12 weeks (end of administration) administration of a compound of the present disclosure (A). Changes in serum albumin concentration before compound administration (baseline) and 9 months after the start of medication are shown (B). 図15は、肝硬変患者を対象とし、プロトコルAの投薬スケジュールで実施した臨床試験の結果に基づく。本開示の化合物投与前(baseline)、および12週間後時(投与終了)のFIB-4インデックスの変化を示す。*p<0.05FIG. 15 is based on the results of a clinical trial conducted on patients with liver cirrhosis using the dosing schedule of Protocol A. Figure 3 shows the change in FIB-4 index before administration of a compound of the present disclosure (baseline) and after 12 weeks (end of administration). *p<0.05 左図は各患者(KOM-6, 7, 9, 10, 12, 13, 15, 17, 22, 28, 26, 30, 32)について、投薬開始時(各棒グラフ左)、投薬終了時(12週間後)(各棒グラフ中央)、投薬開始から9か月後(各棒グラフ右)のフィブロスキャン測定値を示す。右図は、投薬開始時と投薬開始から9か月後の肝組織硬度の変化を示す。**p<0.01;paired t-test。The figure on the left shows each patient (KOM-6, 7, 9, 10, 12, 13, 15, 17, 22, 28, 26, 30, 32) at the start of medication (each bar graph on the left) and at the end of medication (12 Fibroscan measurements are shown after 1 week) (center of each bar graph) and 9 months after the start of medication (right side of each bar graph). The figure on the right shows changes in liver tissue hardness at the start of medication and 9 months after the start of medication. **p<0.01; paired t-test. 図17-1は、本開示の化合物が、肝機能を改善する効果を示した図である。図17-2は、病状がChild-Pugh(CP)分類Bの症例を示した。FIG. 17-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function. Figure 17-2 shows a case with Child-Pugh (CP) classification B. 図18-1は、本開示の化合物が、肝機能を改善する効果を示した図である。図18-2は、病状がChild-Pugh(CP)分類Bの症例を示した。FIG. 18-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function. Figure 18-2 shows a case with Child-Pugh (CP) classification B. 図19-1は、本開示の化合物が、肝機能を改善する効果を示した図である。図19-2は、病状がChild-Pugh(CP)分類Bの症例を示した。FIG. 19-1 is a diagram showing the effect of the compound of the present disclosure on improving liver function. Figure 19-2 shows a case with Child-Pugh (CP) classification B. 図20は、血友病HIV/HCV重複感染肝硬変患者を対象とした本開示の化合物の臨床第I相試験の結果に基づく。肝線維化を改善する効果を示した図である。Figure 20 is based on the results of a Phase I clinical trial of compounds of the present disclosure in hemophilic HIV/HCV coinfected cirrhotic patients. FIG. 2 is a diagram showing the effect of improving liver fibrosis. 図21は、本開示の化合物が、肝線維化を改善する効果を示した図である。FIG. 21 is a diagram showing the effect of the compound of the present disclosure on improving liver fibrosis.
 一部の実施形態において、本開示は、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート(foscenvivint、(別名:ホスセンビビント)、本開示の化合物あるいはPRI-724またはOP―724とも称する)又はそれらの医薬上許容され得る塩を有効成分として含む、肝疾患の予防及び/又は治療用の医薬組成物(以下、「本開示の医薬組成物」とも称する)を提供するものであり、本開示の医薬組成物は、それを必要とする患者に対してfoscenvivintとして100乃至400mg/m(例、280mg/m)を、週1回又は2回(例、週2回)、毎回2乃至5時間(例、4時間)かけて静脈内に投与され、前記投与が2乃至6か月(例、12週間)にわたり継続されることによって、優れた肝硬変(肝線維化)治療効果を示す。
 以下、詳細に説明する。
In some embodiments, the present disclosure provides 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinoline-8- yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate (foscenvivint), a compound of the present disclosure or PRI- 724 or OP-724) or a pharmaceutically acceptable salt thereof as an active ingredient, for the prevention and/or treatment of liver diseases (hereinafter also referred to as "the pharmaceutical composition of the present disclosure") The pharmaceutical composition of the present disclosure provides foscenvivint at a dose of 100 to 400 mg/m 2 (e.g., 280 mg/m 2 ) once or twice a week (e.g. , twice a week), administered intravenously over 2 to 5 hours (e.g., 4 hours) each time, and the administration is continued for 2 to 6 months (e.g., 12 weeks) to improve liver cirrhosis ( Shows therapeutic effect on liver fibrosis.
This will be explained in detail below.
 本開示において、「肝疾患」とは、肝臓における任意の疾患を指す。一部の実施形態において、本発明が対象とする肝疾患は、任意の肝疾患でありうるが、肝線維化、場合によってはさらに進行した症状の肝硬変を伴う疾患であり得る。肝線維化や肝硬変は、ウイルス性肝炎、アルコール性肝障害、自己免疫性肝障害、代謝性肝障害などの様々な肝障害により壊死した肝細胞が修復される過程でコラーゲンや複合糖質からなる細胞外マトリックスと呼ばれる線維組織の産生が促進され、炎症の進行とともに徐々にこれらの線維組織が蓄積した状態をいう。肝線維化や肝硬変を引き起こす疾患は特に限定されないが、ウイルス性肝炎(例、B型肝炎、C型肝炎)、アルコール性肝障害、自己免疫性肝障害(例、原発性胆汁性胆管炎)、薬剤性肝障害、代謝性肝障害(例、脂肪肝、非アルコール性脂肪性肝炎、ヘモクロマトーシス)などの肝障害が挙げられる。一部の実施形態において、本開示の医薬組成物において予防・治療対象となる肝疾患としては、肝線維化又は肝硬変を伴うものが挙げられる。また、一部の実施形態において、B型若しくはC型肝炎原発性胆汁性胆管炎、血友病HIV/HCV重複感染、又は非アルコール性脂肪性肝炎に起因する肝線維化又は肝硬変を伴うものが挙げられる。 In this disclosure, "liver disease" refers to any disease in the liver. In some embodiments, the liver disease targeted by the present invention may be any liver disease, but may be a disease accompanied by liver fibrosis or even more advanced symptoms of liver cirrhosis in some cases. Liver fibrosis and cirrhosis occur during the repair process of liver cells that have become necrotic due to various liver disorders such as viral hepatitis, alcoholic liver injury, autoimmune liver injury, and metabolic liver injury. This is a state in which the production of fibrous tissues called extracellular matrix is promoted, and these fibrous tissues gradually accumulate as inflammation progresses. Diseases that cause liver fibrosis and cirrhosis are not particularly limited, but include viral hepatitis (e.g., hepatitis B, hepatitis C), alcoholic liver damage, autoimmune liver damage (e.g., primary biliary cholangitis), Liver disorders include drug-induced liver injury, metabolic liver injury (eg, fatty liver, non-alcoholic steatohepatitis, hemochromatosis). In some embodiments, liver diseases to be prevented or treated with the pharmaceutical composition of the present disclosure include those accompanied by liver fibrosis or cirrhosis. In some embodiments, hepatitis B or C with primary biliary cholangitis, hemophilia, HIV/HCV co-infection, or liver fibrosis or cirrhosis due to non-alcoholic steatohepatitis. Can be mentioned.
 本開示において、肝疾患の予防又は治療とは、一部の実施形態において、肝機能の改善・維持を意味する。 In the present disclosure, prevention or treatment of liver disease means improvement and maintenance of liver function in some embodiments.
 本開示において、「肝機能」とは、肝臓の有する機能全てを意味し、特に制限はない。肝臓が有する機能としては、一部の実施形態において、血液貯蔵(循環量の調整等);血色素の処理(ヘモグロビンの処理排出等);胆汁や胆汁色素の生成と腸肝循環;血漿タンパク質(急性期タンパク質、アルブミン、血液凝固因子、ステロイド結合タンパク質、他のホルモン結合タンパク質等)の合成等の血液および循環における機能[肝合成能];栄養素とビタミン(グルコースおよび/または他の糖類、アミノ酸、脂質および/または脂肪酸、コレステロール、リポタンパク、脂溶性ビタミン、ならびに水溶性ビタミンなど)の代謝などの栄養素の代謝機能[代謝機能];種々の物質(毒素、エストロゲンおよびアンドロステロンなどのステロイド、ならびに他のホルモンなど)の不活性化などの解毒または分解機能;および免疫機能などが挙げられる。また、肝ミトコンドリアはその電子伝達系により肝臓におけるエネルギー生産に寄与している[電子伝達系機能](Sherlock’s Diseases of the Liver & Biliary System, 13th ed. S.Dooley,A.S.F.Lok, G.Garcia-Tsao他著、2018年06月発行、WILEY-BLACKWELL社、参照)。 In the present disclosure, "liver function" means all functions possessed by the liver, and is not particularly limited. In some embodiments, the functions of the liver include blood storage (e.g., regulation of circulating volume); processing of hemoglobin (e.g., processing and excretion of hemoglobin); production of bile and bile pigments and enterohepatic circulation; Functions in the blood and circulation such as synthesis of phase proteins, albumin, blood clotting factors, steroid binding proteins, other hormone binding proteins, etc. [liver synthesis capacity]; nutrients and vitamins (glucose and/or other sugars, amino acids, lipids) and/or metabolic functions of nutrients, such as the metabolism of fatty acids, cholesterol, lipoproteins, fat-soluble vitamins, and water-soluble vitamins; various substances (toxins, steroids such as estrogen and androsterone, and other Detoxification or decomposition functions such as inactivation of hormones, etc.); and immune functions. Liver mitochondria also contribute to energy production in the liver through their electron transport chain [electron transport chain function] (Sherlock's Diseases of the Liver & Biliary System, 13th ed. S.Dooley, A.S.F.Lok, G.Garcia-Tsao Other authors, published June 2018, WILEY-BLACKWELL, Inc.).
 本開示において、肝機能の「改善・維持」とは、上記した各肝機能の改善や、老化および疾患に伴う肝機能障害による肝機能の低下を防止または抑制する(維持する)ことの他、肝機能障害の原因となる肝線維化や肝硬変に対する治療効果をも包含して言う。肝機能の「改善・維持」効果は、ひいては肝疾患に対する治療効果と言い換えることもできる。一部の実施形態において、肝機能の「改善・維持」とは、特に限定されないが、肝予備能(肝合成能)における改善・維持、胆汁の生成や腸肝循環における改善・維持が挙げられる。 In the present disclosure, "improvement/maintenance" of liver function refers to improving each of the liver functions described above and preventing or suppressing (maintaining) decline in liver function due to liver dysfunction due to aging and disease. The term also includes therapeutic effects on liver fibrosis and cirrhosis, which cause liver dysfunction. The effect of "improving/maintaining" liver function can also be translated into a therapeutic effect on liver diseases. In some embodiments, "improvement/maintenance" of liver function includes, but is not particularly limited to, improvement/maintenance of liver reserve capacity (hepatic synthetic ability), improvement/maintenance of bile production and enterohepatic circulation. .
 一部の実施形態において、肝予備能(肝合成能)における改善・維持は、肝臓で合成されるタンパク質の量や機能を測定する方法によって評価される。一部の実施形態において、肝予備能指標として血清アルブミン濃度を測定することによって評価することができる。一部の実施形態において、血清アルブミン濃度は、吸光光度法(例、ローリー法)、電気泳動-染色デンシトメトリー法及び高速液体クロマトグラフ-紫外吸光検出法等によって測定できる。一部の実施形態において、血清アラニンアミノトランスフェラーゼ(ALT)、アルカリホスファターゼ(ALP)、コリンエステラーゼやコレステロールの量を測定することでも評価できる。一部の実施形態において、他の肝臓で合成されるタンパク質としてプロトロンビンが挙げられ、その機能を評価する方法として、プロトロンビン時間の測定がある。肝機能が損なわれると胆汁の分泌が少なくなり、脂溶性のビタミンKの吸収が阻害される。凝固因子の中で、II、VII、IXおよびXはビタミンK依存性で、肝臓で合成される他の因子よりも減少が早い。この中で第VII因子の半減期が一番短いので、肝合成能が低下すると第VII因子が関与するプロトロンビン時間が鋭敏に延長する。さらに、第VII因子は外因系凝固因子であるが、時間が経てば、内因系凝固因子が関与する活性化部分トロンボプラスチン時間も延長する。 In some embodiments, improvement and maintenance of liver reserve capacity (hepatic synthetic capacity) is evaluated by a method that measures the amount and function of proteins synthesized in the liver. In some embodiments, it can be assessed by measuring serum albumin concentration as an indicator of hepatic reserve. In some embodiments, serum albumin concentration can be measured by spectrophotometry (eg, Lowry method), electrophoresis-dye densitometry, high performance liquid chromatography-ultraviolet absorption detection, and the like. In some embodiments, it can also be evaluated by measuring the amount of serum alanine aminotransferase (ALT), alkaline phosphatase (ALP), cholinesterase, or cholesterol. In some embodiments, other liver-synthesized proteins include prothrombin, and methods of assessing its function include measuring prothrombin time. When liver function is impaired, bile secretion decreases and absorption of fat-soluble vitamin K is inhibited. Among the coagulation factors, II, VII, IX and X are vitamin K dependent and decrease faster than other factors synthesized in the liver. Among these factors, factor VII has the shortest half-life, so when the liver synthesis ability decreases, the prothrombin time in which factor VII is involved is acutely prolonged. Furthermore, although factor VII is an extrinsic coagulation factor, over time, the activated partial thromboplastin time involving endogenous coagulation factors also increases.
 一部の実施形態において、胆汁や胆汁色素の生成や腸肝循環における改善・維持は、胆汁うっ滞を指標として血清中の総胆汁酸濃度を測定することによって評価することができる。胆汁は肝細胞でつくられ、胆道系を通って十二指腸に排泄されるが、この経路のどこかで胆汁の流れが阻害されている状態を胆汁うっ滞という。胆汁の成分が肝臓内や胆管内に停滞し、さらには血液中に漏れ出す。ビリルビンは、古くなった赤血球にあるヘモグロビンが壊れてできる黄色い色素である。ビリルビンは血流に乗ってまず肝臓に運ばれ、そこで処理された後に胆汁に排泄される。肝臓で処理される前のビリルビンを「間接ビリルビン」、処理されて胆汁に入ったビリルビンを「直接ビリルビン」と称し、両者を合わせて「総ビリルビン」という。 In some embodiments, improvement and maintenance of bile and bile pigment production and enterohepatic circulation can be evaluated by measuring total bile acid concentration in serum using cholestasis as an indicator. Bile is produced by liver cells and excreted into the duodenum through the biliary system, but a condition in which the flow of bile is obstructed somewhere along this route is called cholestasis. Bile components remain in the liver and bile ducts and even leak into the blood. Bilirubin is a yellow pigment produced when hemoglobin in old red blood cells breaks down. Bilirubin is carried through the bloodstream to the liver, where it is processed and excreted into the bile. Bilirubin before it is processed by the liver is called "indirect bilirubin", and bilirubin that is processed and enters the bile is called "direct bilirubin", and the two together are called "total bilirubin".
 一部の実施形態において、Child-Pugh score(分類)を用いて、肝予備能を評価することができる。 In some embodiments, the Child-Pugh score can be used to assess liver reserve.
 Child-Pugh score(分類)は、(1)肝性脳症、(2)腹水、(3)血清ビリルビン値、(4)血清アルブミン値、(5)プロトロンビン活性の5項目による肝硬変の機能評価法である(表1)。各項目の点数の合計により、classA(5~6点)、classB(7~9点)、classC(10~15点)の3段階に評価される。
The Child-Pugh score (classification) is a functional evaluation method for liver cirrhosis based on five items: (1) hepatic encephalopathy, (2) ascites, (3) serum bilirubin level, (4) serum albumin level, and (5) prothrombin activity. Yes (Table 1). Based on the total score of each item, the evaluation is divided into three levels: class A (5 to 6 points), class B (7 to 9 points), and class C (10 to 15 points).
 一部の実施形態において、肝疾患は、Child-Pughスコアによる分類A、BまたはCである。 In some embodiments, the liver disease is classified as A, B, or C according to the Child-Pugh score.
 一部の実施形態において、肝疾患は、少なくとも5、または少なくとも6、または少なくとも7、または少なくとも8、または少なくとも9、または少なくとも10、または少なくとも11、または少なくとも12、または少なくとも13、または少なくとも14、または15のChild-Pugh分類による全スコアによって分類される。 In some embodiments, the liver disease is at least 5, or at least 6, or at least 7, or at least 8, or at least 9, or at least 10, or at least 11, or at least 12, or at least 13, or at least 14, Or classified according to the total score according to the Child-Pugh classification of 15.
 一部の実施形態において、肝予備能は、Child-Pugh分類のグレード(A,B,C)の改善によって評価することができる。 In some embodiments, hepatic reserve can be assessed by improvement in Child-Pugh classification grade (A, B, C).
 一部の実施形態において、肝予備能は、Child-Pughスコアの点数(5~15点)の変化量によっても評価することができる。また、Child-Pughスコアを構成する各評価項目(肝性脳症、腹水、血清ビリルビン値、血清アルブミン値、プロトロンビン活性)の評点(1~3点)の変化量によっても評価することができる。 In some embodiments, hepatic reserve can also be evaluated by the amount of change in the Child-Pugh score (5 to 15 points). It can also be evaluated by the amount of change in the scores (1 to 3 points) of each evaluation item (hepatic encephalopathy, ascites, serum bilirubin level, serum albumin level, prothrombin activity) that make up the Child-Pugh score.
 一部の実施形態において、ALBI gradeを用いて、肝予備能を評価することができる。 In some embodiments, the ALBI grade can be used to assess liver reserve.
 ALBI grade(Albumin―bilirubin grade)は、アルブミン値とビリルビン値のみで計算されるALBI scoreを元に肝予備能を3段階に評価する肝予備能評価方法である。 [(0.66×log10ビリルビン(μmol/L))+(-0.085×アルブミン(g/L)):grade 1:2:3 = ≦-2.60:>-2.60 to ≦-1.39:>-1.39。 ALBI grade (Albumin-bilirubin grade) is a liver reserve evaluation method that evaluates liver reserve into three levels based on the ALBI score calculated only from albumin and bilirubin values. [(0.66×log 10 bilirubin (μmol/L)) + (-0.085×albumin (g/L)): grade 1:2:3 = ≦-2.60: >-2.60 to ≦-1.39: >-1.39.
 一部の実施形態において、(MELD)scoreを用いて、肝予備能を評価することができる。 In some embodiments, the (MELD) score can be used to assess liver reserve.
 Model for end―stage liver disease(MELD)scoreは、非代償性肝硬変患者の短期予後予測及び肝移植適応の判断に用いられる。(MELD)scoreは、(1)血清ビリルビン、(2)プロトロビン時間-国際標準化比(PT-INR)、(3)血清クレアチニン値の3項目より、算出される。 The Model for end-stage liver disease (MELD) score is used to predict the short-term prognosis of patients with decompensated liver cirrhosis and to determine suitability for liver transplantation. (MELD) score is calculated from three items: (1) serum bilirubin, (2) prothrobin time-international normalized ratio (PT-INR), and (3) serum creatinine value.
血清ビリルビン
[基準値]0.3~1.2(mg/dL)
 網内系で老廃赤血球のヘモグロビンが分解れヘムが開裂し非抱合型(間接) ビリルビンが産生れる。非抱合型ビリルビンはアルブミンと結合し、肝へ運ばれ類洞側ら肝細胞へ取り込まれる。取り込まれたビリルビンはリガンディンに結合し、小胞体に運ばれbilirubin UDP―glucuronosyl transferase(BUGT)によりグルクロン酸抱合を受け、毛細胆管側へ運ばれmultidrug resistance protein 2(MRP2)により毛細胆管から胆管を経由し十二指腸へ排泄される。このビリルビン代謝、排泄経路の異常により高ビリルビン血症が生じる。ビリルビンが上昇る疾患を表2示す。
serum bilirubin
[Reference value] 0.3 to 1.2 (mg/dL)
In the reticuloendothelial system, hemoglobin in old red blood cells is degraded, heme is cleaved, and unconjugated (indirect) bilirubin is produced. Unconjugated bilirubin binds to albumin, is transported to the liver, and is taken up by hepatocytes from the sinusoidal side. The taken up bilirubin binds to ligandin, is transported to the endoplasmic reticulum, undergoes glucuronidation by bilirubin UDP-glucuronosyl transfer (BUGT), is transported to the bile canaliculi side, and is removed from the bile canaliculi by multidrug resistance protein 2 (MRP2). via bile duct It is excreted into the duodenum. Hyperbilirubinemia occurs due to abnormalities in this bilirubin metabolism and excretion pathway. Table 2 shows diseases in which bilirubin increases.
 プロトロビン時間(PT)
[基準値]PT:80~130(%)
 プロトロビン時間(PT)は外因系凝固因子である第VII、X、V、II因子プロトロビン)、フィブリノーゲンの総合的凝固活性を反映する。(PT)は、実測値(秒)。正常血漿に対する活性%。PTはアルブミンよりも肝予備能(蛋白合成能)の鋭敏な指標であり、重症肝障害、ビタミンK欠乏症、ワルファリン内服より低値となる。
Prothrobin time (PT)
[Reference value] PT: 80-130 (%)
Prothrobin time (PT) reflects the overall coagulation activity of extrinsic coagulation factors VII, X, V, and II factors (prothrobin) and fibrinogen. (PT) is the actual measured value (seconds). % activity relative to normal plasma. PT is a more sensitive indicator of liver reserve capacity (protein synthesis ability) than albumin, and its value is lower than that of severe liver failure, vitamin K deficiency, and oral warfarin administration.
 一部の実施形態において、肝線維化や肝硬変に対する治療効果は、肝実質領域の線維化及び脱線維化の様子を観察することによって評価することができる。一部の実施形態において、病理組織学的手法を用いた組織染色(例、シリウスレッド染色)による。シリウスレッド染色は、線維化を評価するための染色であり、コラーゲン線維を赤色に染色する。一部の実施形態において、さらに、肝組織硬度を測定することによっても評価することができ、一部の実施形態において、その手法としては、例えばフィブロスキャン検査により得られる測定値を指標として肝組織硬度の改善・維持を評価することができる。フィブロスキャン検査とは、体の表面に特殊な「プローブ」をあて、そこから発せられる振動と超音波の伝わり方から肝臓の硬さや肝臓組織内の脂肪量を測ることができる検査である。MRエラストグラフィ(MRE)の基本的原理は、体外振動を起こす加配装置により肝内に生じたずり弾性波の振動位相をプロトンの回転位相に変換させ、この位相差をMRIの位相画像で検出し、ずり弾性率が得られる。肝臓内を伝搬する弾性波の速度(V)は物質のずり弾性率(μ)と密度(ρ)を用いてV=μ/ρの式で表される。 In some embodiments, the therapeutic effect on liver fibrosis and liver cirrhosis can be evaluated by observing the state of fibrosis and defibrosis in the liver parenchymal region. In some embodiments, by tissue staining using histopathological techniques (eg, Sirius Red staining). Sirius red staining is a staining for evaluating fibrosis, and stains collagen fibers red. In some embodiments, the liver tissue stiffness can also be evaluated by measuring the stiffness of the liver tissue, and in some embodiments, the method includes Improvement and maintenance of hardness can be evaluated. A fibroscan test is a test that measures the hardness of the liver and the amount of fat in the liver tissue by applying a special "probe" to the surface of the body and measuring the vibrations and ultrasound transmitted by the probe. The basic principle of MR elastography (MRE) is to convert the vibrational phase of shear elastic waves generated in the liver into the rotational phase of protons using a heating device that generates extracorporeal vibrations, and detect this phase difference using an MRI phase image. , the shear modulus is obtained. The velocity (V) of an elastic wave propagating within the liver is expressed by the formula V 2 =μ/ρ using the shear modulus (μ) and density (ρ) of the substance.
 一部の実施形態において、肝線維化をコラーゲン遺伝子の発現量を測定し評価することによっても、治療効果を確認することができる。 In some embodiments, the therapeutic effect can also be confirmed by evaluating liver fibrosis by measuring the expression level of collagen genes.
 一部の実施形態において、FIB-4 index(フィブフォー・インデックス)を測定し、肝臓の線維化の進展度合いを評価することができる。 In some embodiments, the FIB-4 index can be measured to evaluate the degree of progression of liver fibrosis.
 FIB-4 index(フィブフォー・インデックス)とは、肝臓の線維化の進展度合いを評価するための血液検査データを組み合わせたスコアリングシステムである。この検査方法は、血液検査AST値・ALT値・血小板数・年齢の4項目を組み合わせて計算し、得られた数値から線維化の進展の度合いを評価する。C型肝炎の肝線維化を予測するスコアである。最近ではNASHなどの他の肝疾患にも転用されており、脂肪肝の方においてFIB-4 indexを指標とし、肝線維化の進展例を早期に発見することが、NAFLD患者の経過観察において重要と考えられている。 The FIB-4 index is a scoring system that combines blood test data to evaluate the degree of progression of liver fibrosis. This test method calculates the degree of progression of fibrosis by combining four blood test items: AST value, ALT value, platelet count, and age. This is a score that predicts liver fibrosis in hepatitis C. Recently, it has been used for other liver diseases such as NASH, and it is important to use FIB-4 index as an indicator in fatty liver disease to detect advanced cases of liver fibrosis early in the follow-up of NAFLD patients. It is believed that.
 一部の実施形態において、APRIを測定し、肝臓の線維化の進展度合いを評価することができる。 In some embodiments, APRI can be measured to evaluate the degree of progression of liver fibrosis.
 APRI(アプリ=aspartate aminotransferase to platelet ratio index)は、AST、ALT、血小板数を組み合わせて線維化の度合を評価する。C型肝炎の肝線維化を予測するスコアである。 APRI (app = aspartate aminotransferase to platelet ratio index) evaluates the degree of fibrosis by combining AST, ALT, and platelet count. This is a score that predicts liver fibrosis in hepatitis C.
 一部の実施形態において、肝機能の「改善・維持」効果、即ち、「肝疾患に対する治療効果」は上記した各指標に基づいて評価することができる。一部の実施形態において、該効果を評価する際の補助として、上記した各指標を用いることができる。一部の実施形態において、各指標を測定することによって、治療に最適な用法用量を決定することができる。一部の実施形態において、評価指標として、肝組織硬度(例、フィブロスキャン測定値)、肝予備能指標(例、血清アルブミン濃度)、胆汁うっ滞指標(例、血清中総胆汁酸濃度)の少なくとも1種、2種、又は3種全てを用いることができる。 In some embodiments, the effect of "improving/maintaining" liver function, that is, the "therapeutic effect on liver disease" can be evaluated based on each of the above-mentioned indicators. In some embodiments, each of the indicators described above can be used to assist in evaluating the effectiveness. In some embodiments, by measuring each index, the optimal dosage regimen for treatment can be determined. In some embodiments, the evaluation indicators include liver tissue stiffness (e.g., fibroscan measurements), liver reserve indicators (e.g., serum albumin levels), and cholestatic indicators (e.g., serum total bile acid levels). At least one, two, or all three can be used.
 上記の評価指標は、通常、薬剤の投与を開始する前、薬剤の投与期間中及び薬剤の投与終了時に測定され、薬剤による肝機能の改善・維持効果(肝疾患に対する治療効果)を評価することができる。更に、薬剤の投与終了後一定期間(例えば、6乃至12か月)が経過した時点において測定することで、薬剤による肝機能の更なる改善や維持効果(肝疾患に対する治療効果)の持続性を評価することができる。 The above evaluation indicators are usually measured before starting drug administration, during the drug administration period, and at the end of drug administration, and are used to evaluate the effect of the drug on improving and maintaining liver function (therapeutic effect on liver disease). I can do it. Furthermore, by measuring after a certain period of time (e.g. 6 to 12 months) after the end of drug administration, it is possible to determine the durability of the drug's further improvement in liver function and maintenance effect (therapeutic effect on liver disease). can be evaluated.
 一部の実施形態において、本開示の医薬組成物は、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はそれらの医薬上許容され得る塩を含む。一部の実施形態において、本開示の医薬組成物は、有効成分として、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はそれらの医薬上許容され得る塩を含む。 In some embodiments, the pharmaceutical compositions of the present disclosure contain 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[( quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof . In some embodiments, the pharmaceutical compositions of the present disclosure include 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo- 8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or pharmaceutically acceptable thereof Contains salt obtained.
 本開示において、「医薬上許容され得る」とは、一般に安全で無毒性であり、そして生物学的にもそれ以外にも望ましくないものではない医薬組成物の調製に有用であることを意味し、且つヒトの医薬的用途のみならず獣医学的用途にも許容され得ることを含む。 In this disclosure, "pharmaceutically acceptable" means useful in preparing pharmaceutical compositions that are generally safe, non-toxic, and not biologically or otherwise undesirable. , and is acceptable for human as well as veterinary uses.
 本開示において、「医薬上許容され得る塩」とは、上記で定義したように、医薬上許容され、且つ所望の薬理学的活性を有する本発明化合物の塩を意味する。一部の実施形態において、このような塩としては、例えば、塩酸、臭化水素酸、硫酸、硝酸及びリン酸等の無機酸;又は例えば、酢酸、プロピオン酸、ヘキサン酸、ヘプタン酸、シクロペンタンプロピオン酸、グリコール酸、ピルビン酸、乳酸、マロン酸、コハク酸、リンゴ酸、マレイン酸、フマル酸、酒石酸、クエン酸、安息香酸、o-(4-ヒドロキシベンゾイル)安息香酸、桂皮酸、マンデル酸、メタンスルホン酸、エタンスルホン酸、1,2-エタンジスルホン酸、2-ヒドロキシエタンスルホン酸、ベンゼンスルホン酸、p-クロロベンゼンスルホン酸、2-ナフタレンスルホン酸、p-トルエンスルホン酸、カンファースルホン酸、4-メチルビシクロ[2.2.2]オクト-2-エン-1-カルボン酸、グルコヘプトン酸、4,4’-メチレンビス(3-ヒドロキシ-2-エン-1-カルボン酸)、3-フェニルプロピオン酸、トリメチル酢酸、tert-ブチル酢酸、ラウリル硫酸、グルコン酸、グルタミン酸、ヒドロキシナフトエ酸、サリチル酸、ステアリン酸及びムコン酸等の有機酸で形成された酸付加塩が挙げられる。一部の実施形態において、医薬上許容され得る塩としては、存在する酸性プロトンが無機又は有機の塩基と反応できる場合に形成され得る、塩基付加塩も挙げられる。一部の実施形態において、許容され得る無機塩基としては、水酸化ナトリウム、炭酸ナトリウム、水酸化カリウム、水酸化アルミニウム及び水酸化カルシウムが挙げられる。一部の実施形態において、許容され得る有機塩基としては、エタノールアミン、ジエタノールアミン、トリエタノールアミン、トロメタミン及びN-メチルグルカミン等が挙げられる。 In the present disclosure, the term "pharmaceutically acceptable salt" refers to a salt of the compound of the present invention that is pharmaceutically acceptable and has the desired pharmacological activity, as defined above. In some embodiments, such salts include inorganic acids, such as, for example, hydrochloric, hydrobromic, sulfuric, nitric, and phosphoric acids; or, for example, acetic, propionic, hexanoic, heptanoic, cyclopentane. Propionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid , methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-Methylbicyclo[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropion acid addition salts formed with organic acids such as trimethylacetic acid, tert-butylacetic acid, lauryl sulfate, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid and muconic acid. In some embodiments, pharmaceutically acceptable salts also include base addition salts that may be formed when acidic protons present are capable of reacting with an inorganic or organic base. In some embodiments, acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide, and calcium hydroxide. In some embodiments, acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
 一部の実施形態において、本開示の医薬組成物は、経口的に使用される剤形であってもよく、非経口的に使用される剤形であってもよい。これらの剤形は、当業者であれば、薬学的に許容される担体および添加剤を適宜組み合わせて、一般に認められた製薬実施に要求される単位用量形態で混和することによって製剤化することができる。一部の実施形態において、本開示の医薬組成物は、例えば日本薬局方または米国薬局方(USP)に記載された方法等、既知の方法に従って製造することができる。
 一部の実施形態において、本開示の医薬組成物の投与方法としては、静脈内投与、静脈内持続投与等が挙げられる。
In some embodiments, the pharmaceutical compositions of the present disclosure may be in a dosage form that is used orally or parenterally. These dosage forms can be formulated by those skilled in the art by incorporating appropriate combinations of pharmaceutically acceptable carriers and excipients into unit dosage forms as required by generally accepted pharmaceutical practice. can. In some embodiments, the pharmaceutical compositions of the present disclosure can be manufactured according to known methods, such as those described in the Japanese Pharmacopoeia or the United States Pharmacopeia (USP).
In some embodiments, methods of administering the pharmaceutical compositions of the present disclosure include intravenous administration, continuous intravenous administration, and the like.
 一部の実施形態において、本開示の医薬組成物は、常法により製剤化した医薬製剤(例、注射剤、点滴剤)として、ヒトに対して投与される。一部の実施形態において、前記投与は有効成分の量に換算して、100乃至400mg/mの用量である。 In some embodiments, the pharmaceutical composition of the present disclosure is administered to humans as a pharmaceutical preparation (eg, injection, drip) prepared by a conventional method. In some embodiments, the administration is at a dose of 100 to 400 mg/m 2 of active ingredient.
 一部の実施形態において、前記投与は有効成分の量に換算して、200乃至300mg/mの用量である。 In some embodiments, the administration is at a dose of 200 to 300 mg/m 2 of active ingredient.
 一部の実施形態において、前記投与は有効成分の量に換算して、200mg/m、210mg/m、220mg/m、230mg/m、240mg/m、250mg/m、260mg/m、270mg/m、280mg/m、290mg/m、300mg/mの用量で投与される。 In some embodiments, the administration is in an amount of active ingredient of 200 mg/m 2 , 210 mg/m 2 , 220 mg/m 2 , 230 mg/m 2 , 240 mg/m 2 , 250 mg/m 2 , 260 mg / m2 , 270mg/ m2 , 280mg/ m2 , 290mg/ m2 , 300mg/ m2 .
 一部の実施形態において、前記投与は有効成分の量に換算して、280mg/mの用量である。 In some embodiments, the administration is at a dose of 280 mg/m 2 of active ingredient.
 一部の実施形態において、前記投与は週1回以上、毎回1時間以上かけて静脈内に投与される。 In some embodiments, the administration is administered intravenously over one or more hours each time at least once a week.
 一部の実施形態において、前記投与は週1回又は2回、毎回2乃至5時間かけて静脈内に投与される。一部の実施形態において、前記投与は週2回、毎回4時間かけて静脈内に投与される。 In some embodiments, the administration is administered intravenously once or twice a week over a period of 2 to 5 hours each time. In some embodiments, the administration is administered intravenously twice a week over a 4 hour period each time.
 一部の実施形態において、前記投与が1か月にわたり継続されるように用いられる。 In some embodiments, the administration is continued for one month.
 一部の実施形態において、前記投与が2乃至6か月にわたり継続されるように用いられる。一部の実施形態において、前記投与が12週間にわたり継続されるように用いられる。 In some embodiments, the administration is continued for 2 to 6 months. In some embodiments, the administration is used to continue for 12 weeks.
 一部の実施形態において、本開示の医薬組成物は、常法により製剤化した医薬製剤(例、注射剤、点滴剤)として、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられる。一部の実施形態において、本開示の医薬組成物は、常法により製剤化した医薬製剤(例、注射剤、点滴剤)として、ヒトに対して週2回、毎回4時間かけて静脈内に投与され、前記投与が12週間にわたり継続されるように用いられる。 In some embodiments, the pharmaceutical composition of the present disclosure is administered to humans once or twice a week for 2 to 5 hours each time as a conventionally formulated pharmaceutical preparation (e.g., injection, infusion). The drug is administered intravenously over a period of time, and the administration is continued over a period of 2 to 6 months. In some embodiments, the pharmaceutical composition of the present disclosure is administered intravenously to humans twice a week for 4 hours each time as a pharmaceutical formulation (e.g., injection, infusion) formulated in a conventional manner. It is used such that the administration is continued for 12 weeks.
 以下に実施例を示して、本発明をより詳細に説明するが、これらは本発明の範囲を限定するものではない。 The present invention will be explained in more detail with reference to Examples below, but these are not intended to limit the scope of the present invention.
 すべての実験は、米国科学アカデミーの機関指針(Guide for the Care and Use of Laboratory Animals)に従って実施された。研究計画書は、東京都立駒込病院研究委員会の承認を得た。ヒト試料を用いたすべての分析は、東京都立駒込病院倫理委員会の承認を得た。この研究はヘルシンキ宣言の原則に準拠している。
 すべての実験動物(マウス)は、12時間/12時間の明暗サイクルのもと、換気されたケージ内で、エンリッチメント、水、飼料に自由にアクセスできるように維持された。
All experiments were performed in accordance with the National Academy of Sciences' Guide for the Care and Use of Laboratory Animals. The research protocol was approved by the Tokyo Metropolitan Komagome Hospital Research Committee. All analyzes using human samples were approved by the Tokyo Metropolitan Komagome Hospital Ethics Committee. This study complies with the principles of the Declaration of Helsinki.
All experimental animals (mice) were maintained in ventilated cages under a 12 h/12 h light/dark cycle with free access to enrichment, water, and food.
(略語一覧)
ALP: alkaline phosphatase(アルカリホスファターゼ)
ALT: alanine aminotransferase(アラニンアミノトランスフェラーゼ)
BA: bile acid(胆汁酸)
BDL: bile duct ligation(胆管結紮)
CA: cholic acid(コール酸)
CDCA: chenodeoxycholic acid(ケノデオキシコール酸)
CK19: cytokeratin-19(サイトケラチン19)
FXR: farnesoid X receptor(ファルネソイドX受容体)
MDR2: multidrug resistance-associated protein 2(多剤耐性関連タンパク質2)
PBC: primary biliary cholangitis(原発性胆汁性胆管炎)
PSC: primary sclerosing cholangitis(原発性硬化性胆管炎)
T.Bil: total bilirubin(総ビリルビン)
TBA: total bile acid(総胆汁酸)
(List of abbreviations)
ALP: alkaline phosphatase
ALT: alanine aminotransferase
BA: bile acid
BDL: bile duct ligation
CA: cholic acid
CDCA: chenodeoxycholic acid
CK19: cytokeratin-19 (cytokeratin 19)
FXR: farnesoid X receptor
MDR2: multidrug resistance-associated protein 2
PBC: primary biliary cholangitis
PSC: primary sclerosing cholangitis
T.Bil: total bilirubin
TBA: total bile acid
(材料と方法)
1.Mdr2-KOマウスモデル
 Mdr2-KO(FVB.129P2-Abcb4tm1Bor/J, #002539)マウスはJackson Laboratory(Bar Harbor, ME, USA)から購入し、FVB/NJ背景の野生型同腹子をコントロールとして使用した。8~10週齢の雄のMdr2-KOマウスと同腹のコントロールに、PBSに溶解した20mg/kgの本開示の化合物(foscenvivint;Prism BioLab、日本、東京)またはコントロールとしてのPBSを10週間にわたり週3回腹腔内注射した。
(Materials and methods)
1. Mdr2-KO Mouse Model Mdr2-KO (FVB.129P2-Abcb4tm1Bor/J, #002539) mice were purchased from Jackson Laboratory (Bar Harbor, ME, USA) and wild-type littermates on FVB/NJ background were used as controls. . 8-10 week old male Mdr2-KO mice and littermate controls were treated weekly for 10 weeks with 20 mg/kg of a compound of the present disclosure (foscenvivint; Prism BioLab, Tokyo, Japan) dissolved in PBS or PBS as a control. Three intraperitoneal injections were given.
2.BDLマウスモデル
 8~10週齢の雄の野生型(C57BL/6J)マウスを日本SLC(日本、静岡)から入手し、既報(Osawa Y, et al. Inhibition of Cyclic Adenosine Monophosphate (cAMP)-response Element-binding Protein (CREB)-binding Protein (CBP)/beta-Catenin Reduces Liver Fibrosis in Mice. eBioMedicine 2, 1751-1758 (2015).)と同様に膵臓上の総胆管を切開してBDLを施した。このマウスに20mg/kgの本開示の化合物をPBSに溶解したものを週3回腹腔内投与した。
2. BDL Mouse Model Male wild-type (C57BL/6J) mice aged 8 to 10 weeks were obtained from Japan SLC (Shizuoka, Japan) and used as previously reported (Osawa Y, et al. Inhibition of Cyclic Adenosine Monophosphate (cAMP)-response Element. -binding Protein (CREB)-binding Protein (CBP)/beta-Catenin Reduces Liver Fibrosis in Mice. eBioMedicine 2, 1751-1758 (2015).) The common bile duct above the pancreas was incised and BDL was performed. The mice received 20 mg/kg of a compound of the present disclosure dissolved in PBS intraperitoneally three times a week.
3.DDCマウスモデル
 8週齢のC57BL/6J野生型雄マウスに、0.1%DDC濃縮飼料(Sigma-Aldrich, St.Louis, MO, USA)を18日間与えた。
3. DDC Mouse Model Eight-week-old C57BL/6J wild-type male mice were fed 0.1% DDC concentrate diet (Sigma-Aldrich, St. Louis, MO, USA) for 18 days.
4.ビリルビン
 血清ビリルビンはQuantiChrom Bilirubin Assay Kit(BioAssay Systems, Hayward, CA, USA)を用いて測定した。
4. Bilirubin Serum bilirubin was measured using the QuantiChrom Bilirubin Assay Kit (BioAssay Systems, Hayward, CA, USA).
5.BA分析
 血清および肝臓のTBA、CA、CDCA濃度は、それぞれTotal Bile Acid Assay Kit、Cholic Acid ELISA Kit、Chenodeoxycholic Acid ELISA Kit(Cell Biolabs, San Diego, CA, USA)を用いて、メーカーの説明書に従って解析した。
5. BA analysis Serum and liver TBA, CA, and CDCA concentrations were determined using the Total Bile Acid Assay Kit, Cholic Acid ELISA Kit, and Chenodeoxycholic Acid ELISA Kit (Cell Biolabs, San Diego, CA, USA), respectively, according to the manufacturer's instructions. Analyzed.
6.RT-qPCR
 肝組織および培養細胞からのRNA抽出、DNA除去、逆転写には、RNeasy and DNase Kits(Qiagen, Valencia, CA, USA)及びHigh-Capacity cDNA Reverse Transcription Kit(Applied Biosystems, Foster City, CA, USA)が使用された。qPCRは、Thermo Fisher Scientificから購入したプローブとプライマーのセットとTaqPath qPCR Master Mix,CG(Applied Biosystems)を用いて、LightCycler 480(Roche Applied Science, Mannheim, Germany)により3連で実行された。標的遺伝子の発現量は、各サンプルにおけるGAPDHの発現量に対して正規化した。
(マウスプライマーセット)
6. RT-qPCR
For RNA extraction, DNA removal, and reverse transcription from liver tissue and cultured cells, RNeasy and DNase Kits (Qiagen, Valencia, CA, USA) and High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, USA) were used. was used. qPCR was performed in triplicate on a LightCycler 480 (Roche Applied Science, Mannheim, Germany) using probe and primer sets purchased from Thermo Fisher Scientific and TaqPath qPCR Master Mix, CG (Applied Biosystems). The expression level of the target gene was normalized to the expression level of GAPDH in each sample.
(mouse primer set)
7.組織学的解析
 マウス肝組織を10%ホルマリンで固定し、切片化し、ヘマトキシリン・エオシン(HE)で染色した。コラーゲン沈着はシリウスレッド(飽和ピクリン酸に0.1%direct red 80と0.1%Fast Green FCFを加えたもの)で染色した。さらに、サンプルを、サイトケラチン19(Abcam、ケンブリッジ、英国)に対する抗体を使って免疫組織化学的に染色した。シリウスレッド陽性領域を定量化するために、HistoQuantソフトウェア(3DHISTECH、ブダペスト、ハンガリー)を使用した。
(ヘマトキシリン・エオジン(HE)染色)
 HE染色は、細胞核及び細胞質を染め分け、組織の形態を観察することが可能な染色方法であり、HE染色により細胞や組織構造の全体像を把握することが可能となる。
 Bouin溶液で前固定した肝組織のパラフィンブロックから切片を切り出し、Lillie-Mayer’s Hematoxylin(武藤化学株式会社、日本)及びエオシン溶液(富士フイルム和光純薬工業株式会社、日本)で染色する。
(シリウスレッド染色)
 コラーゲンの沈着を可視化するために、Bouin固定された肝切片をピクロシリウスレッド溶液(Waldeck GmbH & Co., ドイツ)を用いて染色する。定量的解析には、デジタルカメラ(DFC280, Leica, ドイツ)を用いて、200倍拡大で、中心静脈付近でシリウスレッド染色された切片をキャプチャーして、1切片あたり5視野で陽性の領域をImageJ software(ImageJ, National Institute of Health, 米国)を用いて測定する。
7. Histological analysis Mouse liver tissues were fixed in 10% formalin, sectioned, and stained with hematoxylin and eosin (HE). Collagen deposits were stained with Sirius Red (saturated picric acid plus 0.1% Direct Red 80 and 0.1% Fast Green FCF). In addition, samples were immunohistochemically stained using an antibody against cytokeratin 19 (Abcam, Cambridge, UK). To quantify the Sirius Red positive area, HistoQuant software (3DHISTECH, Budapest, Hungary) was used.
(Hematoxylin and eosin (HE) staining)
HE staining is a staining method that allows the cell nucleus and cytoplasm to be dyed separately to observe the morphology of tissues. HE staining makes it possible to grasp the overall picture of cells and tissue structures.
Sections are cut from paraffin blocks of liver tissue prefixed with Bouin solution and stained with Lillie-Mayer's Hematoxylin (Muto Chemical Co., Ltd., Japan) and eosin solution (Fujifilm Wako Pure Chemical Industries, Ltd., Japan).
(Sirius red staining)
To visualize collagen deposition, Bouin-fixed liver sections are stained using picrosirius red solution (Waldeck GmbH & Co., Germany). For quantitative analysis, a digital camera (DFC280, Leica, Germany) was used to capture Sirius Red-stained sections near the central vein at 200x magnification, and positive areas were identified in 5 fields per section using ImageJ. Measure using software (ImageJ, National Institute of Health, USA).
8.Fib-4インデックス測定
 Fib-4インデックス(FIB-4 Index)は、肝線維化を予測するスコアであり、AST、ALT、血小板数、年齢の4項目を組み合わせて計算し、得られた数値から線維化の進展度合いを評価することができる。
8. Fib-4 Index Measurement The Fib-4 Index is a score that predicts liver fibrosis, and is calculated by combining four items: AST, ALT, platelet count, and age. It is possible to evaluate the degree of progress.
9.フィブロスキャン検査
 非侵襲的検査法としてパルス振動波の組織内伝搬速度を超音波画像解析法により弾性度(kPa)で測定するtransient elastographyを原理としたフィブロスキャンでの測定。ECHOSENS社製のFibroScan等が使用される。
9. Fibroscan test As a non-invasive testing method, Fibroscan is a non-invasive testing method based on the principle of transient elastography, which measures the propagation velocity of pulsed vibration waves in tissues in terms of elasticity (kPa) using ultrasonic image analysis. FibroScan manufactured by ECHOSENS is used.
10.血清アルブミン濃度
 血清アルブミンは、肝臓で作られるタンパク質で肝機能の低下とともにその量が減少することが知られている。
 血清アルブミンの測定は常法に従って行うことができる。例えば、アルブミンと結合することで色素を生成するような試薬を用いて、当該色素を分光学的に測定することによって検体中のアルブミン量を求める。このような色素としてはpH4.0付近でアルブミンと反応し青色のアルブミン結合色素を生成するブロモクレゾールグリーンが挙げられる。
 市販の測定用試薬(シカリキッド ALB、関東化学株式会社)を用いることもできる。検体に試薬を混合し、生成した結合色素の吸光度を測定する。
10. Serum Albumin Concentration Serum albumin is a protein produced in the liver, and its amount is known to decrease as liver function declines.
Serum albumin can be measured according to a conventional method. For example, the amount of albumin in a sample is determined by using a reagent that generates a pigment by binding with albumin and measuring the pigment spectroscopically. Examples of such dyes include bromocresol green, which reacts with albumin at around pH 4.0 to produce a blue albumin-binding dye.
A commercially available measurement reagent (Shikaliquid ALB, Kanto Kagaku Co., Ltd.) can also be used. Mix the reagent with the sample and measure the absorbance of the bound dye produced.
実施例1
前臨床試験(PBCのモデルマウスでの評価)1(肝線維化改善作用)
 原発性胆汁性胆管炎(PBC)の非臨床試験モデルとして、胆汁うっ滞により肝線維化を誘導する胆管結紮モデルマウス(BDLマウス)、MDR2ノックアウトマウス(Mdr2-KOマウス)及び胆管障害モデルマウス(DDCマウス)を用い、本開示の化合物の肝線維化改善作用を調べた。
 MDR2KOマウス及びBDLマウスはそれぞれ図1A及び図2Aのプロトコルに従って本開示の化合物で処置した。DDCマウスは、野生型C57BL/6雄性の8週齢マウスに0.1%DCC飼料を18日間与えた。その間、本開示の化合物(1週間あたり3回、20mg/kg)又はPBSを腹腔内投与した。処置後肝臓を採取し、肝組織のシリウスレッド染色により本開示の化合物の抗線維化効果を調べた。シリウスレッド陽性領域はコラーゲンの沈着を示す。BDLマウス及びDDCマウスについてはヘマトキシリン・エオシン染色も同時に行った。いずれのモデルマウスに対しても、シリウスレッド陽性領域の有意な減少、即ち線維化面積の減少を認め(図1~図3)、本開示の化合物の抗線維化効果が確認された。
Example 1
Preclinical test (evaluation in PBC model mice) 1 (hepatic fibrosis improving effect)
As nonclinical test models of primary biliary cholangitis (PBC), we have used bile duct ligation model mice (BDL mice), which induce liver fibrosis due to cholestasis, MDR2 knockout mice (Mdr2-KO mice), and bile duct injury model mice ( The hepatic fibrosis-improving effect of the compound of the present disclosure was investigated using DDC mice).
MDR2KO mice and BDL mice were treated with compounds of the present disclosure according to the protocols in Figures 1A and 2A, respectively. DDC mice were wild-type C57BL/6 male 8-week-old mice fed with 0.1% DCC feed for 18 days. During that time, compounds of the present disclosure (20 mg/kg three times per week) or PBS were administered intraperitoneally. After the treatment, the liver was collected and the anti-fibrotic effect of the compound of the present disclosure was examined by Sirius red staining of the liver tissue. Sirius red positive areas indicate collagen deposition. For BDL mice and DDC mice, hematoxylin and eosin staining was also performed at the same time. A significant decrease in the Sirius red positive area, ie, a decrease in the fibrotic area, was observed in all mouse model mice (FIGS. 1 to 3), confirming the anti-fibrotic effect of the compound of the present disclosure.
実施例2
前臨床試験(PBCのモデルマウスでの評価)2(胆汁うっ滞改善作用)
 実施例1同様、PBCモデルマウスとして、BDLマウス、Mdr2-KOマウス及びDDCマウスを用い、本開示の化合物の胆汁うっ滞改善作用を調べた。胆汁酸(BA)代謝が胆汁うっ滞による肝線維化に関与していることから、BA量(総胆汁酸(TBA)、コール酸(CA)、ケノデオキシコール酸(CDCA))を測定した。結果を図4(Mdr2-KOマウス)、図5(BDLマウス)及び図6(DDCマウス)に示す。Mdr2-KOマウスでは、肝臓のTBAとCAレベルは本開示の化合物の影響を受けなかったが、CDCAレベルは減少した。血清のTBA、CAおよびCDCAレベルは本開示の化合物によって減少した(図4)。同様に、本開示の化合物は、BDLマウスにおいても肝臓のTBA、CA、CDCAレベルを低下させ(図5)、DDCマウスにおいても肝臓及び血清のTBA、CA、CDCAレベルを低下させた(図6)。
Example 2
Preclinical test (evaluation in PBC model mice) 2 (cholestasis improving effect)
As in Example 1, the cholestasis-improving effect of the compound of the present disclosure was investigated using BDL mice, Mdr2-KO mice, and DDC mice as PBC model mice. Since bile acid (BA) metabolism is involved in liver fibrosis due to cholestasis, the amount of BA (total bile acids (TBA), cholic acid (CA), and chenodeoxycholic acid (CDCA)) was measured. The results are shown in FIG. 4 (Mdr2-KO mouse), FIG. 5 (BDL mouse), and FIG. 6 (DDC mouse). In Mdr2-KO mice, hepatic TBA and CA levels were unaffected by compounds of the present disclosure, whereas CDCA levels were decreased. Serum TBA, CA and CDCA levels were reduced by the compounds of the present disclosure (Figure 4). Similarly, compounds of the present disclosure also reduced liver TBA, CA, and CDCA levels in BDL mice (Figure 5), and reduced liver and serum TBA, CA, and CDCA levels in DDC mice (Figure 6). ).
実施例3
前臨床試験(PBCのモデルマウスでの評価)3(肝機能改善作用)
 実施例1同様、PBCモデルマウスとして、BDLマウス、Mdr2-KOマウス及びDDCマウスを用い、本開示の化合物の肝機能改善作用を調べた。血清アラニンアミノトランスフェラーゼ(ALT)は肝細胞でつくられる酵素で肝細胞が破壊されると血液中に放出されるため値が上昇する。アルカリホスファターゼ(ALP)はリン酸化合物を分解する酵素で、肝臓や腎臓、腸粘膜、骨などで作られ、肝臓で処理されて胆汁中に流れ出る。胆石や胆道炎、胆道がんなどで胆道がふさがれて胆汁の流れが悪くなったり(胆汁うっ滞)、肝臓の機能が低下したりすると、胆汁中のALPは逆流して血液中に流れ込む。従って、胆汁うっ滞ではALPは大きく上昇する。ビリルビンは、肝臓で処理される前の間接ビリルビンと処理されて胆汁中に入った直接ビリルビン(併せて総ビリルビン)があり、直接ビリルビンは胆道から排泄される。肝臓の機能が障害されると間接ビリルビンを処理できなくなるため、血液中に間接ビリルビンが大量に残り、一方、胆道系の障害により胆汁の排泄が不十分になると、血液中には直接ビリルビンが増加する。
 本実施例では、各PBCモデルマウスについて、血清中のALT、ALP及び総ビリルビンの量を測定した。結果を図7(Mdr2-KOマウス)、図8(BDLマウス)及び図9(DDCマウス)に示す。いずれのPBCモデルマウスにおいても、ALT、ALP、T.Bil値の低下で示されるように、肝障害の軽減が確認された。
Example 3
Preclinical test (evaluation in PBC model mice) 3 (hepatic function improvement effect)
As in Example 1, the liver function improving effect of the compound of the present disclosure was investigated using BDL mice, Mdr2-KO mice, and DDC mice as PBC model mice. Serum alanine aminotransferase (ALT) is an enzyme produced by liver cells, and when the liver cells are destroyed, it is released into the blood, resulting in increased levels. Alkaline phosphatase (ALP) is an enzyme that breaks down phosphoric acid compounds. It is produced in the liver, kidneys, intestinal mucosa, bones, etc., and is processed in the liver and released into bile. When the bile duct is blocked by gallstones, cholangitis, or biliary tract cancer, resulting in poor bile flow (cholestasis) or when liver function declines, ALP in bile flows backwards into the blood. Therefore, ALP is greatly elevated in cholestasis. Bilirubin includes indirect bilirubin before being processed by the liver, and direct bilirubin that is processed into bile (total bilirubin), and direct bilirubin is excreted from the biliary tract. When liver function is impaired, indirect bilirubin cannot be processed, so a large amount of indirect bilirubin remains in the blood.On the other hand, when biliary system dysfunction causes insufficient bile excretion, direct bilirubin increases in the blood. do.
In this example, the amounts of ALT, ALP, and total bilirubin in the serum of each PBC model mouse were measured. The results are shown in FIG. 7 (Mdr2-KO mouse), FIG. 8 (BDL mouse), and FIG. 9 (DDC mouse). In all of the PBC model mice, reduction in liver damage was confirmed as shown by reductions in ALT, ALP, and T.Bil values.
実施例4
前臨床試験(PBCのモデルマウスでの評価)4(コラーゲン遺伝子発現量改善作用)
 PBCモデルマウスとして、Mdr2-KOマウス及びDDCマウスを用い、本開示の化合物のコラーゲン遺伝子発現量改善作用を調べた。コラーゲン、特にIV型コラーゲンは、肝線維化のマーカーとして知られている。IV型コラーゲンは基底膜を構成する。正常の肝臓の類洞には基底膜は存在しないが、肝線維化進展に伴って基底膜の増生が起こり、IV型コラーゲンの発現量が増加する。
 本実施例では、各PBCモデルマウスについて、肝細胞におけるコラーゲン遺伝子の発現量を測定した。結果を図10(Mdr2-KOマウス)、図11(DDCマウス)に示す。いずれのPBCモデルマウスにおいても、コラーゲン遺伝子の発現量の低下で示されるように、肝線維化の軽減が確認された。
Example 4
Preclinical test (evaluation in PBC model mice) 4 (improving effect on collagen gene expression level)
Using Mdr2-KO mice and DDC mice as PBC model mice, the effect of the compound of the present disclosure on improving collagen gene expression was investigated. Collagen, particularly type IV collagen, is known as a marker of liver fibrosis. Type IV collagen constitutes the basement membrane. Although there is no basement membrane in normal liver sinusoids, proliferation of the basement membrane occurs as liver fibrosis progresses, and the expression level of type IV collagen increases.
In this example, the expression level of collagen gene in hepatocytes was measured for each PBC model mouse. The results are shown in FIG. 10 (Mdr2-KO mouse) and FIG. 11 (DDC mouse). In all of the PBC model mice, reduction in liver fibrosis was confirmed, as indicated by a decrease in collagen gene expression.
実施例5
臨床試験(PBC患者での評価)(胆汁うっ滞改善作用)
 本実施例は、PBC患者における本開示の化合物の安全性および忍容性を調べ、推奨用量を決定することを目的とした第I相試験の結果に基づく。
 被験者は、原発性胆道胆管炎と診断され、肝臓組織検査の結果、線維化の進行(Scheuer stage III以上)と診断された患者である。投薬スケジュールとして、本開示の化合物は、12週間にわたって週2回(4時間)静脈内投与された。ただし、第1サイクル投与の7日前1回、第1サイクルに予定されていた用量を4時間の連続静脈内投与により1回投与し、投与当日から投与翌日までの安全性及び薬物動態を評価した。用量レベルは、2用量(280mg/m/4時間、380mg/m/4時間)とした。
 本開示の化合物の12週間の投薬スケジュール終了後、血清総胆汁酸濃度(TBA濃度)を測定した。結果を図12に示す。
 投薬治療により明らかなTBA濃度の減少が見られ、本開示の化合物の胆汁うっ滞改善作用が確認された。
Example 5
Clinical trial (evaluation in PBC patients) (cholestasis improving effect)
This example is based on the results of a Phase I study designed to examine the safety and tolerability of the disclosed compounds and to determine recommended doses in PBC patients.
The subject was a patient who was diagnosed with primary biliary cholangitis and who was diagnosed with advanced fibrosis (Scheuer stage III or higher) as a result of liver histological examination. As a dosing schedule, compounds of the present disclosure were administered intravenously twice a week (4 hours) for 12 weeks. However, the dose scheduled for the first cycle was administered once 7 days before the first cycle administration by continuous intravenous administration for 4 hours, and safety and pharmacokinetics were evaluated from the day of administration to the day after administration. . Dose levels were 2 doses (280 mg/m 2 /4 hours, 380 mg/m 2 /4 hours).
Serum total bile acid concentrations (TBA concentrations) were measured after the completion of a 12-week dosing schedule of compounds of the present disclosure. The results are shown in FIG.
A clear decrease in TBA concentration was observed as a result of the drug treatment, confirming the cholestasis-improving effect of the compound of the present disclosure.
実施例6(肝炎患者での評価)(肝組織硬度改善作用)
 本実施例は、HCVまたはHBV肝硬変患者に本開示の化合物を投与した場合の安全性および薬物動態を評価し、本開示の化合物の推奨用量を決定することを目的とした第I相試験、及びHCVまたはHBV肝硬変患者に投与される本開示の化合物の推奨用量の有効性および安全性を評価することを目的とした第II相試験の結果に基づく。
 投薬スケジュールとしては、第I相試験では、(レベル1)140mg/m/4時間、(レベル2)280mg/m/4時間、(レベル3)380mg/m/4時間の3用量で、週2回、連続4時間静脈内投与(投与時間の許容範囲:±15分)で行った。これを単回投与と12サイクル(合計12週間)行った。単回投与は、第1サイクル投与の7日前1回、第1サイクルに予定されていた用量を連続4時間静脈内投与(投与時間の許容範囲:±15分)で行い、投与当日から投与翌日までの安全性及び薬物動態を評価した。
 第II(IIa)相試験では、第I相で決定した推奨用量(レベル2)で週2回4時間の連続静脈内投与を行った。これを12サイクル(合計12週間)行った(プロトコルA)。
 第I相試験において、本開示の化合物の12週間の投薬スケジュール終了後、肝組織硬度を、フィブロスキャンを用いて測定した。結果を図13に示す。
 投薬治療により明らかな肝組織硬度の減少が見られ、本開示の化合物の肝組織硬度改善作用が確認された。特に、レベル2での投薬スケジュールで良好な結果が得られた。
 第II相試験において、プロトコルAの投薬スケジュールで、血清アルブミン濃度を測定した。結果を図14Aに示す。本開示の化合物を投与することにより血清アルブミン濃度における改善作用が確認された。プロトコルAで本開示の化合物の投薬スケジュール終了後の血清アルブミン濃度を測定した。結果を図14Bに示す。本開示の化合物投薬開始9か月後も血清アルブミン濃度における改善作用が持続していることが確認された。
 第II相試験において、プロトコルAの投薬スケジュール終了後、FIB-4インデックスを調べた。結果を図15に示す。有意に低下傾向を認めた。
 プロトコルAで本開示の化合物の投薬スケジュール終了後の肝組織硬度についてフィブロスキャンを用いて測定した。結果を図16に示す。本開示の化合物投薬開始9か月後も肝組織硬度の改善効果が持続していることが確認された。
 第II相試験において、プロトコルAの投薬スケジュール終了6か月後のALBIスコアを調べた。結果を図17-1に示す。図17-2は、病状がChild-Pugh(CP)分類Bの症例を示した。本開示の化合物の投与終了6か月後において、ALBIスコアの改善よる肝機能改善が示唆された。第II相試験において、プロトコルAの投薬スケジュール終了6か月後において、血清アルブミン値を調べた。結果を図18-1に示す。図18-2は、病状がChild-Pugh(CP)分類Bの症例を示した。本開示の化合物を投与中にアルブミン製剤を使用した症例(KOM-009)、本開示の化合物を投与終了6か月後に腹水穿刺を実施した症例(KOM-021)、中等量以上の腹水を合併したために本開示の化合物を投与終了28日時点までにアルブミン製剤を使用した症例(KOM-033)が含まれている。肝機能の評価に影響を及ぼすような3例及び本開示の化合物を投与終了6か月後の結果が得られなかった2例(KOM-012、KOM-018)を除いた、CP分類Bの4例のうち2例が、投与終了6か月後においてCPスコア(血清アルブミン値の評点)が2点(2.8~3.5 g/dL)から1点(3.5 g/dL超)に改善がみられた。本開示の化合物の投与終了6か月後において、血清アルブミン値の改善よる肝機能改善が示唆された。
 第II相試験において、プロトコルAの投薬スケジュール終了6か月後において、プロトロンビン時間活性値を調べた。結果を図19―1に示す。図19-2は、病状がChild-Pugh(CP)分類Bの症例を示した。本開示の化合物を投与中にアルブミン製剤を使用した症例(KOM-009)、本開示の化合物を投与終了6か月後に腹水穿刺を実施した症例(KOM-021)、中等量以上の腹水を合併したために本開示の化合物を投与終了28日時点までにアルブミン製剤を使用した症例(KOM-033)が含まれている。肝機能の評価に影響を及ぼすような3例及び本開示の化合物を投与終了6か月後の結果が得られなかった2例(KOM-012、KOM-018)を除いた、CP分類Bの4例のうち3例が、投与終了6か月後においてCPスコア(プロトロンビン時間活性値の評点)が2点(40~70%)から1点(70%超)の改善がみられた。本開示の化合物の投与終了6か月後において、プロトロンビン時間活性値の改善より肝機能改善が示唆された。
Example 6 (evaluation in hepatitis patients) (improving effect on liver tissue hardness)
This example describes a phase I study aimed at evaluating the safety and pharmacokinetics of administering a compound of the present disclosure to patients with HCV or HBV cirrhosis and determining recommended doses of the compound of the present disclosure; Based on the results of a Phase II study aimed at evaluating the efficacy and safety of recommended doses of compounds of the present disclosure administered to patients with HCV or HBV cirrhosis.
As for the dosing schedule, in the phase I study, the drug was administered at three doses: (Level 1) 140 mg/m 2 /4 hours, (Level 2) 280 mg/m 2 /4 hours, and (Level 3) 380 mg/m 2 /4 hours. The drug was administered intravenously for 4 consecutive hours twice a week (acceptable range of administration time: ±15 minutes). This was done with a single dose and 12 cycles (total 12 weeks). For single administration, the dose scheduled for the first cycle is administered intravenously for 4 consecutive hours (tolerable range of administration time: ±15 minutes) once 7 days before the first cycle administration, and from the day of administration to the day after administration. The safety and pharmacokinetics were evaluated.
In the Phase II (IIa) study, continuous intravenous administration was performed for 4 hours twice a week at the recommended dose determined in Phase I (Level 2). This was done for 12 cycles (total 12 weeks) (Protocol A).
In a Phase I study, liver tissue stiffness was measured using Fibroscan after the completion of a 12-week dosing schedule of compounds of the present disclosure. The results are shown in FIG.
A clear decrease in liver tissue stiffness was observed with the drug treatment, confirming the liver tissue stiffness improving effect of the compound of the present disclosure. Particularly good results were obtained with the level 2 dosing schedule.
In a Phase II study, serum albumin concentrations were measured using the Protocol A dosing schedule. The results are shown in Figure 14A. An improving effect on serum albumin concentration was confirmed by administering the compound of the present disclosure. Serum albumin concentrations were determined following the dosing schedule of compounds of the present disclosure in Protocol A. The results are shown in Figure 14B. It was confirmed that the improving effect on serum albumin concentration continued even 9 months after the start of administration of the compound of the present disclosure.
In a Phase II study, the FIB-4 index was examined after completion of the Protocol A dosing schedule. The results are shown in FIG. A significant decreasing trend was observed.
Liver tissue stiffness was measured using Fibroscan after completing the dosing schedule of the compounds of the present disclosure in Protocol A. The results are shown in FIG. It was confirmed that the effect of improving liver tissue hardness continued even 9 months after the start of administration of the compound of the present disclosure.
In a Phase II study, ALBI scores were examined 6 months after the end of the Protocol A dosing schedule. The results are shown in Figure 17-1. Figure 17-2 shows a case with Child-Pugh (CP) classification B. Six months after the end of administration of the compound of the present disclosure, improvement in liver function was suggested by improvement in ALBI score. In the Phase II study, serum albumin levels were determined 6 months after the end of the Protocol A dosing schedule. The results are shown in Figure 18-1. Figure 18-2 shows a case with Child-Pugh (CP) classification B. A case in which an albumin preparation was used during administration of the compound of the present disclosure (KOM-009), a case in which ascites puncture was performed 6 months after the completion of administration of the compound of the present disclosure (KOM-021), and a case with moderate or higher amount of ascites. This includes a case (KOM-033) in which an albumin preparation was used up to 28 days after the end of administration of the compound of the present disclosure. CP class B, excluding 3 cases where the evaluation of liver function was affected and 2 cases where results were not obtained 6 months after the end of administration of the compound of the present disclosure (KOM-012, KOM-018). Two of the four patients showed an improvement in their CP score (serum albumin level rating) from 2 points (2.8 to 3.5 g/dL) to 1 point (over 3.5 g/dL) 6 months after the end of treatment. Ta. Six months after the end of administration of the compound of the present disclosure, improvement in liver function was suggested due to improvement in serum albumin levels.
In a Phase II study, prothrombin time activity values were determined 6 months after the end of the Protocol A dosing schedule. The results are shown in Figure 19-1. Figure 19-2 shows a case with Child-Pugh (CP) classification B. A case in which an albumin preparation was used during administration of the compound of the present disclosure (KOM-009), a case in which ascites puncture was performed 6 months after the completion of administration of the compound of the present disclosure (KOM-021), and a case with moderate or higher amount of ascites. This includes a case (KOM-033) in which an albumin preparation was used up to 28 days after the end of administration of the compound of the present disclosure. CP class B, excluding 3 cases where the evaluation of liver function was affected and 2 cases where results were not obtained 6 months after the end of administration of the compound of the present disclosure (KOM-012, KOM-018). Three of the four patients showed an improvement in their CP score (prothrombin time activity score) from 2 points (40-70%) to 1 point (over 70%) 6 months after the end of treatment. Six months after the end of administration of the compound of the present disclosure, improvement in the prothrombin time activity value suggested improvement in liver function.
実施例7(血友病HIV/HCV重複感染に起因する肝硬変患者での評価)
 本実施例は、血友病HIV/HCV重複感染に起因する肝硬変患者を対象とし、本開示の化合物を投与したときの安全性および忍容性を検討することを目的とする第I相試験の結果に基づく。
 投薬スケジュールとしては、(レベル1)140mg/m2/4時間、(レベル2)280mg/m2/4時間の2用量で、週2回、連続4時間静脈内投与(投与時間の許容範囲:±15分)で行った。これを単回投与と12サイクル(合計12週間)行った。単回投与は、第1サイクル投与の14日前1回、第1サイクルに予定されていた用量を連続4時間静脈内投与(投与時間の許容範囲:±15分)で行い、投与当日から投与翌日までの安全性及び薬物動態を評価した。 
 本開示の化合物の12週間の投薬スケジュール終了後、フィブロスキャンを用いて肝組織硬度の測定、及びFIB-4 indexとAPRIの算出を行った。結果をそれぞれ図20、図21A、図21Bに示す。フィブロスキャンによる肝臓組織の硬さの測定値はFIB-4 index及びAPRIにてレベル1とレベル2のいずれもべースラインからの低下がみられ、なかでもフィブロスキャンによる肝臓組織の硬さの測定値及びAPRIではレベル2でより低下がみられた。本開示の化合物を投与した後、フィブロスキャンでの肝硬度低下、FIB-4 index及びAPRIスコアの減少傾向が確認された。
Example 7 (Evaluation in patients with hemophilia and cirrhosis caused by HIV/HCV co-infection)
This example is a phase I study aimed at examining the safety and tolerability of administering the compound of the present disclosure to patients with hemophilia and liver cirrhosis caused by HIV/HCV co-infection. Based on results.
The dosing schedule was two doses: (Level 1) 140 mg/m2/4 hours and (Level 2) 280 mg/m2/4 hours, twice a week, for 4 consecutive hours (administration time tolerance range: ±15 minutes). This was done with a single dose and 12 cycles (total 12 weeks). For single administration, the dose scheduled for the first cycle is administered intravenously for 4 consecutive hours (tolerable range of administration time: ±15 minutes) once 14 days before the first cycle administration, and from the day of administration to the day after administration. The safety and pharmacokinetics were evaluated.
After completing the 12-week dosing schedule of the compounds of the present disclosure, liver tissue stiffness was measured using Fibroscan and FIB-4 index and APRI were calculated. The results are shown in FIG. 20, FIG. 21A, and FIG. 21B, respectively. The liver tissue stiffness measured by Fibroscan showed a decrease from the baseline in both Level 1 and Level 2 of the FIB-4 index and APRI. and APRI showed a greater decline at level 2. After administering the compound of the present disclosure, a decrease in liver stiffness as determined by Fibroscan, and a decreasing trend in FIB-4 index and APRI score was confirmed.
実施例8(非代償性肝硬変患者での評価)
 本実施例は、HCVまたはHBV感染に起因する非代償性肝硬変患者、及び非アルコール性脂肪肝炎(疑いを含む)に起因する非代償性肝硬変患者に本開示の化合物を投与した場合の有効性、安全性および薬物動態を評価することを目的とした第II相試験の計画に基づくものである。非代償性肝硬変の重症度は、Child-Pugh分類Bであり、HCVまたはHBV感染に起因する非代償性肝硬変患者をコホートA、非アルコール性脂肪肝炎(疑いを含む)に起因する非代償性肝硬変患者をコホートBに分けて、当該化合物又はプラセボを二重盲検下で投与する。
 投与スケジュールとしては、280mg/m2の用量で、1回あたり連続3~4時間静脈内投与(投与時間の許容範囲:±15分)で行う。コホートAでは3群(当該化合物を週2回投与する群、当該化合物とプラセボを週1回ずつ投与する群、プラセボを週2回投与する群)、コホートBでは2群(当該化合物を週2回投与する群、プラセボを週2回投与する群)に無作為に割付けて、24週間投与を行い、投与開始52週間まで観察を行う。
 有効性評価項目として、肝予備能指標(Child-Pughスコア、ALBIスコア、MELDスコア、Child-Pugh分類、mALBIグレード、血清アルブミン値、血清ビリルビン値、プロトロンビン活性値)、肝線維化指標(フィブロスキャン肝組織硬度、MRE肝組織硬度、血清線維化マーカー、FIB-4 index)を評価する。その他、腹水の治療で使用されるアルブミン製剤の投与量及び投与回数、非代償性肝硬変に伴うイベント(食道・胃静脈瘤出血、腹水など)、腹水及び肝性脳症の重症度についても評価する。
Example 8 (Evaluation in patients with decompensated liver cirrhosis)
This example demonstrates the efficacy of compounds of the present disclosure when administered to patients with decompensated cirrhosis due to HCV or HBV infection, and patients with decompensated cirrhosis due to (suspected) non-alcoholic steatohepatitis; It is based on the design of a Phase II study aimed at evaluating safety and pharmacokinetics. The severity of decompensated cirrhosis is Child-Pugh classification B, and patients with decompensated cirrhosis due to HCV or HBV infection are classified into Cohort A, and patients with decompensated cirrhosis due to non-alcoholic steatohepatitis (including suspected). Patients will be divided into Cohort B and will receive the compound or placebo in a double-blind manner.
The administration schedule is 280 mg/m2, administered intravenously for 3 to 4 consecutive hours each time (acceptable range of administration time: ±15 minutes). Cohort A has 3 groups (the compound is administered twice a week, the compound and placebo are administered once a week, and the placebo is administered twice a week), and Cohort B has 2 groups (the compound is administered twice a week). Patients were randomly assigned to a group receiving twice a week administration of a placebo and a group receiving a placebo twice a week, and were administered for 24 weeks, with observation being made until 52 weeks after the start of administration.
Efficacy evaluation items include liver reserve index (Child-Pugh score, ALBI score, MELD score, Child-Pugh classification, mALBI grade, serum albumin level, serum bilirubin level, prothrombin activity value), liver fibrosis index (fibroscan Evaluate liver tissue stiffness, MRE liver tissue stiffness, serum fibrosis marker, FIB-4 index). In addition, the dose and frequency of administration of albumin preparations used to treat ascites, events associated with decompensated cirrhosis (esophageal/gastric variceal bleeding, ascites, etc.), and the severity of ascites and hepatic encephalopathy will also be evaluated.
 4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェートは、100乃至400mg/mの用量で、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられることにより、優れた肝硬変(肝線維化)治療効果を示すことが示唆される。従って、該化合物を含有する医薬組成物は、肝疾患予防及び/又は治療に有効であることが示唆される。 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2 ,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate is administered to humans at a dose of 100 to 400 mg/m 2 once or twice a week, each time 2 to 5 It is suggested that when administered intravenously over time and the administration is continued for 2 to 6 months, it exhibits an excellent therapeutic effect on liver cirrhosis (hepatic fibrosis). Therefore, it is suggested that a pharmaceutical composition containing the compound is effective for preventing and/or treating liver diseases.

Claims (25)

  1.  100乃至400mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週1回または2回、毎回2乃至5時間かけて静脈内に投与され、前記投与が2乃至6か月にわたり継続されるように用いられることを特徴とする、4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩を含有する、肝疾患の予防及び/又は治療用医薬組成物。 100 to 400 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl] Octahydro-2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered to humans once or twice a week. 4-({(6S,9S,9aS)-1 -(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2,1-c][1,2,4]triazine -6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, for the prevention and/or treatment of liver diseases.
  2.  280mg/mの4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩が、ヒトに対して週2回、毎回4時間かけて静脈内に投与され、前記投与が12週間にわたり継続されるように用いられることを特徴とする、請求項1記載の医薬組成物。 280 mg/m 2 of 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro- 2H-pyrazino[2,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, is administered to humans twice a week for 4 hours each time. 2. The pharmaceutical composition according to claim 1, wherein the pharmaceutical composition is administered intravenously over a period of 12 weeks.
  3.  肝疾患が、肝線維化又は肝硬変を伴う、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the liver disease is accompanied by liver fibrosis or cirrhosis.
  4.  肝線維化又は肝硬変がB型若しくはC型肝炎または原発性胆汁性胆管炎(PBC)に起因する疾患であることを特徴とする請求項3記載の医薬組成物。 The pharmaceutical composition according to claim 3, wherein the liver fibrosis or cirrhosis is a disease caused by hepatitis B or C or primary biliary cholangitis (PBC).
  5.  肝線維化又は肝硬変が血友病HIV/HCV重複感染、又は非アルコール性脂肪性肝炎に起因する疾患であることを特徴とする請求項3記載の医薬組成物。 The pharmaceutical composition according to claim 3, wherein the liver fibrosis or cirrhosis is a disease caused by hemophilia, HIV/HCV co-infection, or non-alcoholic steatohepatitis.
  6.  肝疾患が、Child-Pughスコアによる分類A、BまたはCのものである、請求項3記載の医薬組成物。 The pharmaceutical composition according to claim 3, wherein the liver disease is classified as A, B, or C according to the Child-Pugh score.
  7.  肝組織硬度の維持又は改善用である、請求項1~6のいずれか1項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 6, which is used for maintaining or improving liver tissue hardness.
  8.  維持又は改善が、薬剤投与中または薬剤投与終了時点において、薬剤投与開始前と比較して評価されるものである、請求項7記載の医薬組成物。 8. The pharmaceutical composition according to claim 7, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the drug administration started.
  9.  維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、請求項7記載の医薬組成物。 8. The pharmaceutical composition according to claim 7, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
  10.  肝組織硬度がフィブロスキャン測定値で示される、請求項7記載の医薬組成物。 8. The pharmaceutical composition according to claim 7, wherein liver tissue hardness is indicated by Fibroscan measurement value.
  11.  肝組織硬度がMRエラストグラフィ測定値で示される、請求項7記載の医薬組成物。 The pharmaceutical composition according to claim 7, wherein liver tissue hardness is indicated by MR elastography measurements.
  12.  肝予備能指標の維持又は改善用である、請求項1~6のいずれか1項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 6, which is used for maintaining or improving a liver reserve index.
  13.  維持又は改善が、薬剤投与中または薬剤投与終了時点において、薬剤投与開始前と比較して評価されるものである、請求項12記載の医薬組成物。 13. The pharmaceutical composition according to claim 12, wherein maintenance or improvement is evaluated during drug administration or at the end of drug administration compared to before the drug administration started.
  14.  維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、請求項12記載の医薬組成物。 13. The pharmaceutical composition according to claim 12, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
  15.  肝予備能指標が血清アルブミン濃度で示される、請求項12記載の医薬組成物。 13. The pharmaceutical composition according to claim 12, wherein the hepatic reserve index is represented by serum albumin concentration.
  16.  肝予備能指標が血清ビリルビン濃度、プロトロンビン活性値、又はALBIスコアで示される、請求項12記載の医薬組成物。 The pharmaceutical composition according to claim 12, wherein the hepatic reserve index is represented by serum bilirubin concentration, prothrombin activity value, or ALBI score.
  17. [17]肝予備能指標が、Child Pugh分類(A,B,C)の変化またはスコア(点数)の変化量で示される、請求項12記載の医薬組成物 [17] The pharmaceutical composition according to claim 12, wherein the hepatic reserve index is indicated by a change in Child Pugh classification (A, B, C) or a change in score.
  18.  胆汁うっ滞指標の維持又は改善用である、請求項1~6のいずれか1項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 6, which is used for maintaining or improving a cholestasis index.
  19.  維持又は改善が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して評価されるものである、請求項18記載の医薬組成物。 19. The pharmaceutical composition according to claim 18, wherein maintenance or improvement is evaluated 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
  20.  胆汁うっ滞指標が血清中総胆汁酸濃度で示される、請求項18記載の医薬組成物。 The pharmaceutical composition according to claim 18, wherein the cholestasis index is expressed by serum total bile acid concentration.
  21.  4-({(6S,9S,9aS)-1-(ベンジルカルバモイル)-2,9-ジメチル-4,7-ジオキソ-8-[(キノリン-8-イル)メチル]オクタヒドロ-2H-ピラジノ[2,1-c][1,2,4]トリアジン-6-イル}メチル)フェニル ジヒドロゲン ホスフェート、又はその医薬上許容され得る塩の肝疾患に対する治療効果の評価を補助する方法であって、肝組織硬度、肝予備能指標及び胆汁うっ滞指標から選択される少なくとも1つを測定することを特徴とする方法。 4-({(6S,9S,9aS)-1-(benzylcarbamoyl)-2,9-dimethyl-4,7-dioxo-8-[(quinolin-8-yl)methyl]octahydro-2H-pyrazino[2 ,1-c][1,2,4]triazin-6-yl}methyl)phenyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof, for assisting in the evaluation of the therapeutic effect on liver diseases, the method comprising: A method characterized by measuring at least one selected from hardness, liver reserve index, and cholestasis index.
  22.  該測定が、薬剤投与終了後6乃至12か月の時点において、薬剤投与終了時点と比較して為されるものである、請求項21記載の方法。 22. The method according to claim 21, wherein the measurement is performed 6 to 12 months after the end of drug administration compared to the time point at which drug administration ended.
  23.  肝組織硬度がフィブロスキャン測定値で示される、請求項21又は22記載の方法。 23. The method according to claim 21 or 22, wherein the liver tissue stiffness is indicated by a fibroscan measurement value.
  24.  肝予備能指標が血清アルブミン濃度で示される、請求項21又は22記載の方法。 23. The method according to claim 21 or 22, wherein the hepatic reserve index is indicated by serum albumin concentration.
  25.  胆汁うっ滞指標が血清中総胆汁酸濃度で示される、請求項21又は22記載の方法。 23. The method according to claim 21 or 22, wherein the cholestasis index is indicated by serum total bile acid concentration.
PCT/JP2023/016556 2022-04-28 2023-04-26 Liver fibrosis therapeutic regimen for cbp/catenin inhibitor WO2023210716A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2022-075421 2022-04-28
JP2022075421 2022-04-28

Publications (1)

Publication Number Publication Date
WO2023210716A1 true WO2023210716A1 (en) 2023-11-02

Family

ID=88518760

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2023/016556 WO2023210716A1 (en) 2022-04-28 2023-04-26 Liver fibrosis therapeutic regimen for cbp/catenin inhibitor

Country Status (1)

Country Link
WO (1) WO2023210716A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020122022A1 (en) * 2018-12-10 2020-06-18 東京都 Liver function improving agent

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020122022A1 (en) * 2018-12-10 2020-06-18 東京都 Liver function improving agent

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "History of Changes for Study: NCT04047160", 27 December 2021 (2021-12-27), XP093105564, Retrieved from the Internet <URL:https://classic.clinicaltrials.gov/ct2/history/NCT04047160?A=7&B=7&C=merged#StudyPageTop> *
ANONYMOUS: "History of Changes for Study: NCT04688034", 16 March 2022 (2022-03-16), XP093105563, Retrieved from the Internet <URL:https://classic.clinicaltrials.gov/ct2/history/NCT04688034?A=6&B=6&C=merged#StudyPageTop> *
KIMURA KIMINORI, IKOMA AKEMI, SHIBAKAWA MAKI, SHIMODA SHINJI, HARADA KENICHI, SAIO MASANAO, IMAMURA JUN, OSAWA YOSUKE, KIMURA MASA: "Safety, Tolerability, and Preliminary Efficacy of the Anti-Fibrotic Small Molecule PRI-724, a CBP/β-Catenin Inhibitor, in Patients with Hepatitis C Virus-related Cirrhosis: A Single-Center, Open-Label, Dose Escalation Phase 1 Trial", EBIOMEDICINE, ELSEVIER BV, NL, vol. 23, 1 September 2017 (2017-09-01), NL , pages 79 - 87, XP093105576, ISSN: 2352-3964, DOI: 10.1016/j.ebiom.2017.08.016 *
KIMURA KIMINORI, KANTO TATSUYA, SHIMODA SHINJI, HARADA KENICHI, KIMURA MASAMICHI, NISHIKAWA KOJI, IMAMURA JUN, OGAWA EIICHI, SAIO : "Safety, tolerability, and anti-fibrotic efficacy of the CBP/β-catenin inhibitor PRI-724 in patients with hepatitis C and B virus-induced liver cirrhosis: An investigator-initiated, open-label, non-randomised, multicentre, phase 1/2a study", EBIOMEDICINE, ELSEVIER BV, NL, vol. 80, 20 May 2022 (2022-05-20), NL , pages 104069, XP093105575, ISSN: 2352-3964, DOI: 10.1016/j.ebiom.2022.104069 *
TANAKA NAOMI, OSUGA TOSHIAKI, TORII MASAO, ODA TOSHITSUGU, MASHIGE FUMIKO: "Clinical significance of determination of serum bile acid in liver diseases", KANZO, vol. 22, no. 6, 1 January 1981 (1981-01-01), pages 785 - 802, XP093105572, DOI: 10.2957/kanzo.22.785 *

Similar Documents

Publication Publication Date Title
US20110021475A1 (en) Compositions And Methods for Treatment Of Fibrosis
Paumgartner Biliary physiology and disease: Reflections of a physician‐scientist
WO2017170434A1 (en) Medicine obtained by combining fxr agonist and arb
KR102067848B1 (en) Methods of treating fibrosis
KR20110107287A (en) Pharmaceutical composition for the prevention and the treatment of non-alcoholic fatty liver disease
JP2019505559A (en) Methods for improving liver regeneration
Smith et al. Endogenous ursodeoxycholic acid and cholic acid in liver disease due to cystic fibrosis
US20220288053A1 (en) Compositions and methods to treat non-alcoholic fatty liver diseases (nafld)
JP6915050B2 (en) New regime of FXR agonists
EA014077B1 (en) Use of 24-nor-ursodeoxycholic acid
Kim et al. Cigarette smoking differentially regulates inflammatory responses in a mouse model of nonalcoholic steatohepatitis depending on exposure time point
CN114144185A (en) Pharmaceutical composition comprising an FXR agonist and a fibrate for the treatment of cholestatic liver diseases
Ma et al. Activation of G0/G1 switch gene 2 by endoplasmic reticulum stress enhances hepatic steatosis
WO2023210716A1 (en) Liver fibrosis therapeutic regimen for cbp/catenin inhibitor
RU2752089C1 (en) Application of the compound for the preparation of a drug for the treatment of cerebral microangiopathy
Navas‐Madroñal et al. Targeting mitochondrial stress with Szeto‐Schiller 31 prevents experimental abdominal aortic aneurysm: Crosstalk with endoplasmic reticulum stress
Wang et al. Rifampicin induces bicarbonate-rich choleresis in rats: Involvement of anion exchanger 2
CA2688079C (en) 5beta, 14beta-androstane derivatives useful for the treatment of restenosis after angioplastic or endoartherectomy and diseases due to organ fibrosis
US20230181602A1 (en) Treatment of non-alcoholic steatohepatitis (nash)
US20230181601A1 (en) Treatment of non-alcoholic steatohepatitis (nash)
Zhuang et al. Gpbar-1/cAMP/PKA signaling mitigates macrophage-mediated acute cholestatic liver injury via antagonizing NLRP3-ASC inflammasome
WO2021237149A2 (en) Treatment of non-alcoholic steatohepatitis (nash)
WO2023147141A1 (en) Combination therapy
Asgarova et al. CHANGES OF INSULIN IN THE BLOOD OF RABBITS UNDER CONDITIONS OF HYPOXIA
Gu et al. Mirna Database

Legal Events

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

Ref document number: 23796452

Country of ref document: EP

Kind code of ref document: A1