[go: up one dir, main page]

WO2025127097A1 - Medicine for treating steatotic liver diseases - Google Patents

Medicine for treating steatotic liver diseases Download PDF

Info

Publication number
WO2025127097A1
WO2025127097A1 PCT/JP2024/043962 JP2024043962W WO2025127097A1 WO 2025127097 A1 WO2025127097 A1 WO 2025127097A1 JP 2024043962 W JP2024043962 W JP 2024043962W WO 2025127097 A1 WO2025127097 A1 WO 2025127097A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
fatty liver
liver disease
formula
composition according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/JP2024/043962
Other languages
French (fr)
Japanese (ja)
Inventor
慶介 楠本
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shionogi and Co Ltd
Original Assignee
Shionogi and Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shionogi and Co Ltd filed Critical Shionogi and Co Ltd
Publication of WO2025127097A1 publication Critical patent/WO2025127097A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/45Non condensed piperidines, e.g. piperocaine having oxo groups directly attached to the heterocyclic ring, e.g. cycloheximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a new pharmaceutical composition for treating and/or preventing fatty liver disease, particularly non-alcoholic fatty liver disease.
  • Liver disease is a leading cause of death worldwide. Liver disease can be caused by infection, injury, exposure to drugs or toxic compounds, alcohol, impurities in food, and abnormal accumulation of normal substances in the blood, autoimmune processes, genetic defects (such as hemochromatosis), or unknown cause(s). Liver disease is commonly classified as acute or chronic based on the duration of the disease. Fatty liver disease (SLD) is a general term for diseases in which accumulation of neutral fat in the liver causes liver damage. A case in which 30% or more of hepatocytes in liver tissue contain lipid droplets is called fatty liver.
  • SLD Fatty liver disease
  • Fatty liver is a state where neutral fat accumulates in the liver, and since the accumulation of fat in the liver is a non-progressive (reversible) change, the accumulation of fat in the liver returns to normal when the cause is removed. However, if the accumulation of fat progresses and liver dysfunction occurs, it may develop into cirrhosis or liver cancer.
  • Alcoholic steatohepatitis which is caused by drinking alcohol, is a typical example of fatty liver.
  • fatty liver can occur even in people who consume very little alcohol, and this condition is called nonalcoholic fatty liver disease (NAFLD).
  • NAFLD nonalcoholic fatty liver disease
  • NAFLD nonalcoholic steatohepatitis
  • NASH is considered to be an important cause of cirrhosis and liver cancer.
  • the histological findings of alcoholic and non-alcoholic hepatitis are similar, and common mechanisms of onset, such as oxidative stress in the liver, are being studied.
  • NAFLD can progress to NASH, which causes serious adverse effects.
  • NASH can lead to liver fibrosis, cirrhosis, liver failure, or hepatocellular carcinoma.
  • An estimated 16 million adults in the United States have NASH, and roughly 50% have advanced liver fibrosis (bridging fibrosis or cirrhosis) associated with NASH.
  • NASH is currently the leading cause of hepatocellular carcinoma in the United States and the second most common indication for liver transplantation after hepatitis C. NASH is characterized by the presence of steatosis, as well as other characteristics including hepatocellular degeneration (balloon, Mallory hyaline), inflammatory cell infiltration, and the development of progressive fibrosis. There are no currently approved therapies for the treatment of NASH or that reduce fibrosis and/or steatosis in NASH patients. Thus, there remains a need to provide new effective agents for treating liver disease or symptoms of liver disease.
  • Patent Document 1 describes a method for treating and/or preventing NASH and/or primary biliary cirrhosis, which comprises administering a pharmaceutical composition containing eicosapentaenoic acid or a derivative thereof to a subject in need thereof. It has been reported that obeticholic acid, a semisynthetic bile acid and an agonist for the nuclear receptor FXR (farnesoid X receptor), has been applied for in Europe and the United States for the treatment of fibrosis caused by NASH.
  • FXR nuclear receptor
  • Non-Patent Document 1 In monoacylglycerol acyltransferase 2 (hereinafter also referred to as MGAT2) knockout mice, it has been confirmed that high-fat diet-induced weight gain, insulin resistance, blood cholesterol elevation, fatty liver formation, and energy consumption are suppressed (Non-Patent Document 1). In addition, it has been reported that compounds having MGAT2 inhibitory activity (JTP-103237, BMS-963272 ) improve fatty liver in high sucrose-loaded model mice and improve liver fibrosis in NASH model mice (Non-Patent Documents 2 and 3). Furthermore, it has been reported that expression of MGAT2 is increased in the liver of NAFLD patients (Non-Patent Document 4). Based on these findings, it is expected that MGAT2 inhibitors are promising therapeutic agents for fatty liver diseases including NASH, but no drug that shows efficacy in humans has yet been developed (Non-Patent Document 5).
  • Patent documents 2 to 4 disclose MGAT2 inhibitors of the formula: The compound represented by the formula: Furthermore, Patent Documents 5 and 6 disclose MGAT2 inhibitors of the formula: The compound represented by the formula: However, Patent Documents 2 to 6 do not disclose that they are effective in treating and/or preventing non-alcoholic fatty liver disease.
  • the object of the present invention is to provide a pharmaceutical composition for the treatment and/or prevention of fatty liver disease, particularly non-alcoholic fatty liver disease, which has excellent MGAT2 inhibitory activity and is free of side effects such as hemostatic abnormalities, liver damage, renal damage, or embryonic lethality.
  • the present inventors discovered that, among the compounds having MGAT2 inhibitory activity described in Patent Documents 2 to 4, certain compounds are effective in treating and/or preventing non-alcoholic fatty liver disease and are free of side effects such as hemostatic abnormalities, liver damage, renal damage, or embryonic lethality, and thus completed the present invention.
  • the present invention relates to the following.
  • Each R 6 is independently halogen, haloalkyl, or haloalkyloxy;
  • R 4a is a group represented by the formula: and
  • R 4b is a haloalkyloxyalkyl, a phenyl group optionally substituted with halogen, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group ⁇ ,
  • Ring B is represented by the formula: (In the formula, each symbol has the same meaning as [1] or [2].)
  • the pharmaceutical composition according to [1] or [2], [4] The pharmaceutical composition according to [1], which contains a compound selected from the group consisting of compounds I-8, I-23, I-34, I-190, I-212, I-236, I-253, I-275, I-276, II-93, II-103, II-121, II-151, II-168, II-174, II-203, II-225, II-233 and II-295, or a pharma- ceutical acceptable salt thereof.
  • the pharmaceutical composition according to [1] which contains a compound selected from the group consisting of compounds I-236, I-253, I-275, II-103, II-121, II-174, II-203, II-225 and II-233, or a pharma- ceutical acceptable salt thereof.
  • the pharmaceutical composition according to [1], wherein the fatty liver disease is nonalcoholic fatty liver disease (NAFLD).
  • NAFLD nonalcoholic fatty liver disease
  • the fatty liver disease is metabolic dysfunction-associated fatty liver disease (MASLD), cryptogenic fatty liver disease (Cryptogenic SLD) or specific aetiology fatty liver disease (Specific aetiology SLD).
  • the pharmaceutical composition according to [1], wherein the fatty liver disease is nonalcoholic steatohepatitis (NASH).
  • NASH nonalcoholic steatohepatitis
  • MASH metabolic dysfunction-associated steatohepatitis
  • the pharmaceutical composition according to [1], wherein the fatty liver disease is hepatic fibrosis caused by NASH or MASH.
  • the pharmaceutical composition according to [1], wherein the fatty liver disease is cirrhosis caused by NASH or MASH.
  • the pharmaceutical composition according to [1], wherein the fatty liver disease is hepatocellular carcinoma (HCC) caused by NASH or MASH.
  • HCC hepatocellular carcinoma
  • Test Example 5 Evaluation of toxicity by repeated administration for four days in rats The toxicity of the compounds and/or drugs according to the present invention was evaluated in Crl:CD (SD) rats.
  • Six-week-old male Crl:CD (SD) rats were administered the compound according to the present invention or BMS-963272 by gavage once a day for 4 days, and general observations, body weight and food intake measurements, hematology and blood chemistry tests, and pathological tests were performed. (result) In the groups administered with compounds II-121 and II-203, no toxicity was observed up to the maximum dose of 500 mg/kg/day.
  • reaction conditions were as follows: substrate, 0.5 ⁇ mol/L ethoxyresorufin (CYP1A2), 100 ⁇ mol/L tolbutamide (CYP2C9), 50 ⁇ mol/L S-mephenytoin (CYP2C19), 5 ⁇ mol/L dextromethorphan (CYP2D6), 1 ⁇ mol/L terfenadine (CYP3A4); reaction time, 15 minutes; reaction temperature, 37° C.; enzyme, pooled human liver microsome, 0.2 mg protein/mL; compound of the present invention concentration, 1, 5, 10, 20 ⁇ mol/L (4 points).
  • resorufin (CYP1A2 metabolite) in the centrifugal supernatant was quantified by a fluorescent multi-label counter or LC/MS/MS, and tolbutamide hydroxylated form (CYP2C9 metabolite), mephenytoin 4' hydroxylated form (CYP2C19 metabolite), dextrorphan (CYP2D6 metabolite), and terfenadine alcohol form (CYP3A4 metabolite) were quantified by LC/MS/MS.
  • a reaction system in which only DMSO, the solvent in which the drug was dissolved, was added was used as a control (100%). The remaining activity (%) was calculated, and the IC50 was calculated by inverse estimation using a logistic model using the concentration and inhibition rate.
  • Compound II-103 5 species >20 ⁇ mol/L
  • the area under the plasma concentration-time curve (AUC) of the compound according to the present invention was calculated using the nonlinear least squares program WinNonlin (registered trademark) for the plasma concentration transition of the compound according to the present invention, and the bioavailability (BA) of the compound according to the present invention was calculated from the AUC of the oral administration group and the intravenous administration group.
  • Test Example 12 CYP3A4 (MDZ) MBI Test This is a test to evaluate the mechanism based inhibition (MBI) ability of the compound of the present invention from the enhancement by metabolic reaction regarding CYP3A4 inhibition. CYP3A4 inhibition was evaluated using pooled human liver microsomes with the 1-hydroxylation reaction of midazolam (MDZ) as an index.
  • reaction conditions were as follows: substrate, 10 ⁇ mol/L MDZ; pre-reaction time, 0 or 30 minutes; reaction time, 2 minutes; reaction temperature, 37° C.; pooled human liver microsomes, 0.5 mg/mL at pre-reaction and 0.05 mg/mL (at 10-fold dilution) at reaction; concentration of the compound according to the present invention at pre-reaction, 1, 5, 10, and 20 ⁇ mol/L (4 points).
  • a portion of the solution was transferred to another plate so as to be diluted 1/10 with the substrate and K-Pi buffer, and a reaction using the indicator was started.
  • the plates in which the respective indicator reactions were performed were centrifuged at 3000 rpm for 15 minutes, and 1-hydroxymidazolam in the centrifugal supernatant was quantified by LC/MS/MS.
  • the reaction system was treated as a control (100%) with only DMSO, the solvent in which the compound according to the present invention was dissolved, and the residual activity (%) was calculated when the compound according to the present invention was added at each concentration.
  • the IC was calculated by inverse estimation using a logistic model using the concentration and inhibition rate.
  • the IC at 0 min preincubation/IC at 30 min preincubation was used as the Shifted IC value, and if the Shifted IC was 1.5 or more, it was considered positive, and if the Shifted IC was 1.0 or less, it was considered negative.
  • Test Example 13 Powder solubility test An appropriate amount of the compound according to the present invention was placed in an appropriate container, and 200 ⁇ L of JP-1 solution (2.0 g of sodium chloride, 7.0 mL of hydrochloric acid, and water added to make 1000 mL), JP-2 solution (500 mL of water added to 500 mL of phosphate buffer solution at pH 6.8), and 20 mmol/L sodium taurocholate (TCA)/JP-2 solution (1.08 g of TCA and JP-2 solution added to make 100 mL) were added to each container. If the entire amount was dissolved after the addition of the test solution, the compound according to the present invention was appropriately added. The container was sealed and shaken at 37° C.
  • JP-1 solution 2.0 g of sodium chloride, 7.0 mL of hydrochloric acid, and water added to make 1000 mL
  • JP-2 solution 500 mL of water added to 500 mL of phosphate buffer solution at pH 6.8
  • TCA sodium taurocholate
  • the compound according to the present invention was quantified using HPLC by the absolute calibration curve method.
  • Test Example 14 Fluctuation Ames test The mutagenicity of the compound according to the present invention was evaluated. 20 ⁇ L of frozen Salmonella typhimurium (Salmonella typhimurium TA98 strain, TA100 strain) was inoculated into 10 mL of liquid nutrient medium (2.5% Oxoid nutrient broth No. 2) and pre-cultured with shaking for 10 hours at 37° C. For the TA98 strain, 8.0 mL of the bacterial solution was centrifuged (2000 ⁇ g, 10 minutes) to remove the culture medium.
  • the bacteria were suspended in 8.0 mL of Micro F buffer (K 2 HPO 4 : 3.5 g / L, KH 2 PO 4 : 1 g / L, (NH 4 ) 2 SO 4 : 1 g / L, trisodium citrate dihydrate: 0.25 g / L, MgSO 4 ⁇ 7H 2 0: 0.1 g / L) and added to 120 mL of Exposure medium (Micro F buffer containing biotin: 8 ⁇ g / mL, histidine: 0.2 ⁇ g / mL, glucose: 8 mg / mL).
  • Exposure medium Micro F buffer containing biotin: 8 ⁇ g / mL, histidine: 0.2 ⁇ g / mL, glucose: 8 mg / mL.
  • TA100 strain was added to 120 mL of Exposure medium per 3.1 mL of bacterial solution to prepare a test bacterial solution.
  • the compound DMSO solution according to the present invention (diluted in several stages at a common ratio of 2 to 3 times from the maximum dose of 50 mg / mL), DMSO as a negative control, 50 ⁇ g / mL 4-nitroquinoline-1-oxide DMSO solution for the TA98 strain as a positive control under non-metabolic activation conditions, 0.25 ⁇ g / mL 2- (2-furyl) -3- (5-nitro-2-furyl) acrylamide DMSO solution for the TA100 strain, 40 ⁇ g / mL 2-aminoanthracene DMSO solution for the TA98 strain under metabolic activation conditions, 20 ⁇ g / mL 2-aminoanthracene DMSO solution for the TA100 strain were mixed with 12 ⁇ L each and 588 ⁇ L of test bacteria solution (a mixture of 498 ⁇ L of test bacteria solution and 90 ⁇ L of S9 mix under metabolic activation conditions), and cultured with shaking at 37 ° C.
  • bacterial solution exposed to the compound according to the present invention was mixed with 1150 ⁇ L of Indicator medium (MicroF buffer containing biotin: 8 ⁇ g/mL, histidine: 0.2 ⁇ g/mL, glucose: 8 mg/mL, bromocresol purple: 37.5 ⁇ g/mL), and 50 ⁇ L was dispensed into 48 wells/dose of a microplate, and cultured at 37° C. for 3 days.
  • Indicator medium MicroF buffer containing biotin: 8 ⁇ g/mL, histidine: 0.2 ⁇ g/mL, glucose: 8 mg/mL, bromocresol purple: 37.5 ⁇ g/mL
  • Test Example 15 For the purpose of evaluating the risk of electrocardiogram QT interval prolongation of the compound according to the present invention, the effect of the compound according to the present invention on delayed rectifier K + current (I Kr ), which plays an important role in the ventricular repolarization process, was investigated using CHO cells expressing human ether-a-go-go related gene (hERG) channel.
  • I Kr delayed rectifier K + current
  • the cells were held at a membrane potential of -80 mV by the whole-cell patch clamp method, and a leak potential of -50 mV was applied, followed by a depolarizing stimulus of +20 mV for 2 seconds and a repolarizing stimulus of -50 mV for 2 seconds.
  • the I Kr evoked by this was recorded.

Landscapes

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

Abstract

Provided is a pharmaceutical composition for treating and/or preventing steatotic liver diseases, particularly non-alcoholic fatty liver diseases, which has an excellent MGAT2 inhibitory effect and does not involve adverse side effects such as hemostatic abnormality, hepatic and renal disorders or embryonic lethality. The pharmaceutical composition contains a compound represented by formula (I) or a pharmaceutically acceptable salt thereof. Formula (I): (wherein: R2a and R2b together, in conjunction with the adjacent carbon atom, form a ring B; the ring B is represented by any one of the formulae shown in the middle part of FIG. 1 (wherein R6's each independently represent a halogen atom or the like); R4a is represented by the formula shown in the lower part of FIG. 1 (wherein L3 represents an alkylene group, and R7 represents an alkylsulfonyl group); R4b represents an alkyl group which may be substituted by a group α substituent, or the like; the group α substituent is a halogen atom, or the like; and a group β substituent is a halogen atom, or the like.)

Description

脂肪性肝疾患の治療用医薬Medicine for treating fatty liver disease

 本発明は、脂肪性肝疾患、特に非アルコール性脂肪肝疾患を治療及び/又は予防するための、新たな医薬組成物に関する。 The present invention relates to a new pharmaceutical composition for treating and/or preventing fatty liver disease, particularly non-alcoholic fatty liver disease.

 肝疾患は、世界中で主要な死因である。肝疾患は、感染、傷害、薬物又は毒性の化合物、アルコール、食物中の不純物への曝露、及び血液中の正常な物質の異常な蓄積、自己免疫過程、遺伝子欠損(ヘモクロマトーシスなど)又は未知の原因(複数可)により引き起こされ得る。肝疾患は、疾患の期間に基づいて急性又は慢性と一般的に分類される。
 脂肪性肝疾患(steatotic liver disease: SLD)は、肝臓に中性脂肪が蓄積して肝障害を引き起こす疾患の総称である。肝臓の組織で、脂肪滴を伴う肝細胞が30%以上認められる場合を脂肪肝という。
 脂肪肝は肝臓内に中性脂肪が蓄積した状態であり、肝臓での脂肪の蓄積は非進行性(可逆的)変化であるため、原因を除去すると肝臓での脂肪蓄積は正常に戻る。しかし、脂肪の蓄積が進行し、肝機能障害が生じることになると、肝硬変や肝臓ガンに発展する場合がある。飲酒が原因のアルコール性脂肪性肝炎(alcoholic steatohepatitis: ASH)が脂肪肝の典型例である。
 一方で、アルコールをほとんど摂取しない人にも脂肪肝が生じる場合があり、これは非アルコール性脂肪肝疾患(nonalcoholic fatty liver disease: NAFLD)と呼ばれている。NAFLDの中でも重度のものは、非アルコール性脂肪肝炎(nonalcoholic steato-hepatitis:NASH)と呼ばれている。NASHは肝硬変や肝臓ガンの重要な原因と考えられている。
 アルコール性と非アルコール性、ふたつの肝炎の組織像は類似しており、肝臓内の酸化ストレスなど共通の発症メカニズムが研究されている。
 米国肝臓協会によると、人口の20%より多くがNAFLDを有する。無処置のままだと、NAFLDは、重篤な有害作用を引き起こすNASHに進行し得る。さらにNASHは、未処置である場合、肝線維症、肝硬変症、肝不全、又は肝細胞癌をもたらす可能性がある。米国における概算1600万人の成人がNASHを有し、概ね50%はNASHに関連する進行した肝線維症(架橋状線維症又は肝硬変)を有する。NASHは現在、米国において、肝細胞癌の主要な原因であり、C型肝炎に次いで2番目に多い肝移植のための主要な適応症である。NASHは、脂肪症の存在によって、ならびに肝細胞性変性(気球状、マロリーヒアリン)、炎症性細胞浸潤及び進行性線維症の発達を含む他の特性によって特徴付けられる。
 NASHの処置のための現在承認されている療法も、NASH患者において線維症及び/又は脂肪症を低減する療法もない。したがって、肝疾患又は肝疾患の症状を処置するための新しい有効な薬剤を提供する必要性がまだある。
Liver disease is a leading cause of death worldwide. Liver disease can be caused by infection, injury, exposure to drugs or toxic compounds, alcohol, impurities in food, and abnormal accumulation of normal substances in the blood, autoimmune processes, genetic defects (such as hemochromatosis), or unknown cause(s). Liver disease is commonly classified as acute or chronic based on the duration of the disease.
Fatty liver disease (SLD) is a general term for diseases in which accumulation of neutral fat in the liver causes liver damage. A case in which 30% or more of hepatocytes in liver tissue contain lipid droplets is called fatty liver.
Fatty liver is a state where neutral fat accumulates in the liver, and since the accumulation of fat in the liver is a non-progressive (reversible) change, the accumulation of fat in the liver returns to normal when the cause is removed. However, if the accumulation of fat progresses and liver dysfunction occurs, it may develop into cirrhosis or liver cancer. Alcoholic steatohepatitis (ASH), which is caused by drinking alcohol, is a typical example of fatty liver.
On the other hand, fatty liver can occur even in people who consume very little alcohol, and this condition is called nonalcoholic fatty liver disease (NAFLD). The most severe form of NAFLD is called nonalcoholic steatohepatitis (NASH). NASH is considered to be an important cause of cirrhosis and liver cancer.
The histological findings of alcoholic and non-alcoholic hepatitis are similar, and common mechanisms of onset, such as oxidative stress in the liver, are being studied.
According to the American Liver Association, more than 20% of the population has NAFLD. If left untreated, NAFLD can progress to NASH, which causes serious adverse effects. Furthermore, if untreated, NASH can lead to liver fibrosis, cirrhosis, liver failure, or hepatocellular carcinoma. An estimated 16 million adults in the United States have NASH, and roughly 50% have advanced liver fibrosis (bridging fibrosis or cirrhosis) associated with NASH. NASH is currently the leading cause of hepatocellular carcinoma in the United States and the second most common indication for liver transplantation after hepatitis C. NASH is characterized by the presence of steatosis, as well as other characteristics including hepatocellular degeneration (balloon, Mallory hyaline), inflammatory cell infiltration, and the development of progressive fibrosis.
There are no currently approved therapies for the treatment of NASH or that reduce fibrosis and/or steatosis in NASH patients. Thus, there remains a need to provide new effective agents for treating liver disease or symptoms of liver disease.

 特許文献1には、NASH及び/又は原発性胆汁性肝硬変の治療及び/又は予防方法が記載されており、該方法は、エイコサペンタエン酸又はこれらの誘導体を含む医薬組成物を、それを必要としている対象に投与することを含む。半合成胆汁酸で、核内受容体FXR(farnesoid X receptor)に対するアゴニストであるオベチコール酸は、欧米ではNASHに起因する線維症を適応として申請されたことが報告されている。 Patent Document 1 describes a method for treating and/or preventing NASH and/or primary biliary cirrhosis, which comprises administering a pharmaceutical composition containing eicosapentaenoic acid or a derivative thereof to a subject in need thereof. It has been reported that obeticholic acid, a semisynthetic bile acid and an agonist for the nuclear receptor FXR (farnesoid X receptor), has been applied for in Europe and the United States for the treatment of fibrosis caused by NASH.

 モノアシルグリセロールアシルトランスフェラーゼ2(monoacylglycerol acyltransferase2、以下MGAT2とも称する)ノックアウ卜マウスにおいて、高脂肪食負荷による体重増加の抑制、インスリン抵抗性惹起の抑制、血中コレステロール上昇の抑制、脂肪肝形成などの抑制、エネルギー消費の亢進が確認されている(非特許文献1)。また、MGAT2阻害活性を有する化合物(JTP-103237、BMS-963272が、高スクロース負荷モデルマウスの脂肪肝を改善させることや、NASHモデルマウスの肝臓線維化病態を改善させることが報告されている(非特許文献2、3)。さらに、NAFLD患者の肝臓でΜGAT2の発現が亢進したことが報告されている(非特許文献4)。
 これらの知見から、MGAT2阻害剤がNASHを含む脂肪性肝疾患治療薬として有望であることが期待されているが、ヒトで効果を示す薬剤は未だ開発されていない(非特許文献5)。
In monoacylglycerol acyltransferase 2 (hereinafter also referred to as MGAT2) knockout mice, it has been confirmed that high-fat diet-induced weight gain, insulin resistance, blood cholesterol elevation, fatty liver formation, and energy consumption are suppressed (Non-Patent Document 1). In addition, it has been reported that compounds having MGAT2 inhibitory activity (JTP-103237, BMS-963272 ) improve fatty liver in high sucrose-loaded model mice and improve liver fibrosis in NASH model mice (Non-Patent Documents 2 and 3). Furthermore, it has been reported that expression of MGAT2 is increased in the liver of NAFLD patients (Non-Patent Document 4).
Based on these findings, it is expected that MGAT2 inhibitors are promising therapeutic agents for fatty liver diseases including NASH, but no drug that shows efficacy in humans has yet been developed (Non-Patent Document 5).

 特許文献2~4にはMGAT2阻害剤である式:

Figure JPOXMLDOC01-appb-C000006

で示される化合物が記載されている。
 また特許文献5~6には、MGAT2阻害剤である式:
Figure JPOXMLDOC01-appb-C000007

で示される化合物が記載されている。
 しかしながら、これら特許文献2~6には非アルコール性脂肪肝疾患の治療及び/又は予防に有効であることは記載されていない。 Patent documents 2 to 4 disclose MGAT2 inhibitors of the formula:
Figure JPOXMLDOC01-appb-C000006

The compound represented by the formula:
Furthermore, Patent Documents 5 and 6 disclose MGAT2 inhibitors of the formula:
Figure JPOXMLDOC01-appb-C000007

The compound represented by the formula:
However, Patent Documents 2 to 6 do not disclose that they are effective in treating and/or preventing non-alcoholic fatty liver disease.

米国出願公開公報2014/187633号US Patent Publication No. 2014/187633 国際公開第2019013311号International Publication No. 2019013311 国際公開第2019013312号International Publication No. 2019013312 特願2020-02515号Patent Application No. 2020-02515 国際公開第2020145369号International Publication No. 2020145369 特願2021-113901号Patent application No. 2021-113901

Nature Medicine volume 15, pages442-446 (2009)Nature Medicine volume 15, pages442-446 (2009) Journal of Pharmacological Sciences 128 (2015) 150e157Journal of Pharmacological Sciences 128 (2015) 150e157 Cell Metabolism 34, 1732-1748, 2022Cell Metabolism 34, 1732-1748, 2022 Journal of Lipid Research Volume 53, 2012, p990-999Journal of Lipid Research Volume 53, 2012, p990-999 J. Med. Chem. 2018, 61, 9879-9888J. Med. Chem. 2018, 61, 9879-9888

 本発明の課題は、優れたMGAT2阻害作用を有し、止血異常、肝障害腎障害又は胚致死性などの副作用を伴わない、脂肪性肝疾患、特に非アルコール性脂肪肝疾患の治療及び/又は予防のための医薬組成物を提供することにある。 The object of the present invention is to provide a pharmaceutical composition for the treatment and/or prevention of fatty liver disease, particularly non-alcoholic fatty liver disease, which has excellent MGAT2 inhibitory activity and is free of side effects such as hemostatic abnormalities, liver damage, renal damage, or embryonic lethality.

 本発明者らは、前記課題を解決するために検討を重ねた結果、特許文献2~4に記載のMGAT2阻害作用を有する化合物のうち、特定の化合物が非アルコール性脂肪肝疾患の治療及び/又は予防に有効であり、かつ、止血異常、肝障害腎障害又は胚致死性などの副作用を伴わないことを見出し、本発明を完成するに至った。本発明は、以下に関する。 As a result of extensive investigations to solve the above problems, the present inventors discovered that, among the compounds having MGAT2 inhibitory activity described in Patent Documents 2 to 4, certain compounds are effective in treating and/or preventing non-alcoholic fatty liver disease and are free of side effects such as hemostatic abnormalities, liver damage, renal damage, or embryonic lethality, and thus completed the present invention. The present invention relates to the following.

[1]式(I):

Figure JPOXMLDOC01-appb-C000008

(式中、
 R2a及びR2bは、隣接する炭素原子と一緒になって、環Bを形成し;
 環Bは、式:
Figure JPOXMLDOC01-appb-C000009

(式中、Rは、それぞれ独立して、ハロゲン、ハロアルキル、アルキルオキシ、ハロアルキルオキシ、又はシクロプロパニルである)であり;
 R4aは、式:
Figure JPOXMLDOC01-appb-C000010

(式中、
 Lは、アルキレンであり、
 Rは、アルキルスルフォニルである)であり;
 R4bは、置換基群αで置換されていてもよいアルキル、置換基群βで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり;
 置換基群αは、ハロゲン、ハロアルキルオキシ、及びシクロプロパニルであり、
 置換基群βは、ハロゲン、シアノ、アルキル、ハロアルキル、及びシクロプロパニルである)で示される化合物又はその製薬上許容される塩を含有する、脂肪性肝疾患を治療及び/又は予防するための、医薬組成物。
[2]Rが、それぞれ独立して、ハロゲン、ハロアルキル、又はハロアルキルオキシであり、
4aが、式:
Figure JPOXMLDOC01-appb-C000011

であり、
4bが、ハロアルキルオキシアルキル、ハロゲンで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり、
 置換基群βは、ハロゲン及びアルキルである、[1]記載の医薬組成物。
[3]環Bが、式:
Figure JPOXMLDOC01-appb-C000012

(式中、各記号は[1]又は[2]と同意義)
である、[1]又は[2]記載の医薬組成物。
[4]化合物I-8、I-23、I-34、I-190、I-212、I-236、I-253、I-275、I-276、II-93、II-103、II-121、II-151、II-168、II-174、II-203、II-225、II-233及びII-295からなる群から選択される化合物又はその製薬上許容される塩を含有する、[1]記載の医薬組成物。
[5]化合物I-236、I-253、I-275、II-103、II-121、II-174、II-203、II-225及びII-233からなる群から選択される化合物又はその製薬上許容される塩を含有する、[1]記載の医薬組成物。
[6]前記脂肪性肝疾患が非アルコール性脂肪肝疾患(NAFLD)である、[1]記載の医薬組成物。
[7]前記脂肪性肝疾患が代謝機能障害関連脂肪性肝疾患(MASLD)、成因不明脂肪性肝疾患(Cryptogenic SLD)又は特定成因脂肪性肝疾患(Specific aetiology SLD)である、[1]記載の医薬組成物。
[8]前記脂肪性肝疾患が非アルコール性脂肪肝炎(NASH)である、[1]記載の医薬組成物。
[9]前記脂肪性肝疾患が代謝機能障害関連脂肪肝炎(MASH)である、[1]記載の医薬組成物。
[10]前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝線維症である、[1]記載の医薬組成物。
[11]前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝硬変である、[1]記載の医薬組成物。
[12]前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝細胞癌(HCC)である、[1]記載の医薬組成物。
[13]前記医薬組成物の投与によって、止血異常、肝障害、腎障害及び胚致死性の少なくとも1つの副作用を伴わない、[1]記載の医薬組成物。
[14]身体質量指数(BMI)が25kg/m未満である対象における脂肪性肝疾患を治療及び/又は予防するための、[1]~[13]のいずれかに記載の医薬組成物。
[15]脂肪性肝疾患を治療及び/又は予防するための、[1]~[14]のいずれかに記載の医薬組成物であって、有効量の化合物が対象に投与される、医薬組成物。
[16]前記化合物の有効量が1日あたり0.5mg~100mgの範囲である、[15]記載の医薬組成物。
[17]前記化合物の有効量が1日あたり1mg~50mgの範囲である、[16]記載の医薬組成物。
[18]前記化合物の有効量が1日あたり3mg~30mgの範囲である、[17]記載の医薬組成物。
[19]脂肪性肝疾患の治療及び/又は予防方法であって、式:
Figure JPOXMLDOC01-appb-C000013

(式中、
 R2a及びR2bは、隣接する炭素原子と一緒になって、環Bを形成し;
 環Bは、式:
Figure JPOXMLDOC01-appb-C000014

(式中、Rは、それぞれ独立して、ハロゲン、ハロアルキル、アルキルオキシ、ハロアルキルオキシ、又はシクロプロパニルである)であり;
 R4aは、式:
Figure JPOXMLDOC01-appb-C000015

(式中、
 Lは、アルキレンであり、
 Rは、アルキルスルフォニルである)であり;
 R4bは、置換基群αで置換されていてもよいアルキル、置換基群βで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり;
 置換基群αは、ハロゲン、ハロアルキルオキシ、及びシクロプロパニルであり、
 置換基群βは、ハロゲン、シアノ、アルキル、ハロアルキル、及びシクロプロパニルである)で示される化合物又はその製薬上許容される塩の有効量を、脂肪性肝疾患の治療及び/又は予防を必要とする個体に投与する工程を含む、脂肪性肝疾患の治療及び/又は予防方法。
[20]脂肪性肝疾患の治療及び/又は予防用医薬を製造するための、式:
Figure JPOXMLDOC01-appb-C000016

(式中、
 R2a及びR2bは、隣接する炭素原子と一緒になって、環Bを形成し;
 環Bは、式:
Figure JPOXMLDOC01-appb-C000017

(式中、Rは、それぞれ独立して、ハロゲン、ハロアルキル、アルキルオキシ、ハロアルキルオキシ、又はシクロプロパニルである)であり;
 R4aは、式:
Figure JPOXMLDOC01-appb-C000018

(式中、
 Lは、アルキレンであり、
 Rは、アルキルスルフォニルである)であり;
 R4bは、置換基群αで置換されていてもよいアルキル、置換基群βで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり;
 置換基群αは、ハロゲン、ハロアルキルオキシ、及びシクロプロパニルであり、
 置換基群βは、ハロゲン、シアノ、アルキル、ハロアルキル、及びシクロプロパニルである)で示される化合物又はその製薬上許容される塩の使用。
[21]脂肪性肝疾患を治療及び/又は予防するための、式:
Figure JPOXMLDOC01-appb-C000019

(式中、
 R2a及びR2bは、隣接する炭素原子と一緒になって、環Bを形成し;
 環Bは、式:
Figure JPOXMLDOC01-appb-C000020

(式中、Rは、それぞれ独立して、ハロゲン、ハロアルキル、アルキルオキシ、ハロアルキルオキシ、又はシクロプロパニルである)であり;
 R4aは、式:
Figure JPOXMLDOC01-appb-C000021

(式中、
 Lは、アルキレンであり、
 Rは、アルキルスルフォニルである)であり;
 R4bは、置換基群αで置換されていてもよいアルキル、置換基群βで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり;
 置換基群αは、ハロゲン、ハロアルキルオキシ、及びシクロプロパニルであり、
 置換基群βは、ハロゲン、シアノ、アルキル、ハロアルキル、及びシクロプロパニルである)で示される化合物又はその製薬上許容される塩。 [1] Formula (I):
Figure JPOXMLDOC01-appb-C000008

(In the formula,
R 2a and R 2b together with adjacent carbon atoms form ring B;
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000009

wherein each R6 is independently halogen, haloalkyl, alkyloxy, haloalkyloxy, or cyclopropanyl;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000010

(In the formula,
L3 is alkylene;
R 7 is alkylsulfonyl;
R 4b is an alkyl group optionally substituted with substituent group α, a phenyl group optionally substituted with substituent group β, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β;
Substituent group α is halogen, haloalkyloxy, and cyclopropanyl;
A pharmaceutical composition for treating and/or preventing fatty liver disease, comprising a compound represented by the formula (I) or a pharma- ceutically acceptable salt thereof, wherein substituent group β is halogen, cyano, alkyl, haloalkyl, and cyclopropanyl.
[2] Each R 6 is independently halogen, haloalkyl, or haloalkyloxy;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000011

and
R 4b is a haloalkyloxyalkyl, a phenyl group optionally substituted with halogen, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β,
The pharmaceutical composition according to [1], wherein the substituent group β is halogen and alkyl.
[3] Ring B is represented by the formula:
Figure JPOXMLDOC01-appb-C000012

(In the formula, each symbol has the same meaning as [1] or [2].)
The pharmaceutical composition according to [1] or [2],
[4] The pharmaceutical composition according to [1], which contains a compound selected from the group consisting of compounds I-8, I-23, I-34, I-190, I-212, I-236, I-253, I-275, I-276, II-93, II-103, II-121, II-151, II-168, II-174, II-203, II-225, II-233 and II-295, or a pharma- ceutical acceptable salt thereof.
[5] The pharmaceutical composition according to [1], which contains a compound selected from the group consisting of compounds I-236, I-253, I-275, II-103, II-121, II-174, II-203, II-225 and II-233, or a pharma- ceutical acceptable salt thereof.
[6] The pharmaceutical composition according to [1], wherein the fatty liver disease is nonalcoholic fatty liver disease (NAFLD).
[7] The pharmaceutical composition according to [1], wherein the fatty liver disease is metabolic dysfunction-associated fatty liver disease (MASLD), cryptogenic fatty liver disease (Cryptogenic SLD) or specific aetiology fatty liver disease (Specific aetiology SLD).
[8] The pharmaceutical composition according to [1], wherein the fatty liver disease is nonalcoholic steatohepatitis (NASH).
[9] The pharmaceutical composition according to [1], wherein the fatty liver disease is metabolic dysfunction-associated steatohepatitis (MASH).
[10] The pharmaceutical composition according to [1], wherein the fatty liver disease is hepatic fibrosis caused by NASH or MASH.
[11] The pharmaceutical composition according to [1], wherein the fatty liver disease is cirrhosis caused by NASH or MASH.
[12] The pharmaceutical composition according to [1], wherein the fatty liver disease is hepatocellular carcinoma (HCC) caused by NASH or MASH.
[13] The pharmaceutical composition according to [1], wherein administration of the pharmaceutical composition does not result in at least one side effect of hemostatic abnormalities, liver damage, kidney damage, and embryonic lethality.
[14] The pharmaceutical composition according to any one of [1] to [13], for treating and/or preventing fatty liver disease in a subject having a body mass index (BMI) of less than 25 kg/ m2 .
[15] The pharmaceutical composition according to any one of [1] to [14] for treating and/or preventing fatty liver disease, wherein an effective amount of the compound is administered to a subject.
[16] The pharmaceutical composition according to [15], wherein the effective amount of the compound is in the range of 0.5 mg to 100 mg per day.
[17] The pharmaceutical composition according to [16], wherein the effective amount of the compound is in the range of 1 mg to 50 mg per day.
[18] The pharmaceutical composition according to [17], wherein the effective amount of the compound is in the range of 3 mg to 30 mg per day.
[19] A method for treating and/or preventing fatty liver disease, comprising administering to a patient having the formula:
Figure JPOXMLDOC01-appb-C000013

(In the formula,
R 2a and R 2b together with adjacent carbon atoms form ring B;
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000014

wherein each R6 is independently halogen, haloalkyl, alkyloxy, haloalkyloxy, or cyclopropanyl;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000015

(In the formula,
L3 is alkylene;
R 7 is alkylsulfonyl;
R 4b is an alkyl group optionally substituted with substituent group α, a phenyl group optionally substituted with substituent group β, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β;
Substituent group α is halogen, haloalkyloxy, and cyclopropanyl;
and R 1 is an integer from 1 to 3. The present invention relates to a method for treating and/or preventing fatty liver disease, the method comprising the step of administering an effective amount of a compound represented by the formula:
[20] A compound according to the formula:
Figure JPOXMLDOC01-appb-C000016

(In the formula,
R 2a and R 2b together with adjacent carbon atoms form ring B;
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000017

wherein each R6 is independently halogen, haloalkyl, alkyloxy, haloalkyloxy, or cyclopropanyl;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000018

(In the formula,
L3 is alkylene;
R 7 is alkylsulfonyl;
R 4b is an alkyl group optionally substituted with substituent group α, a phenyl group optionally substituted with substituent group β, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β;
Substituent group α is halogen, haloalkyloxy, and cyclopropanyl;
Substituent group β is halogen, cyano, alkyl, haloalkyl, and cyclopropanyl, or a pharma- ceutically acceptable salt thereof.
[21] A compound of the formula:
Figure JPOXMLDOC01-appb-C000019

(In the formula,
R 2a and R 2b together with adjacent carbon atoms form ring B;
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000020

wherein each R6 is independently halogen, haloalkyl, alkyloxy, haloalkyloxy, or cyclopropanyl;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000021

(In the formula,
L3 is alkylene;
R 7 is alkylsulfonyl;
R 4b is an alkyl group optionally substituted with substituent group α, a phenyl group optionally substituted with substituent group β, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β;
Substituent group α is halogen, haloalkyloxy, and cyclopropanyl;
Substituent group β is halogen, cyano, alkyl, haloalkyl, and cyclopropanyl, or a pharma- ceutically acceptable salt thereof.

 本発明の医薬組成物は、脂肪性肝疾患、特に非アルコール性脂肪肝疾患の治療及び/又は予防に有効であるという優れた効果を奏するものである。また、止血異常、肝障害腎障害及び胚致死性の少なくとも1つの副作用を伴わない、高い安全性を有する。 The pharmaceutical composition of the present invention has an excellent effect of being effective in treating and/or preventing fatty liver disease, particularly non-alcoholic fatty liver disease. In addition, it has a high level of safety, being free from at least one of the side effects of hemostatic abnormalities, liver damage, renal damage, and embryonic lethality.

は化合物II-203、II-121及びBMS-963272をHepG2細胞に一晩処置した際の細胞内脂肪蓄積含量を示す。1 shows the intracellular fat accumulation content in HepG2 cells after overnight treatment with compounds II-203, II-121 and BMS-963272. は化合物II-203、II-121及びBMS-963272をHepG2細胞に48時間処置した際の細胞内ATP含量を示す。1 shows the intracellular ATP content when compounds II-203, II-121 and BMS-963272 were treated in HepG2 cells for 48 hours. は化合物II-203、II-121及びBMS-963272をHepG2細胞に24時間処置した際の培養上清中LDH活性を示す。4 shows LDH activity in the culture supernatant of HepG2 cells treated with compounds II-203, II-121 and BMS-963272 for 24 hours.

 以下に本明細書において用いられる各用語の意味を説明する。各用語は特に断りのない限り、単独で用いられる場合も、又は他の用語と組み合わせて用いられる場合も、同一の意味で用いられる。
 「からなる」という用語は、構成要件のみを有することを意味する。
 「含む」という用語は、構成要件に限定されず、記載されていない要素を排除しないことを意味する。
 以下、本発明について実施形態を示しながら説明する。本明細書の全体にわたり、単数形の表現は、特に言及しない限り、その複数形の概念をも含むことが理解されるべきである。従って、単数形の冠詞(例えば、英語の場合は「a」、「an」、「the」など)は、特に言及しない限り、その複数形の概念をも含むことが理解されるべきである。
 また、本明細書において使用される用語は、特に言及しない限り、当上記分野で通常用いられる意味で用いられることが理解されるべきである。したがって、他に定義されない限り、本明細書中で使用される全ての専門用語及び科学技術用語は、本発明の属する分野の当業者によって一般的に理解されるのと同じ意味を有する。矛盾する場合、本明細書(定義を含めて)が優先する。
The meaning of each term used in this specification will be explained below. Unless otherwise specified, each term has the same meaning whether used alone or in combination with other terms.
The term "consisting of" means having only the constituent elements.
The term "comprising" is meant to be open ended and does not exclude unrecited elements.
Hereinafter, the present invention will be described with reference to the embodiments. Throughout this specification, it should be understood that the singular expression includes the plural concept unless otherwise specified. Therefore, it should be understood that the singular article (for example, in the case of English, "a", "an", "the", etc.) includes the plural concept unless otherwise specified.
In addition, it should be understood that the terms used in this specification are used in the same sense as commonly used in the above field unless otherwise specified. Therefore, unless otherwise defined, all technical and scientific terms used in this specification have the same meaning as commonly understood by those skilled in the art to which this invention belongs. In case of conflict, the present specification (including definitions) shall prevail.

 「ハロゲン」とは、フッ素原子、塩素原子、臭素原子、及びヨウ素原子を包含する。特にフッ素原子及び塩素原子が好ましい。 "Halogen" includes fluorine, chlorine, bromine, and iodine atoms. Fluorine and chlorine atoms are particularly preferred.

 「アルキル」とは、炭素数1~15、好ましくは炭素数1~10、より好ましくは炭素数1~6、さらに好ましくは炭素数1~4の直鎖又は分枝状の炭化水素基を包含する。例えば、メチル、エチル、n-プロピル、イソプロピル、n-ブチル、イソブチル、sec-ブチル、tert-ブチル、n-ペンチル、イソペンチル、ネオペンチル、n-ヘキシル、イソヘキシル、n-へプチル、イソヘプチル、n-オクチル、イソオクチル、n-ノニル、n-デシル等が挙げられる。
 「アルキル」の好ましい態様として、メチル、エチル、n-プロピル、イソプロピル、n-ブチル、イソブチル、sec-ブチル、tert-ブチル、n-ペンチルが挙げられる。さらに好ましい態様として、メチル、エチル、n-プロピル、イソプロピル、tert-ブチルが挙げられる。
The term "alkyl" includes straight-chain or branched hydrocarbon groups having 1 to 15 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms, and even more preferably 1 to 4 carbon atoms. Examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, isohexyl, n-heptyl, isoheptyl, n-octyl, isooctyl, n-nonyl, and n-decyl.
Preferred embodiments of "alkyl" include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and n-pentyl. More preferred embodiments include methyl, ethyl, n-propyl, isopropyl, and tert-butyl.

 「アルキレン」とは、炭素数1~15、好ましくは炭素数1~10、より好ましくは炭素数1~6、さらに好ましくは炭素数1~4の直鎖又は分枝状の2価の炭化水素基を包含する。例えば、メチレン、エチレン、トリメチレン、プロピレン、テトラメチレン、ペンタメチレン、ヘキサメチレン等が挙げられる。 "Alkylene" includes straight-chain or branched divalent hydrocarbon groups having 1 to 15 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms, and even more preferably 1 to 4 carbon atoms. Examples include methylene, ethylene, trimethylene, propylene, tetramethylene, pentamethylene, and hexamethylene.

 「芳香族複素環式基」とは、O、S及びNから任意に選択される同一又は異なるヘテロ原子を環内に1以上有する、単環又は2環以上の、芳香族環式基を意味する。
 2環以上の芳香族複素環式基は、単環又は2環以上の芳香族複素環式基に、上記「芳香族炭素環式基」における環が縮合したものも包含し、該結合手はいずれの環に有していても良い。
 単環の芳香族複素環式基としては、5~8員が好ましく、より好ましくは5員又は6員である。5員芳香族複素環式基としては、例えば、ピロリル、イミダゾリル、ピラゾリル、トリアゾリル、テトラゾリル、フリル、チエニル、イソオキサゾリル、オキサゾリル、オキサジアゾリル、イソチアゾリル、チアゾリル、チアジアゾリル等が挙げられる。6員芳香族複素環式基としては、例えば、ピリジル、ピリダジニル、ピリミジニル、ピラジニル、トリアジニル等が挙げられる。
 2環の芳香族複素環式基としては、8~10員が好ましく、より好ましくは9員又は10員である。例えば、インドリル、イソインドリル、インダゾリル、インドリジニル、キノリニル、イソキノリニル、シンノリニル、フタラジニル、キナゾリニル、ナフチリジニル、キノキサリニル、プリニル、プテリジニル、ベンズイミダゾリル、ベンズイソオキサゾリル、ベンズオキサゾリル、ベンズオキサジアゾリル、ベンズイソチアゾリル、ベンゾチアゾリル、ベンゾチアジアゾリル、ベンゾフリル、イソベンゾフリル、ベンゾチエニル、ベンゾトリアゾリル、イミダゾピリジル、トリアゾロピリジル、イミダゾチアゾリル、ピラジノピリダジニル、オキサゾロピリジル、チアゾロピリジル等が挙げられる。
 3環以上の芳香族複素環式基としては、13~15員が好ましい。例えば、カルバゾリル、アクリジニル、キサンテニル、フェノチアジニル、フェノキサチイニル、フェノキサジニル、ジベンゾフリル等が挙げられる。
The term "aromatic heterocyclic group" means a monocyclic or bicyclic or more aromatic cyclic group having one or more identical or different heteroatoms selected from O, S and N in the ring.
The aromatic heterocyclic group having two or more rings also includes a monocyclic or bicyclic or more ring aromatic heterocyclic group condensed with a ring in the above-mentioned "aromatic carbocyclic group", and the bond may be on any of the rings.
The monocyclic aromatic heterocyclic group is preferably 5- to 8-membered, more preferably 5- or 6-membered. Examples of the 5-membered aromatic heterocyclic group include pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, oxazolyl, oxadiazolyl, isothiazolyl, thiazolyl, thiadiazolyl, etc. Examples of the 6-membered aromatic heterocyclic group include pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc.
The bicyclic aromatic heterocyclic group is preferably an 8- to 10-membered ring, more preferably an 9- or 10-membered ring, and examples thereof include indolyl, isoindolyl, indazolyl, indolizinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, naphthyridinyl, quinoxalinyl, purinyl, pteridinyl, benzimidazolyl, benzisoxazolyl, benzoxazolyl, benzoxadiazolyl, benzisothiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, isobenzofuryl, benzothienyl, benzotriazolyl, imidazopyridyl, triazolopyridyl, imidazothiazolyl, pyrazinopyridazinyl, oxazolopyridyl, thiazolopyridyl, and the like.
The aromatic heterocyclic group having three or more rings is preferably a group having 13 to 15 members, and examples thereof include carbazolyl, acridinyl, xanthenyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, and dibenzofuryl.

 本明細書中、「置換基群αで置換されてもよい」とは、「置換基群αから選択される1以上の基で置換されていてもよい」ことを意味する。置換基群βについても同様である。 In this specification, "may be substituted with substituent group α" means "may be substituted with one or more groups selected from substituent group α." The same applies to substituent group β.

 脂肪性肝疾患(SLD)は、肝臓に中性脂肪が蓄積して肝障害を引き起こす疾患の総称である。以前はアルコールが原因であることが多いとされてきたが、最近では飲酒習慣が無いものの肥満を有している人などに増えてきている。このような明らかな飲酒歴が無いにもかかわらず、肝組織所見はアルコール性肝障害に類似した肝脂肪沈着を特徴とする肝障害のことを非アルコール性脂肪肝疾患(NAFLD)と呼んでいる。 Fatty liver disease (SLD) is a general term for diseases in which triglycerides accumulate in the liver, causing liver damage. In the past, it was believed that alcohol was often the cause, but recently it has become more common among obese people who do not drink alcohol. Liver damage characterized by fatty deposits in the liver similar to alcoholic liver disease in liver tissue findings, even in cases where there is no clear history of drinking, is called non-alcoholic fatty liver disease (NAFLD).

 NAFLDは、組織診断、あるいは画像診断で脂肪肝を認め、アルコール性肝障害など他の肝疾患を除外した病態である。NAFLDは肝細胞における脂肪の蓄積によって特徴付けられ、メタボリックシンドロームの一部の態様(例えば、2型真性糖尿病、インスリン抵抗性、高脂血症、高血圧)にしばしば関連している。この疾患の頻度は、炭水化物に富む及び高脂肪の食事の消費のためにますます一般的になっている。NAFLD患者のサブセットは、非アルコール性脂肪肝炎(NASH)を起こす。 NAFLD is a condition characterized by the presence of fatty liver tissue or imaging findings, after exclusion of other liver diseases such as alcoholic liver disease. NAFLD is characterized by the accumulation of fat in hepatocytes and is often associated with some aspects of the metabolic syndrome (e.g., type 2 diabetes mellitus, insulin resistance, hyperlipidemia, hypertension). The frequency of this disease is becoming increasingly common due to the consumption of carbohydrate-rich and high-fat diets. A subset of patients with NAFLD develops nonalcoholic steatohepatitis (NASH).

 脂肪性肝疾患のサブタイプであるNASHは、NAFLDのより重度の病態である。それは、大滴性脂肪変性(macrovesicular  steatosis)、肝細胞のバルーン変性(balloon  degeneration)及び/又は肝臓の瘢痕化(すなわち、線維症)に最終的につながる炎症によって特徴付けられる。NASHと診断された患者は、進行したステージの肝臓線維症及び最終的に肝硬変に進行する。末期疾患の肝硬変NASH患者のための現在の処置は、肝移植である。 NASH, a subtype of fatty liver disease, is a more severe form of NAFLD. It is characterized by macrovesicular steatosis, balloon degeneration of liver cells, and/or inflammation that ultimately leads to scarring of the liver (i.e., fibrosis). Patients diagnosed with NASH progress to advanced stages of liver fibrosis and ultimately cirrhosis. The current treatment for end-stage cirrhotic NASH patients is liver transplantation.

 上記で定義されるNAFLDには、代謝機能障害関連脂肪性肝疾患(MASLD)、成因不明脂肪性肝疾患(Cryptogenic SLD)、及び特定成因脂肪性肝疾患(Specific aetiology SLD)が含まれるものとする。 NAFLD as defined above includes metabolic dysfunction-associated fatty liver disease (MASLD), cryptogenic fatty liver disease (Cryptogenic SLD), and specific aetiology fatty liver disease (Specific Aetiology SLD).

 代謝機能障害関連脂肪性肝疾患(MASLD)とは、脂肪肝に加えて、肥満、耐糖能異常、高血圧、高中性脂肪血症、低HDL血症に関する基準のうち1つ以上を満たす疾患をいう。前記基準としては、例えば、J Hepatol, 2023; 79 (6): 1542-56に記載のものを挙げることができる。 Metabolic dysfunction-associated fatty liver disease (MASLD) refers to a disease that, in addition to fatty liver, meets one or more of the following criteria: obesity, impaired glucose tolerance, hypertension, hypertriglyceridemia, and low HDL cholesterol. Examples of the criteria include those described in J Hepatol, 2023; 79 (6): 1542-56.

 成因不明脂肪性肝疾患(Cryptogenic SLD)とは、上記で定義されるNAFLD/NASHのうち、肥満、耐糖能異常、高血圧、高中性脂肪血症、低HDL血症に関する基準のいずれも満たさない疾患をいう。前記基準としては、例えば、J Hepatol, 2023; 79 (6): 1542-56に記載のものを挙げることができる。 Cryptogenic fatty liver disease (Cryptogenic SLD) refers to a disease among the NAFLD/NASH defined above that does not meet any of the criteria for obesity, impaired glucose tolerance, hypertension, hypertriglyceridemia, or hypo-HDL cholesterol. Examples of the criteria include those described in J Hepatol, 2023; 79 (6): 1542-56.

 特定成因脂肪性肝疾患(Specific aetiology SLD)とは、代謝機能障害を併発しない脂肪性肝疾患であって、特定の薬剤などの脂肪性肝疾患の原因がある疾患をいう。 Specific aetiology fatty liver disease (SLD) refers to fatty liver disease that is not accompanied by metabolic dysfunction and is caused by specific drugs or other factors.

 上記で定義されるNASHには、代謝機能障害関連脂肪肝炎(MASH)が含まれるものとする。したがって、上記NAFLDまたはMASLDの患者のサブセットは、MASHを起こすことがある。 As defined above, NASH includes metabolic dysfunction-associated steatohepatitis (MASH). Thus, a subset of patients with NAFLD or MASLD may develop MASH.

 代謝機能障害関連脂肪肝炎(MASH)とは、上記で定義されるMASLDのうち、組織学的にバルーン形成(balooning)やMallory-Denk体などを認めるものをいう。 Metabolic dysfunction-associated steatohepatitis (MASH) refers to the MASLD defined above in which ballooning and Mallory-Denk bodies are histologically observed.

 肝線維症は、ほとんどのタイプの慢性肝疾患において起こる、コラーゲンを含む細胞外マトリクスタンパク質の過剰な蓄積である。進行した肝線維症は肝硬変、肝不全及び門脈圧亢進をもたらし、肝移植をしばしば必要とする。 Liver fibrosis is the excessive accumulation of extracellular matrix proteins, including collagen, that occurs in most types of chronic liver disease. Advanced liver fibrosis leads to cirrhosis, liver failure, and portal hypertension, often requiring liver transplantation.

 肝線維症が進むと肝細胞の周囲が線維化で囲まれ肝硬変になる。肝硬変が進行すると、浮腫、腹水、黄疸などの症状がみられるようになり、食道胃静脈瘤などの消化管の病変を併発すると、吐血などを生じる場合もある。また、肝線維化が進むにつれて、肝がんを発生しやすくなる。 As liver fibrosis progresses, liver cells become surrounded by fibrosis, resulting in liver cirrhosis. As liver cirrhosis progresses, symptoms such as edema, ascites, and jaundice appear, and if gastrointestinal lesions such as esophageal and gastric varices occur at the same time, vomiting of blood may occur. In addition, as liver fibrosis progresses, the risk of developing liver cancer increases.

 肝細胞癌は、肝臓に由来するがんの中で最もよくみられ、通常は、肝臓に重度の瘢痕(肝硬変)がある患者に発生する。進行した肝臓線維化及び肝硬変患者は5年以内に5-30%の頻度で肝癌を発生すると報告されている。 Hepatocellular carcinoma is the most common cancer originating from the liver and usually occurs in patients with severe scarring of the liver (cirrhosis). Patients with advanced liver fibrosis and cirrhosis are reported to develop liver cancer within five years at a rate of 5-30%.

 アルコール性肝疾患(alcoholic liver disease; ALD)は、脂肪肝(alcoholic fatty liver;AFL)、アルコール性肝炎(alcoholic steatohepatitis; ASH)、重症型アルコール性肝炎(severe alcoholic hepatitis;SAH)、肝線維症(alcoholic liver fibrosis)、肝硬変と幅広い疾患を包含する。
 本発明に係る化合物は、優れたMGAT2阻害作用を有するため、ALDの治療剤及び/又は予防剤としても有用である。
Alcoholic liver disease (ALD) encompasses a wide range of diseases, including alcoholic fatty liver (AFL), alcoholic steatohepatis (ASH), severe alcoholic hepatitis (SAH), alcoholic liver fibrosis, and liver cirrhosis.
Since the compound according to the present invention has an excellent MGAT2 inhibitory effect, it is also useful as a therapeutic and/or preventive agent for ALD.

 明細書中、身体質量指数(BMI)は成人(15歳以上)の集団あるいは個人を過体重や肥満に分類する際に共通して使用されている身長体重比の単純指数を意味し、メートルで表す身長の二乗で割ったキログラムで表す体重(kg/m)である。BMIが25kg/m以上対象にも効果を発揮するが、特に、BMIが25kg/m未満である対象に有効である。 In the specification, Body Mass Index (BMI) refers to a simple index of weight to height commonly used to classify adult (15 years or older) populations or individuals as overweight or obese, and is weight in kilograms divided by height in meters squared (kg/ m2 ). It is particularly effective in subjects with a BMI of less than 25 kg/ m2 , although it is also effective in subjects with a BMI of 25 kg/ m2 or more.

 式(I)で示される化合物としては、特に以下に示される化合物、又はその製薬上許容される塩が好ましい。

Figure JPOXMLDOC01-appb-C000022
As the compound represented by formula (I), the following compounds or pharma- ceutically acceptable salts thereof are particularly preferred.
Figure JPOXMLDOC01-appb-C000022

 本発明に用いられる化合物は、特定の異性体に限定するものではなく、全ての可能な異性体(例えば、ケト-エノール異性体、イミン-エナミン異性体、ジアステレオ異性体、光学異性体、回転異性体等)、ラセミ体又はそれらの混合物を含む。 The compounds used in the present invention are not limited to specific isomers, but include all possible isomers (e.g., keto-enol isomers, imine-enamine isomers, diastereoisomers, optical isomers, rotamers, etc.), racemates, or mixtures thereof.

 本発明に用いられる化合物の一つ以上の水素、炭素及び/又は他の原子は、それぞれ水素、炭素及び/又は他の原子の同位体で置換され得る。そのような同位体の例としては、それぞれH、H、11C、13C、14C、15N、18O、17O、31P、32P、35S、18F、123I及び36Clのように、水素、炭素、窒素、酸素、リン、硫黄、フッ素、ヨウ素及び塩素が包含される。本発明に用いられる化合物は、そのような同位体で置換された化合物も包含する。該同位体で置換された化合物は、医薬品としても有用であり、本発明に用いられる化合物のすべての同位体及び放射性標識体を包含する。また該「放射性標識体」を製造するための「放射性標識化方法」も本発明に包含され、該「放射性標識体」は、代謝薬物動態研究、結合アッセイにおける研究及び/又は診断のツールとして有用である。 One or more hydrogen, carbon and/or other atoms of the compounds used in the present invention may be replaced with isotopes of hydrogen, carbon and/or other atoms, respectively. Examples of such isotopes include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine, such as 2H , 3H , 11C , 13C, 14C, 15N, 18O , 17O , 31P , 32P , 35S, 18F , 123I and 36Cl . The compounds used in the present invention also include compounds replaced with such isotopes. The isotope-substituted compounds are also useful as pharmaceuticals, and include all isotopes and radiolabeled compounds of the compounds used in the present invention. The "radiolabeling method" for producing the "radiolabel" is also included in the present invention, and the "radiolabel" is useful as a research and/or diagnostic tool in metabolism pharmacokinetic studies and binding assays.

 本発明に用いられる化合物の放射性標識体は、当該技術分野で周知の方法で調製できる。例えば、式(I)で示されるトリチウム標識化合物は、トリチウムを用いた触媒的脱ハロゲン化反応によって、式(I)で示される特定の化合物にトリチウムを導入することで調製できる。この方法は、適切な触媒、例えばPd/Cの存在下、塩基の存在下又は非存在下で、式(I)で示される化合物が適切にハロゲン置換された前駆体とトリチウムガスとを反応させることを包含する。トリチウム標識化合物を調製するための他の適切な方法は、“Isotopes in the Physical and Biomedical Sciences,Vol.1,Labeled Compounds (Part A),Chapter 6 (1987年)”を参照することができる。14C-標識化合物は、14C炭素を有する原料を用いることによって調製できる。 Radiolabeled compounds used in the present invention can be prepared by methods well known in the art. For example, tritium-labeled compounds of formula (I) can be prepared by introducing tritium into a particular compound of formula (I) by catalytic dehalogenation using tritium. This method involves reacting a precursor of a compound of formula (I) in which the halogen is appropriately substituted with tritium gas in the presence of a suitable catalyst, such as Pd/C, in the presence or absence of a base. Other suitable methods for preparing tritium-labeled compounds can be found in "Isotopes in the Physical and Biomedical Sciences, Vol. 1, Labeled Compounds (Part A), Chapter 6 (1987)". 14 C-labeled compounds can be prepared by using a raw material having 14 C carbon.

 本発明に用いられる化合物の製薬上許容される塩としては、例えば、本発明に用いられる化合物と、アルカリ金属(例えば、リチウム、ナトリウム、カリウム等)、アルカリ土類金属(例えば、カルシウム、バリウム等)、マグネシウム、遷移金属(例えば、亜鉛、鉄等)、アンモニア、有機塩基(例えば、トリメチルアミン、トリエチルアミン、ジシクロヘキシルアミン、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、メグルミン、エチレンジアミン、ピリジン、ピコリン、キノリン等)及びアミノ酸との塩、又は無機酸(例えば、塩酸、硫酸、硝酸、炭酸、臭化水素酸、リン酸、ヨウ化水素酸等)、及び有機酸(例えば、ギ酸、酢酸、プロピオン酸、トリフルオロ酢酸、クエン酸、乳酸、酒石酸、シュウ酸、マレイン酸、フマル酸、コハク酸、マンデル酸、グルタル酸、リンゴ酸、安息香酸、フタル酸、アスコルビン酸、ベンゼンスルホン酸、p-トルエンスルホン酸、メタンスルホン酸、エタンスルホン酸、トリフルオロ酢酸等)との塩が挙げられる。これらの塩は、通常行われる方法によって形成させることができる。 Pharmaceutically acceptable salts of the compounds used in the present invention include, for example, salts of the compounds used in the present invention with alkali metals (e.g., lithium, sodium, potassium, etc.), alkaline earth metals (e.g., calcium, barium, etc.), magnesium, transition metals (e.g., zinc, iron, etc.), ammonia, organic bases (e.g., trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, meglumine, ethylenediamine, pyridine, pyridine, etc.), and the like. Examples of the salts include salts with inorganic acids (e.g., hydrochloric acid, sulfuric acid, nitric acid, carbonic acid, hydrobromic acid, phosphoric acid, hydroiodic acid, etc.) and amino acids, and salts with inorganic acids (e.g., hydrochloric acid, sulfuric acid, nitric acid, carbonic acid, hydrobromic acid, phosphoric acid, hydroiodic acid, etc.) and organic acids (e.g., formic acid, acetic acid, propionic acid, trifluoroacetic acid, citric acid, lactic acid, tartaric acid, oxalic acid, maleic acid, fumaric acid, succinic acid, mandelic acid, glutaric acid, malic acid, benzoic acid, phthalic acid, ascorbic acid, benzenesulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, trifluoroacetic acid, etc.). These salts can be formed by conventional methods.

 本発明に用いられる化合物又はその製薬上許容される塩は、溶媒和物(例えば、水和物等)、共結晶及び/又は結晶多形を形成する場合があり、本発明はそのような各種の溶媒和物、共結晶及び結晶多形も包含する。「溶媒和物」は、本発明に用いられる化合物に対し、任意の数の溶媒分子(例えば、水分子等)と配位していてもよい。本発明に用いられる化合物又はその製薬上許容される塩を、大気中に放置することにより、水分を吸収し、吸着水が付着する場合や、水和物を形成する場合がある。また、本発明に用いられる化合物又はその製薬上許容される塩を、再結晶することで結晶多形を形成する場合がある。「共結晶」は、本発明に用いられる化合物又は塩とカウンター分子が同一結晶格子内に存在することを意味し、任意の数のカウンター分子を含んでいても良い。 The compound or its pharma- ceutically acceptable salt used in the present invention may form a solvate (e.g., hydrate, etc.), co-crystal, and/or crystalline polymorph, and the present invention includes such various solvates, co-crystals, and crystalline polymorphs. A "solvate" may be coordinated with any number of solvent molecules (e.g., water molecules, etc.) to the compound used in the present invention. When the compound or its pharma- ceutically acceptable salt used in the present invention is left in the air, it may absorb moisture, and adsorbed water may adhere to it, or a hydrate may be formed. In addition, the compound or its pharma- ceutically acceptable salt used in the present invention may form a crystalline polymorph by recrystallizing it. A "co-crystal" means that the compound or salt used in the present invention and a counter molecule are present in the same crystal lattice, and may contain any number of counter molecules.

 本発明に用いられる化合物又はその製薬上許容される塩は、プロドラッグを形成する場合があり、本発明はそのような各種のプロドラッグも包含する。プロドラッグは、化学的又は代謝的に分解できる基を有する本発明化合物の誘導体であり、加溶媒分解により又は生理学的条件下でインビボにおいて薬学的に活性な本発明化合物となる化合物である。プロドラッグは、生体内における生理条件下で酵素的に酸化、還元、加水分解等を受けて式(I)で示される化合物に変換される化合物、胃酸等により加水分解されて式(I)で示される化合物に変換される化合物等を包含する。適当なプロドラッグ誘導体を選択する方法及び製造する方法は、例えば “Design of Prodrugs, Elsevier, Amsterdam, 1985”に記載されている。プロドラッグは、それ自身が活性を有する場合がある。 The compounds or pharma- ceutically acceptable salts thereof used in the present invention may form prodrugs, and the present invention also includes such various prodrugs. Prodrugs are derivatives of the compounds of the present invention having a group that can be decomposed chemically or metabolically, and are compounds that become the pharma- ceutically active compounds of the present invention in vivo by solvolysis or under physiological conditions. Prodrugs include compounds that are converted to the compounds of formula (I) by enzymatic oxidation, reduction, hydrolysis, etc. under physiological conditions in the living body, and compounds that are converted to the compounds of formula (I) by hydrolysis by gastric acid, etc. Methods for selecting and producing appropriate prodrug derivatives are described, for example, in "Design of Prodrugs, Elsevier, Amsterdam, 1985". Prodrugs may themselves have activity.

 本発明に係る化合物は、優れたMGAT2阻害作用を有するため、脂肪性肝疾患、特に非アルコール性脂肪肝疾患の治療剤及び/又は予防剤として有用である。非アルコール性脂肪肝疾患は、非アルコール性脂肪肝炎(NASH)に限定されず、NASHによって引き起こされる肝線維症、肝硬変又は肝細胞癌(HCC)の治療剤及び/又は予防剤としても有用である。
 さらに本発明に係る化合物は、医薬としての有用性を備えており、好ましくは、下記のいずれか、又は複数の優れた特徴を有している。
a)CYP酵素(例えば、CYP1A2、CYP2C9、CYP2C19、CYP2D6、CYP3A4等)に対する阻害作用が弱い。
b)高いバイオアベイラビリティー、適度なクリアランス等良好な薬物動態を示す。
c)代謝安定性が高い。
d)CYP酵素(例えば、CYP3A4)に対し、本明細書に記載する測定条件の濃度範囲内で不可逆的阻害作用を示さない。
e)変異原性を有さない。
f)心血管系のリスクが低い。
g)高い溶解性を示す。
h)止血異常、肝障害腎障害及び胚致死性の少なくとも1つの副作用を伴わない。
The compound according to the present invention has an excellent MGAT2 inhibitory effect, and is therefore useful as a therapeutic and/or preventive agent for fatty liver disease, particularly non-alcoholic fatty liver disease. Non-alcoholic fatty liver disease is not limited to non-alcoholic steatohepatitis (NASH), and is also useful as a therapeutic and/or preventive agent for liver fibrosis, liver cirrhosis, or hepatocellular carcinoma (HCC) caused by NASH.
Furthermore, the compound according to the present invention has pharmaceutical utility and preferably has one or more of the following excellent characteristics:
a) It has a weak inhibitory effect on CYP enzymes (e.g., CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, etc.).
b) It exhibits favorable pharmacokinetics, such as high bioavailability and moderate clearance.
c) High metabolic stability.
d) It does not exhibit irreversible inhibitory effects on CYP enzymes (e.g., CYP3A4) within the concentration range of the measurement conditions described herein.
e) It is not mutagenic.
f) Lower cardiovascular risk.
g) It exhibits high solubility.
h) It is free of at least one of the side effects of hemostatic abnormalities, liver damage, renal damage, and embryonic lethality.

 本発明の医薬組成物は、経口的、非経口的のいずれの方法でも投与することができる。非経口投与の方法としては、経皮、皮下、静脈内、動脈内、筋肉内、腹腔内、経粘膜、吸入、経鼻、点眼、点耳、膣内投与等が挙げられる。 The pharmaceutical composition of the present invention can be administered either orally or parenterally. Parenteral administration methods include transdermal, subcutaneous, intravenous, intraarterial, intramuscular, intraperitoneal, transmucosal, inhalation, nasal, ophthalmic, otic, and vaginal administration.

 経口投与の場合は常法に従って、内用固形製剤(例えば、錠剤、散剤、顆粒剤、カプセル剤、丸剤、フィルム剤等)、内用液剤(例えば、懸濁剤、乳剤、エリキシル剤、シロップ剤、リモナーデ剤、酒精剤、芳香水剤、エキス剤、煎剤、チンキ剤等)等の通常用いられるいずれの剤型に調製して投与すればよい。錠剤は、糖衣錠、フィルムコーティング錠、腸溶性コーティング錠、徐放錠、トローチ錠、舌下錠、バッカル錠、チュアブル錠又は口腔内崩壊錠であってもよく、散剤及び顆粒剤はドライシロップであってもよく、カプセル剤は、ソフトカプセル剤、マイクロカプセル剤又は徐放性カプセル剤であってもよい。  In the case of oral administration, the drug may be prepared and administered in any of the commonly used dosage forms, such as solid preparations for internal use (e.g., tablets, powders, granules, capsules, pills, films, etc.) and liquid preparations for internal use (e.g., suspensions, emulsions, elixirs, syrups, lemonades, spirits, aromatic preparations, extracts, decoctions, tinctures, etc.), in accordance with the usual methods. Tablets may be sugar-coated tablets, film-coated tablets, enteric-coated tablets, sustained-release tablets, troches, sublingual tablets, buccal tablets, chewable tablets, or orally disintegrating tablets, powders and granules may be dry syrups, and capsules may be soft capsules, microcapsules, or sustained-release capsules.

 非経口投与の場合は、注射剤、点滴剤、外用剤(例えば、点眼剤、点鼻剤、点耳剤、エアゾール剤、吸入剤、ローション剤、注入剤、塗布剤、含嗽剤、浣腸剤、軟膏剤、硬膏剤、ゼリー剤、クリーム剤、貼付剤、パップ剤、外用散剤、坐剤等)等の通常用いられるいずれの剤型でも好適に投与することができる。注射剤は、O/W、W/O、O/W/O、W/O/W型等のエマルジョンであってもよい。 In the case of parenteral administration, any of the commonly used dosage forms such as injections, drops, topical preparations (e.g., eye drops, nasal drops, ear drops, aerosols, inhalants, lotions, injections, liniments, mouthwashes, enemas, ointments, plasters, jellies, creams, patches, poultices, topical powders, suppositories, etc.) can be suitably administered. Injections may be emulsions such as O/W, W/O, O/W/O, and W/O/W types.

 本発明に係る化合物の有効量にその剤型に適した賦形剤、結合剤、崩壊剤、滑沢剤等の各種医薬用添加剤を必要に応じて混合し、医薬組成物とすることができる。さらに、該医薬組成物は、本発明化合物の有効量、剤型及び/又は各種医薬用添加剤を適宜変更することにより、小児用、高齢者用、重症患者用又は手術用の医薬組成物とすることもできる。例えば、小児用医薬組成物は、新生児(出生後4週未満)、乳児(出生後4週~1歳未満)、幼児(1歳以上7歳未満)、小児(7歳以上15歳未満)若しくは15歳~18歳の患者に投与されうる。例えば、高齢者用医薬組成物は、65歳以上の患者に投与されうる。 A pharmaceutical composition can be prepared by mixing an effective amount of the compound of the present invention with various pharmaceutical additives suitable for the dosage form, such as excipients, binders, disintegrants, lubricants, etc., as necessary. Furthermore, the pharmaceutical composition can be prepared as a pharmaceutical composition for children, elderly people, seriously ill patients, or surgical patients by appropriately changing the effective amount of the compound of the present invention, the dosage form, and/or various pharmaceutical additives. For example, a pharmaceutical composition for children can be administered to newborns (less than 4 weeks after birth), infants (4 weeks to less than 1 year after birth), toddlers (1 year to less than 7 years), children (7 years to less than 15 years), or patients aged 15 to 18 years. For example, a pharmaceutical composition for the elderly can be administered to patients aged 65 years or older.

 本発明に係る化合物の投与量は、患者の年齢、体重、疾病の種類や程度、投与経路等を考慮した上で設定することが望ましいが、通常、経口投与の場合、成人1日あたり約0.5mg~100mg、好ましくは、約1mg~50mg、さらに好ましくは、約3mg~30mgを、要すれば分割して投与してもよい。また、小児の場合は、1日あたり約0.5~100mgを投与すればよい。また、非経口投与の場合、成人1日あたり約0.1mg~100mg、好ましくは、約0.5mg~50mg、約1mg~30mgを投与する。これを1日1回~数回に分けて投与すれば良い。 The dosage of the compound according to the present invention is preferably determined taking into consideration the age and weight of the patient, the type and severity of the disease, the route of administration, etc., but typically, in the case of oral administration, about 0.5 mg to 100 mg, preferably about 1 mg to 50 mg, and more preferably about 3 mg to 30 mg per day for adults, which may be administered in divided doses if necessary. In the case of children, about 0.5 to 100 mg may be administered per day. In the case of parenteral administration, about 0.1 mg to 100 mg, preferably about 0.5 mg to 50 mg, or about 1 mg to 30 mg, is administered per day for adults. This may be administered once or in divided doses several times a day.

 本発明の式:

Figure JPOXMLDOC01-appb-C000023

(式中、各記号は前記と同意義である。)で示される化合物又はその製薬上許容される塩は、該化合物の作用の増強又は該化合物の投与量の低減等を目的として、抗肥満作用を有する薬剤、血糖値をコントロールするための薬剤、血中のコレステロール及び/又はトリグリセライドをコントロールするための薬剤、及び血圧をコントロールするための薬剤からなる群から選択される少なくとも1つの薬剤と組み合わせて用いることができる。 The formula of the present invention:
Figure JPOXMLDOC01-appb-C000023

(wherein each symbol has the same meaning as defined above) or a pharma- ceutically acceptable salt thereof can be used in combination with at least one drug selected from the group consisting of drugs having anti-obesity action, drugs for controlling blood glucose level, drugs for controlling blood cholesterol and/or triglyceride, and drugs for controlling blood pressure, for the purpose of enhancing the action of the compound or reducing the dose of the compound, etc.

 「抗肥満作用を有する薬剤、血糖値をコントロールするための薬剤、血中のコレステロール及び/又はトリグリセライドをコントロールするための薬剤、及び血圧をコントロールするための薬剤からなる群から選択される少なくとも1つの薬剤」としては、市販又は開発中のものには限定されないが、市販又は開発中のものとしては、オルリスタット、セチリスタット、フェンテルミン、マジンドール、ベンズフェタミン、アンフェプラモン、メタンフェタミン、フェンテルミン塩酸塩/トピラマート、ナルトレキソン塩酸塩/ブプロピオン塩酸塩、リラグルチド、セマグルチド、セトメラノチド(setmelanotide、RM-493)、メトレレプチン、トピラメート、ナルトレキソン、ブプロピオン、アカルボース、ボグリボース、ミグリトール、イプラグリフロジン、ダパグリフロジン、レモグリフロジン、KGT-1075、ルセオグリフロジン、トホグリフロジン、カナグリフロジン、エンパグリフロジン、エルツグリフロジン、ベクサグリフロジン(bexagliflozin)、エナボグリフロジン(enavogliflozin)、ヘナグリフロジン(henagliflozin)、ジャナグリフロジン(janagliflozin)、ソタグリフロジン、インスリン アスパルト、インスリン リスプロ、インスリン グルリジン、生合成ヒト中性インスリン、ヒトインスリン、生合成ヒトイソフェンインスリン、ヒトイソフェンインスリン、中間型インスロンリスプロ、インスリン デテミル、インスリン グラルギン、インスリン デグルデク、グリベンクラミド、グリクラジド、グリメピリド、グリピジド(glipizide)、グリキドン(gliquidone)、ナテグリニド、ミチグリニドカルシウム水和物、レパグリニド、メトホルミン塩酸塩、ブホルミン塩酸塩、ピオグリタゾン塩酸塩、ロシグリタゾン(rosiglitazone)、ロベグリタゾン(lobeglitazone)、シタグリプチンリン酸塩、ビルダグリプチン、アログリプチン安息香酸塩、リナグリプチン、テネリグリプチン臭化水素酸塩、アナグリプチン、サキサグリプチン、トレラグリプチンコハク酸塩、オマリグリプチン、ゲミグリプチン、エボグリプチン(evogliptin)、リキシセナチド、エキセナチド、デュラグルチド、チルゼパチド、イメグリミン、シタグリプチンリン酸塩/イプラグリフロジン、ピオグリタゾン塩酸塩/メトホルミン、ピオグリタゾン塩酸塩/グリメピリド、テネリグリプチン臭化水素酸塩/カナグリフロジン、アログリプチン安息香酸塩/ピオグリタゾン塩酸塩、アログリプチン安息香酸塩/メトホルミン塩酸塩、ビルダグリプチン/メトホルミン塩酸塩、ミチグリニドカルシウム/ボグリボース、プラバスタチン、シンバスタチン、フルバスタチン、アトルバスタチン、ピタバスタチン、ロスバスタチン、ロバスタチン(lovastatin)、クリノフィブラート、クロフィブラート、ベザフィブラート、フェノフィブラート、シプロフィブラート(ciprofibrate)、ペマフィブラート、ジェムフィブロジル、コレスチミド、コレスチラミン、コレセベラム塩酸塩、コレスチポール(colestipol)、エゼチミブ、ブロブコール、ニコモール、トコフェロールニコチン酸エステル、ニセリトロール、イコサペント酸エチル、ω-3脂肪酸エチル、エボロクマブ、アリロクマブ、ニフェジピン、アムロジピン、エホニジピン、シルニジピン、ニカルジピン、ニソルジピン、ニトレンジピン、ニルバジピン、バルニジピン、フェロジピン、ベンジピン、マニジピン、アゼルニジピン、アラニジピン、ジルチアゼム、トルクロルメチアジド、ベンチルヒドロクロロチアジド、ヒドロクロロチアジド、メイクラン、インバダミド、トリパミド、メフルシド、フロセミド、トリアムテレン、スピノロラクトン、エプレレノン、トルバプタン、トラセミド、ヒドロクロロチアジド(hydrochlorothiazide)、ブメタニド(bumetanide)、クロルタリドン(chlortalidone)、イソソルビド、メトラゾン、ロサルタン、カンデサルタン、バルサルタン、テルミサルタン、オルメサルタン、イルベサルタン、アジルサルタン、カプトプリル、エナラプリル、アラセプリル、デラプリル、シラザプリル、リシノプリル、ベナゼプリル、イミダプリル、テモカプリル、キナプリル、トランドラプリル、ペリンドプリルエルブミン、ウラピジル、テラゾシン、ブラゾシン、ドキサゾシン、ブナゾシン、アテノロール、ビソブロロール、メトプロロール、アセブトロール、セリプロロール、プロプラノロール、ナドロール、ニプラジロール、カルテオロール、ピンドロール、ネビボロール、カルベジロール、ラベタロール、ソタロール、ランジオロール、アロチノロール、アモスラロール、アロチノロール、カルベジロール、ラベタロール、ベバントロール、クロニジン、グアナベンズ、メチルドパ、レセルピン、ヒドララジン、ニトロプルシド、アリスキレン、カリジノゲナーゼ、アルプロスタジルアルファデクス、ジヒドロエルゴトキシン、ドキサゾシン、ウラピジル、ヒドララジン(hydralazine)、プラゾシン、モクソニジン(moxonidine)、グアンファシン、リルメニジン(rilmenidine)、アムロジピン/アトロバスタチン、ロサルタン/ヒドロクロロチアジド、バルサルタン/ヒドロクロロチアジド、カンデサルタン/ヒドロクロロチアジド、テルミサルタン/ヒドロクロロチアジド、イルベサルタン/トリクロルチアジド、バルサルタン/アムロジピン、オルメサルタン/アゼルニジピン、カンデサルタン/アムロジピン、テルミサルタン/アムロジピン、イルベサルタン/アムロジピン、バルサルタン/シルニジピン、アジルサルタン/アムロジピン、チルゼパチド、SCO-094、efinopegdutide(HM12525A)、BI-456906、DD01、NN-9423、LY-3437943、ダヌグリプロン(danuglipron)、PF07081532、LY-3502970、RGT-075、エフペグレナチド(efpeglenatide)、エキセナチド、ノイグルチド(noiiglutide)、GMA105、HM-15136、ノリグリコペプチド(noliglycopeptide、SHR-20004)、carbetocin(LV-101)、PYY-1562(NNC0165-1562)、PYY-1875(NNC0165-1875)、コタデュチド(cotadutide)/AM833(NN9838、NNC-01740833)、コタデュチド(cotadutide、MEDI0382)、LY3305677、ペガパモデュチド(pegapamodutide、LY2944876、OPK88003)、NGM395、YH34160、CT-388、CT-868、SCO-267、SCO-792、ジアゾキシド、テソフェンシン(tesofensine)、ナモデノソン(namodenoson)、ERX1000、AMG133、ASC41、Xla1、HDV Biotin、EMP16、メトプロロール(metoprolol)/テソフェンシン(tesofensine)、RZL-012、CB4211、BI1356225、AMG171、NO-13065、バルドキソロンメチル、HSG4112、YHC2129、YHC2134、KTX-0200、オベチコール酸、cilofexor(GS-9674)、tropifexor(LJN452)、EDP-305、EYP-001、レスメティロム(resmetirom)、VK-2809、セニクリビロク(cenicriviroc)、サログリタザール(saroglitazar)、ラニフィブラノール(lanifibranor)、セロンセルチブ、PF-06835919、ペグベルフェルミン(pegbelfermin)、エフルグキシフェルミン(efrugxifermin)、アルダフェルミン(aldafermin)、アラムコル(aramcol)、MK-3655、MSDC-0602K、ベラペクチン(belapectin)、firsocostat(GS-0976)、PF-05221304、エルボガスト(ervogastat)、ION-224、AXA-1125、HU-6、MET-409、MET-642、TERN-101、TERN-501、LPCN-1144、デニファンステート(denifanstate)、フルキシフェルミン(fruxifermin)、レロンリマブ(leronlimab)、ペゴザフェルミン(pegozafermin)、レンコフィルスタット(rencofilstat)、レトルチド(retatrutide)、ティペルカスト(tipelukast)、S-237648、S-723595等が挙げられる。更にこれら薬剤が塩を形成していない場合は、それらの製薬上許容される塩も含まれ、塩を形成している場合は、それ以外の製薬上許容される塩を形成していてもよい。
 好ましくは、オルリスタット、セチリスタット、フェンテルミン、マジンドール、ベンズフェタミン、アンフェプラモン、メタンフェタミン、フェンテルミン塩酸塩/トピラマート、ナルトレキソン塩酸塩/ブプロピオン塩酸塩、リラグルチド、セマグルチド、セトメラノチド(setmelanotide、RM-493)、メトレレプチン、トピラメート、ナルトレキソン、ブプロピオン、チルゼパチド、SCO-094、efinopegdutide(HM12525A)、BI-456906、DD01、NN-9423、LY-3437943、ダヌグリプロン(danuglipron)、PF07081532、LY-3502970、RGT-075、エフペグレナチド(efpeglenatide)、エキセナチド、ノイグルチド(noiiglutide)、GMA105、HM-15136、ノリグリコペプチド(noliglycopeptide、SHR-20004)、carbetocin(LV-101)、PYY-1562(NNC0165-1562)、PYY-1875(NNC0165-1875)、コタデュチド(cotadutide)/AM833(NN9838、NNC-01740833)、コタデュチド(cotadutide、MEDI0382)、LY3305677、ペガパモデュチド(pegapamodutide、LY2944876、OPK88003)、NGM395、YH34160、CT-388、CT-868、SCO-267、SCO-792、ジアゾキシド、テソフェンシン(tesofensine)、ナモデノソン(namodenoson)、ERX1000、AMG133、ASC41、Xla1、HDV Biotin、EMP16、メトプロロール(metoprolol)/テソフェンシン(tesofensine)、RZL-012、CB4211、BI1356225、AMG171、NO-13065、バルドキソロンメチル、HSG4112、YHC2129、YHC2134、KTX-0200、S-237648、S-723595等が挙げられる。
 更に好ましくは、リラグルチド、セマグルチド、チルゼパチド、BI-456906、ダヌグリプロン、PF07081532、LY-3502970、RGT-075、S-237648又はS-723595等が挙げられる。
The "at least one drug selected from the group consisting of drugs having an anti-obesity effect, drugs for controlling blood glucose levels, drugs for controlling blood cholesterol and/or triglycerides, and drugs for controlling blood pressure" is not limited to those on the market or under development, but examples of those on the market or under development include orlistat, cetilistat, phentermine, mazindol, benzphetamine, amfepramone, methamphetamine, phentermine hydrochloride/topiramate, naltrexone hydrochloride/bupropion hydrochloride, liraglutide, semaglutide, setomelanotide (s etmelanotide, RM-493), metreleptin, topiramate, naltrexone, bupropion, acarbose, voglibose, miglitol, ipragliflozin, dapagliflozin, remogliflozin, KGT-1075, luseogliflozin, tofogliflozin, canagliflozin, empagliflozin, ertugliflozin, bexagliflozin, enavogliflozin, henagliflozin, janagliflozin, sotagliflozin, insulin Aspart, insulin lispro, insulin glulisine, biosynthetic human neutral insulin, human insulin, biosynthetic human isophane insulin, human isophane insulin, intermediate-acting insulin lispro, insulin detemir, insulin glargine, insulin Degludec, glibenclamide, gliclazide, glimepiride, glipizide, gliquidone, nateglinide, mitiglinide calcium hydrate, repaglinide, metformin hydrochloride, buformin hydrochloride, pioglitazone hydrochloride, rosiglitazone, lobeglitazone, sitagliptin phosphate, vildagliptin, alogliptin benzoate, linagliptin, teneligliptin hydrobromide, anagliptin, saxagliptin, trelagliptin succinate, omarigliptin, gemigliptin, evogliptin in), lixisenatide, exenatide, dulaglutide, tirzepatide, imeglimin, sitagliptin phosphate/ipragliflozin, pioglitazone hydrochloride/metformin, pioglitazone hydrochloride/glimepiride, teneligliptin hydrobromide/canagliflozin, alogliptin benzoate/pioglitazone hydrochloride, alogliptin benzoate/metformin hydrochloride, vildagliptin/metformin hydrochloride, mitiglinide calcium/voglibose, pravastatin, simvastatin, fluvastatin, atorvastatin, pitavastatin, rosuvastatin, lovastatin, clinofibrate, clofibrate acetaminophen, bezafibrate, fenofibrate, ciprofibrate, pemafibrate, gemfibrozil, colestimide, cholestyramine, colesevelam hydrochloride, colestipol, ezetimibe, brobucol, nicomol, tocopherol nicotinate, niceritrol, ethyl icosapentate, omega-3 fatty acid ethyl ester, evolocumab, alirocumab, nifedipine, amlodipine, efonidipine, cilnidipine, nicardipine, nisoldipine, nitrendipine, nilvadipine, barnidipine, felodipine, bendipine, manidipine, azelnidipine, aranidipine, ziltipine Azem, tolchlormethiazide, benzylhydrochlorothiazide, hydrochlorothiazide, meicran, invadamide, tripamide, mefruside, furosemide, triamterene, spinolactone, eplerenone, tolvaptan, torasemide, hydrochlorothiazide, bumetanide, chlortalidone, isosorbide, metolazone, losartan, candesartan, valsartan, telmisartan, olmesartan, irbesartan, azilsartan, captopril, enalapril, alacepril, delapril, cilazapril, lisinopril, bena Zepril, imidapril, temocapril, quinapril, trandolapril, perindopril erbumine, urapidil, terazosin, brazosin, doxazosin, bunazosin, atenolol, bisobrolol, metoprolol, acebutolol, celiprolol, propranolol, nadolol, nipradilol, carteolol, pindolol, nebivolol, carvedilol, labetalol, sotalol, landiolol, arotinolol, amosulalol, arotinolol, carvedilol, labetalol, bevantolol, clonidine, guanabenz, methyldopa, reserpine, hydralazine, nitroprusside, aliskiren, kalizinogena azepam, alprostadil alfadex, dihydroergotoxine, doxazosin, urapidil, hydralazine, prazosin, moxonidine, guanfacine, rilmenidine, amlodipine/atorvastatin, losartan/hydrochlorothiazide, valsartan/hydrochlorothiazide, candesartan/hydrochlorothiazide, telmisartan/hydrochlorothiazide, irbesartan/trichlorthiazide, valsartan/amlodipine, olmesartan/azelnidipine, candesartan/amlodipine, telmisartan/amlodipine, irbesartan /Amlodipine, Valsartan/Cilnidipine, Azilsartan/Amlodipine, Tirzepatide, SCO-094, Efinopegdutide (HM12525A), BI-456906, DD01, NN-9423, LY-3437943, Danuglipron, PF07081532, LY -3502970, RGT-075, efpeglenatide, exenatide, noiglutide, GMA105, HM-15136, noliglycopeptide (SHR-20004), carbetocin (LV-101), PYY-1 562 (NNC0165-1562), PYY-1875 (NNC0165-1875), cotadutide/AM833 (NN9838, NNC-01740833), cotadutide (cotadutide, MEDI0382), LY3305677, pegapamodutide ide, LY2944876, OPK88003), NGM395, YH34160, CT-388, CT-868, SCO-267, SCO-792, diazoxide, tesofensine, namodenoson, ERX1000, AMG133, ASC41, Xla1, HDV Biotin, EMP16, metoprolol/tesofensine, RZL-012, CB4211, BI1356225, AMG171, NO-13065, bardoxolone methyl, HSG4112, YHC2129, YHC2134, KTX-0200, obeticholic acid, cilofexor (GS-9674), tropifexor (LJN452) , EDP-305, EYP-001, resmetirom, VK-2809, cenicriviroc, saroglitazar, lanifibranor, selonsertib, PF-06835919, pegbelfermin, efrugxifermin, Aldafermin, aramcol, MK-3655, MSDC-0602K, belapectin, firsocostat (GS-0976), PF-05221304, ervogastat, ION-224, AXA-1125, HU-6, MET-409, MET-642, TERN-101, TERN-501, LPC Examples of such drugs include N-1144, denifanstate, fluxifermin, leronlimab, pegozafermin, rencofilstat, retatrutide, tipelukast, S-237648, and S-723595. Furthermore, when these drugs do not form a salt, their pharma- ceutically acceptable salts are also included, and when they form a salt, they may form other pharma- ceutically acceptable salts.
Preferably, orlistat, cetilistat, phentermine, mazindol, benzphetamine, amfepramone, methamphetamine, phentermine hydrochloride/topiramate, naltrexone hydrochloride/bupropion hydrochloride, liraglutide, semaglutide, setmelanotide (RM-493), metreleptin, topiramate, naltrexone, bupropion, tirze Patide, SCO-094, efinopegdutide (HM12525A), BI-456906, DD01, NN-9423, LY-3437943, danuglipron, PF07081532, LY-3502970, RGT-075, efpeglenatide, exenatide, neuglutide, GMA105, H M-15136, noliglycopeptide (SHR-20004), carbetocin (LV-101), PYY-1562 (NNC0165-1562), PYY-1875 (NNC0165-1875), cotadutide/AM833 (NN9838, NNC-01740833), cotadutide (MEDI0 382), LY3305677, pegapamodutide (LY2944876, OPK88003), NGM395, YH34160, CT-388, CT-868, SCO-267, SCO-792, diazoxide, tesofensine, namodenoson, ERX1000, AMG133, ASC41, Xla1, HDV Biotin, EMP16, metoprolol/tesofensine, RZL-012, CB4211, BI1356225, AMG171, NO-13065, bardoxolone methyl, HSG4112, YHC2129, YHC2134, KTX-0200, S-237648, S-723595, and the like.
More preferred examples include liraglutide, semaglutide, tirzepatide, BI-456906, danugliplon, PF07081532, LY-3502970, RGT-075, S-237648, and S-723595.

 本発明に係る化合物と上記の組み合わせて用いられる併用薬剤の投与時期は限定されず、これらを投与対象に対し、同時に又は連続的に投与してもよいし、時間差をおいて投与してもよい。さらに、本発明化合物と併用薬剤とは、それぞれの活性成分を含む2種類以上の製剤として投与されてもよいし、それらの活性成分を含む単一の製剤として投与されてもよい。 The administration timing of the compound of the present invention and the concomitant drug used in combination as described above is not limited, and they may be administered to the subject simultaneously or consecutively, or may be administered with a time lag. Furthermore, the compound of the present invention and the concomitant drug may be administered as two or more types of preparations containing the respective active ingredients, or as a single preparation containing those active ingredients.

 併用薬剤の投与量は、臨床上用いられている用量を基準として適宜選択することができる。また、本発明化合物と併用薬剤の配合比は、投与対象、投与ルート、対象疾患、症状、組み合わせ等により適宜選択することができる。例えば、投与対象がヒトである場合、本発明化合物1重量部に対し、併用薬剤を0.01~100重量部用いればよい。 The dosage of the concomitant drug can be appropriately selected based on the dose used clinically. The mixing ratio of the compound of the present invention and the concomitant drug can be appropriately selected depending on the subject, administration route, target disease, symptoms, combination, etc. For example, when the subject is a human, 0.01 to 100 parts by weight of the concomitant drug may be used per 1 part by weight of the compound of the present invention.

 以下に本発明の実施例及び試験例を挙げて本発明をさらに詳しく説明するが、本発明はこれらにより限定されるものではない。また、本発明の範囲を逸脱しない範囲で変化させてもよい。尚、以下の実施例及び比較例において示された化合物名は必ずしもIUPAC命名法に従うものではない。 The present invention will be explained in more detail below with reference to examples and test examples, but the present invention is not limited to these. Furthermore, changes may be made within the scope of the present invention. The compound names given in the following examples and comparative examples do not necessarily conform to the IUPAC nomenclature.

(合成例1)化合物II-121

Figure JPOXMLDOC01-appb-C000024

 市販化合物から、特許文献2に記載の方法に従い、化合物II-121を合成した。
1H-NMR(CDCl3) δ: 2.09-2.18(m, 2H), 2.29-2.36(m, 2H), 3.03(d, J=16.3 Hz, 1H), 3.21(s, 3H), 3.31(d, J=16.3 Hz, 1H), 3.99-4.35(m, 10H), 5.92(s, 1H), 6.40(d, J=2.4 Hz, 1H), 6.60(dd, J=8.8, 2.4 Hz, 1H), 7.41(d, J=8.8 Hz, 1H), 9.56(s, 1H). (Synthesis Example 1) Compound II-121
Figure JPOXMLDOC01-appb-C000024

Compound II-121 was synthesized from a commercially available compound according to the method described in Patent Document 2.
1 H-NMR(CDCl 3 ) δ: 2.09-2.18(m, 2H), 2.29-2.36(m, 2H), 3.03(d, J=16.3 Hz, 1H), 3.21(s, 3H), 3.31(d, J=16.3 Hz, 1H), 3.99-4.35(m, 10H), 5.92(s, 1H), 6.40(d, J=2.4 Hz, 1H), 6.60(dd, J=8.8, 2.4 Hz, 1H), 7.41(d, J=8.8 Hz, 1H), 9.56(s, 1H).

 市販化合物から、特許文献2~4に記載の方法に従い、以下の化合物を合成した。

Figure JPOXMLDOC01-appb-T000025
The following compounds were synthesized from commercially available compounds according to the methods described in Patent Documents 2 to 4.
Figure JPOXMLDOC01-appb-T000025

Figure JPOXMLDOC01-appb-T000026
Figure JPOXMLDOC01-appb-T000026

 各化合物の物理データを以下に示す。
 表中に「MS」とあるのは、LC/MSでの質量(M+H)を表す。
[測定条件A]
カラム:ACQUITY UPLC(登録商標)BEH C18 (1.7μm i.d.2.1x50mm)(Waters)
流速:0.8 mL/分
UV検出波長:254nm
移動相:[A]は0.1%ギ酸含有水溶液、[B]は0.1%ギ酸含有アセトニトリル溶液
3.5分間で5%-100%溶媒[B]のリニアグラジエントを行った後、0.5分間、100%溶媒[B]を維持した。
[測定条件B]
カラム:Shim-pack XR-ODS (2.2μm i.d.50x3.0mm) (Shimadzu)
流速:1.6 mL/分
UV検出波長:254nm;
移動相:[A]は0.1%ギ酸含有水溶液、[B]は0.1%ギ酸含有アセトニトリル溶液
グラジェント:3分間で10%-100%溶媒[B]のリニアグラジエントを行い、0.5分間、100%溶媒[B]を維持した。
[測定条件C]
カラム:ACQUITY UPLC(登録商標)BEH C18 (1.7μm i.d.2.1x50mm)(Waters)
流速:0.55 mL/分
UV検出波長:254nm
移動相:[A]は0.1%ギ酸含有水溶液、[B]は0.1%ギ酸含有アセトニトリル溶液
グラジェント:3分間で5%-100%溶媒[B]のリニアグラジエントを行い、0.5分間、100%溶媒[B]を維持した。

Figure JPOXMLDOC01-appb-T000027
The physical data of each compound is shown below.
In the table, "MS" indicates the mass (M+H) measured by LC/MS.
[Measurement condition A]
Column: ACQUITY UPLC (registered trademark) BEH C18 (1.7 μm id2.1x50 mm) (Waters)
Flow rate: 0.8 mL/min UV detection wavelength: 254 nm
Mobile phase: [A] was an aqueous solution containing 0.1% formic acid; [B] was an acetonitrile solution containing 0.1% formic acid. A linear gradient of 5%-100% solvent [B] was performed over 3.5 minutes, followed by maintaining 100% solvent [B] for 0.5 minutes.
[Measurement condition B]
Column: Shim-pack XR-ODS (2.2 μm id50x3.0 mm) (Shimadzu)
Flow rate: 1.6 mL/min UV detection wavelength: 254 nm;
Mobile phase: [A] is an aqueous solution containing 0.1% formic acid; [B] is an acetonitrile solution containing 0.1% formic acid. Gradient: A linear gradient of 10%-100% solvent [B] was performed over 3 minutes, and 100% solvent [B] was maintained for 0.5 minutes.
[Measurement conditions C]
Column: ACQUITY UPLC (registered trademark) BEH C18 (1.7 μm id2.1x50 mm) (Waters)
Flow rate: 0.55 mL/min UV detection wavelength: 254 nm
Mobile phase: [A] is an aqueous solution containing 0.1% formic acid; [B] is an acetonitrile solution containing 0.1% formic acid. Gradient: A linear gradient of 5%-100% solvent [B] was performed over 3 minutes, and 100% solvent [B] was maintained for 0.5 minutes.
Figure JPOXMLDOC01-appb-T000027

 各実施例で得られたNMR分析は300MHzで行い、DMSO-d、CDClを用いて測定した。

Figure JPOXMLDOC01-appb-T000028
The NMR analysis obtained in each example was performed at 300 MHz using DMSO-d 6 and CDCl 3 .
Figure JPOXMLDOC01-appb-T000028

試験例1:ヒトMGAT2阻害活性の測定
 各々の本発明に係る化合物のDMSO溶液0.2μLを分注したコーニング社製ポリスチレン製384穴マイクロプレートに、アッセイ緩衝液(2mmol/l DTTを含む100mmol/Lリン酸緩衝液(pH7.4))で調製した酵素溶液5μLと基質溶液(100mmol/Lリン酸緩衝液(pH7.4)、30μmol/L 2-Oleoylglycerol、10μmol/L Oleoyl-CoA)5μLを添加し、撹拌及び遠心後、湿潤箱中で室温1時間インキュベーションした。酵素反応後、Internal Standard(IS)を含む停止液(0.2μmol/L Diolein-d5、0.4%ギ酸及び50%イソプロパノールを含む)50μLを添加により反応を停止し、島津GLC社製プレートにシール後、撹拌及び遠心し、RapidFire360及びAgilent 6550 Q-TOF質量分析装置を用いてエレクトロスプレーイオン化法で測定を行った。基質である2-Oleoylglycerolの反応産物(P) DioleinとISのアンモニウム付加体イオンを検出し、そのピーク高さを用いてピーク強度比P/ISを算出し阻害活性を評価した。阻害活性は酵素添加あり/なしをそれぞれControl(+)/Control(-)と規定し、阻害率をそれぞれ0%及び100%阻害として、本発明に係る化合物を添加した時のピーク強度比P/ISをSampleとして、下記の数式によりTIBCO Spotfire(TIBCO Software社製)にて算出した。
 阻害活性(%)=[1-{Sample-Control(-)} / {Control(+)-Control(-)}] * 100
 各々の本発明に係る化合物の阻害活性結果を次の表に示す。表中のIC50(nM)とは、50%の酵素阻害を呈する濃度を示す。
Test Example 1: Measurement of human MGAT2 inhibitory activity To a 384-well polystyrene microplate manufactured by Corning Incorporated, into which 0.2 μL of a DMSO solution of each compound according to the present invention had been dispensed, 5 μL of an enzyme solution prepared with an assay buffer (100 mmol/L phosphate buffer (pH 7.4) containing 2 mmol/L DTT) and 5 μL of a substrate solution (100 mmol/L phosphate buffer (pH 7.4), 30 μmol/L 2-Oleoylglycerol, 10 μmol/L Oleoyl-CoA) were added, stirred and centrifuged, and then incubated in a humidified box at room temperature for 1 hour. After the enzyme reaction, the reaction was stopped by adding 50 μL of a stop solution (containing 0.2 μmol/L Diolein-d5, 0.4% formic acid and 50% isopropanol) containing an internal standard (IS), and the mixture was sealed on a Shimadzu GLC plate, stirred and centrifuged, and then measured by electrospray ionization using a RapidFire360 and an Agilent 6550 Q-TOF mass spectrometer. The reaction product (P) Diolein of the substrate 2-oleoylglycerol and the ammonium adduct ion of IS were detected, and the peak intensity ratio P/IS was calculated using the peak height to evaluate the inhibitory activity. The inhibitory activity was calculated using TIBCO Spotfire (TIBCO Software) according to the following formula, with and without the addition of enzyme defined as Control (+)/Control (-), the inhibition rate was 0% and 100%, respectively, and the peak intensity ratio P/IS when the compound according to the present invention was added was taken as Sample.
Inhibitory activity (%) = [1-{Sample-Control(-)} / {Control(+)-Control(-)}] * 100
The inhibitory activity results of each compound according to the present invention are shown in the following table, in which IC 50 (nM) indicates the concentration exhibiting 50% enzyme inhibition.

Figure JPOXMLDOC01-appb-T000029
Figure JPOXMLDOC01-appb-T000029

試験例2:高脂肪餌負荷肥満マウスにおける肝臓脂肪蓄積に対する作用の検証
 5週齢の雄性C57BL/6Jマウスに高脂肪餌(TestDiet;58Y1)を4週間給餌し、高脂肪餌負荷肥満マウスを作成した。化合物投与の3週間前より1日2回、媒体(0.5%HPMC)を投与した。この馴化投与期間中の体重、摂餌量変化によって無作為化を行い、群分けを実施した。Day0よりDay35もしくはDay88まで1日2回、0.5%HPMCの強制経ロ投与(以下、コントロール群)、あるいは、本発明に係る化合物の強制経ロ投与(以下、II-121及びII-203投与群)を実施した。経口反復投与終了後に血漿及び肝臓を採取し、肝重量及び肝中中性脂肪含量を測定した。また、血漿中肝障害マーカー測定及び肝臓中線維化関連遺伝子発現解析を実施した。
(結果)
 解剖時点のコントロール群に対する肝重量、肝臓中中性脂肪、血漿中肝障害マーカー、肝臓中線維化関連遺伝子発現の減少率は以下であった。これらの結果から肝臓における肝臓脂肪蓄積やNASH病態マーカーの程度を評価し、本発明に係る化合物による肝臓脂肪蓄積及びNASH病態マーカーレベルの抑制を認めた。

Figure JPOXMLDOC01-appb-T000030

Figure JPOXMLDOC01-appb-T000031
Test Example 2: Verification of the effect on hepatic fat accumulation in high-fat diet-fed obese mice Five-week-old male C57BL/6J mice were fed a high-fat diet (TestDiet; 58Y1) for 4 weeks to prepare high-fat diet-fed obese mice. From 3 weeks before compound administration, the vehicle (0.5% HPMC) was administered twice a day. Randomization was performed according to changes in body weight and food intake during this habituation administration period, and grouping was performed. From Day 0 to Day 35 or Day 88, 0.5% HPMC was administered by forced oral administration (hereinafter, control group), or the compound according to the present invention was administered by forced oral administration (hereinafter, II-121 and II-203 administration groups). After the end of repeated oral administration, plasma and liver were collected, and liver weight and hepatic neutral fat content were measured. In addition, plasma liver damage markers and liver fibrosis-related gene expression analysis were performed.
(result)
At the time of dissection, the reduction rates of liver weight, hepatic neutral fat, plasma liver damage markers, and hepatic fibrosis-related gene expression compared to the control group were as follows. From these results, the degree of hepatic fat accumulation and NASH pathology markers in the liver were evaluated, and it was confirmed that the compound according to the present invention suppressed hepatic fat accumulation and NASH pathology marker levels.
Figure JPOXMLDOC01-appb-T000030

Figure JPOXMLDOC01-appb-T000031

試験例3:in vitro肝細胞株を用いた肝臓脂肪蓄積、肝毒性に対する作用の検証
 本発明に係る化合物及び/又は薬剤のin vitro肝脂肪蓄積抑制作用及び細胞毒性作用をヒト肝細胞株 HepG2細胞を用いて検証した。
 HepG2細胞へPBS、本発明に係る化合物又はBMS-963272を10、30μM添加し、一晩培養後の細胞内脂肪滴蓄積を評価した。
(結果)
 結果を図1に示す。化合物II-121及びII-203処置群はPBS処置群と比較し濃度依存的に脂肪滴蓄積を抑制した。一方、BMS-963272処置群は有意な脂肪滴蓄積抑制作用を示さなかった。

 次に、HepG2細胞を用いて本発明に係る化合物の細胞障害性について評価した。
 細胞障害性は各化合物(12.5-100μM)を添加し48時間培養後に、細胞生存率として細胞内ATP含量を、また死細胞評価として培養上清中LDH活性を測定することで評価した。
(結果)
 結果を図2、3に示す。化合物II-203(100μM)により細胞内ATP含量が約5%低下したが、培養上清中LDH活性に変動は認められなかった。同様に、化合物II-121(100μM)により細胞内ATP含量が約7%低下したが、培養上清中LDH活性に変動は認められなかった。一方で、BMS-963272(100μM)により細胞内ATP含量は約67%低下し、培養上清中LDH活性は約9倍上昇した。これらの結果から、BMS-963272は、化合物II-203及びII-121と比較し細胞障害性が強いことが示唆された。
Test Example 3: Verification of effects on hepatic fat accumulation and hepatotoxicity using in vitro hepatic cell line The in vitro hepatic fat accumulation inhibitory effects and cytotoxic effects of the compounds and/or agents according to the present invention were verified using human hepatic cell line HepG2 cells.
PBS, the compound according to the present invention, or BMS-963272 was added to HepG2 cells at 10 or 30 μM, and the accumulation of intracellular lipid droplets after overnight culture was evaluated.
(result)
The results are shown in Figure 1. The compound II-121 and II-203 treatment groups inhibited lipid droplet accumulation in a concentration-dependent manner compared to the PBS treatment group. On the other hand, the BMS-963272 treatment group did not show a significant lipid droplet accumulation inhibitory effect.

Next, the cytotoxicity of the compounds according to the present invention was evaluated using HepG2 cells.
Cytotoxicity was evaluated by adding each compound (12.5-100 μM) and culturing for 48 hours, and then measuring the intracellular ATP content as the cell viability and LDH activity in the culture supernatant as the cell death evaluation.
(result)
The results are shown in Figures 2 and 3. Compound II-203 (100 μM) reduced the intracellular ATP content by about 5%, but no change was observed in the LDH activity in the culture supernatant. Similarly, compound II-121 (100 μM) reduced the intracellular ATP content by about 7%, but no change was observed in the LDH activity in the culture supernatant. On the other hand, BMS-963272 (100 μM) reduced the intracellular ATP content by about 67%, and increased the LDH activity in the culture supernatant by about 9 times. These results suggest that BMS-963272 has stronger cytotoxicity than compounds II-203 and II-121.

試験例4:高脂肪コリン欠乏メチオニン減量飼料給餌マウスにおける肝臓線維化進展に対する作用の検証
 7週齢の雄性C57BL/6Jマウスに高脂肪コリン欠乏メチオニン減量飼料(RESEARCH DIETS、A06071302)を給餌し、給餌4週間後から、1日1回、媒体(0.5%メチルセルロース溶液)の経口反復投与(以下、コントロール群)あるいは3~30mg/kg/10mlの投与量となるように0.5%メチルセルロース溶液に懸濁した本発明に係る化合物の経口反復投与を8週間行った。経口反復投与終了後に肝臓を採取し、採取した肝臓から肝臓中ヒドロキシプロリン含有量を測定した。
(結果)
 解剖時点のコントロール群に対する肝臓中ヒドロキシプロリン含有量の減少率は以下であった。この結果から肝臓における線維化進展の程度を評価し、本発明に係る化合物による線維化進展抑制を認めた。

Figure JPOXMLDOC01-appb-T000032
Test Example 4: Verification of the effect on the progression of liver fibrosis in mice fed a high-fat, choline-deficient, methionine-reduced diet Seven-week-old male C57BL/6J mice were fed a high-fat, choline-deficient, methionine-reduced diet (RESEARCH DIETS, A06071302), and from 4 weeks after feeding, the mice were orally administered a vehicle (0.5% methylcellulose solution) (hereinafter, referred to as the control group) or the compound according to the present invention suspended in a 0.5% methylcellulose solution at a dose of 3 to 30 mg/kg/10 ml once a day for 8 weeks. After the end of the oral administration, the liver was harvested, and the hydroxyproline content in the liver was measured.
(result)
The reduction rates of hydroxyproline content in the liver at the time of dissection relative to the control group were as follows: From these results, the degree of progression of fibrosis in the liver was evaluated, and inhibition of fibrosis progression by the compound according to the present invention was confirmed.
Figure JPOXMLDOC01-appb-T000032

試験例5:ラット4日間反復毒性評価
 本発明に係る化合物及び/又は薬剤の毒性評価をCrl:CD(SD)ラットにおいて検討した。
 6週齢の雄性Crl:CD(SD)ラットに4日間、1日1回、本発明に係る化合物又はBMS-963272の強制経ロ投与を実施し、一般状態観察、体重及び摂餌量測定、血液学及び血液化学的検査並びに病理検査を行った。
(結果)
 化合物II-121及びII-203投与群では、最高用量の500mg/kg/dayの用量まで毒性は認められなかった。一方、BMS-963272投与群は500及び1000mg/kg/dayの用量で血液凝固時間の延長、肝胆管系障害マーカーの上昇が、1000mg/kg/dayの用量で肝障害、腎障害、一般状態の悪化、及び体重/摂餌量の減少が認められた。
Test Example 5: Evaluation of toxicity by repeated administration for four days in rats The toxicity of the compounds and/or drugs according to the present invention was evaluated in Crl:CD (SD) rats.
Six-week-old male Crl:CD (SD) rats were administered the compound according to the present invention or BMS-963272 by gavage once a day for 4 days, and general observations, body weight and food intake measurements, hematology and blood chemistry tests, and pathological tests were performed.
(result)
In the groups administered with compounds II-121 and II-203, no toxicity was observed up to the maximum dose of 500 mg/kg/day. On the other hand, in the group administered with BMS-963272 , prolongation of blood coagulation time and elevation of hepatic biliary system injury markers were observed at doses of 500 and 1000 mg/kg/day, and liver injury, kidney injury, deterioration of general condition, and reduction in body weight/food intake were observed at the dose of 1000 mg/kg/day.

試験例6:代謝安定性試験
 市販のプールドヒト肝ミクロソームと本発明に係る化合物を一定時間反応させ、反応サンプルと未反応サンプルの比較により残存率を算出し、本発明に係る化合物が肝で代謝される程度を評価した。
Test Example 6: Metabolic stability test Commercially available pooled human liver microsomes were reacted with the compound of the present invention for a certain period of time, and the remaining rate was calculated by comparing the reacted sample with the unreacted sample, to evaluate the degree to which the compound of the present invention was metabolized in the liver.

 ヒト肝ミクロソーム0.5mgタンパク質/mLを含む0.2mLの緩衝液(50mmol/L Tris-HCl pH7.4、150mmol/L 塩化カリウム、10mmol/L 塩化マグネシウム)中で、1mmol/L NADPH存在下で37℃、0分あるいは30分間反応させた(酸化的反応)。反応後、メタノール/アセトニトリル=1/1(v/v)溶液の100μLに反応液50μLを添加、混合し、3000rpmで15分間遠心した。その遠心上清中の本発明化合物をLC/MS/MSにて定量し、反応後の本発明に係る化合物の残存量を0分反応時の化合物量を100%として計算した。
(結果)化合物濃度0.5μmol/Lでの残存率を示す。

Figure JPOXMLDOC01-appb-T000033
The mixture was reacted in 0.2 mL of a buffer solution (50 mmol/L Tris-HCl pH 7.4, 150 mmol/L potassium chloride, 10 mmol/L magnesium chloride) containing 0.5 mg protein/mL of human liver microsomes in the presence of 1 mmol/L NADPH at 37°C for 0 or 30 minutes (oxidative reaction). After the reaction, 50 μL of the reaction solution was added to 100 μL of a 1/1 (v/v) methanol/acetonitrile solution, mixed, and centrifuged at 3000 rpm for 15 minutes. The compound of the present invention in the centrifugal supernatant was quantified by LC/MS/MS, and the remaining amount of the compound of the present invention after the reaction was calculated by assuming the amount of the compound at 0 minutes of reaction as 100%.
(Results) The residual rate at a compound concentration of 0.5 μmol/L is shown.
Figure JPOXMLDOC01-appb-T000033

試験例7:溶解性試験
 本発明に係る化合物の溶解度は、1%DMSO添加条件下で決定した。DMSOにて10mmol/L化合物溶液を調製し、本発明に係る化合物溶液6μLをpH6.8人工腸液(0.2mol/L リン酸二水素カリウム試液250mLに0.2mol/L NaOH試液118mL、水を加えて1000mLとした)594μLに添加した。25℃で16時間静置させた後、混液を吸引濾過した。濾液をメタノール/水=1/1(V/V)にて2倍希釈し、絶対検量線法によりHPLC又はLC/MS/MSを用いて濾液中濃度を測定した。
(結果)

Figure JPOXMLDOC01-appb-T000034
Test Example 7: Solubility Test The solubility of the compound according to the present invention was determined under the condition of adding 1% DMSO. A 10 mmol/L compound solution was prepared in DMSO, and 6 μL of the compound solution according to the present invention was added to 594 μL of pH 6.8 artificial intestinal fluid (250 mL of 0.2 mol/L potassium dihydrogen phosphate test solution, 118 mL of 0.2 mol/L NaOH test solution, and water were added to make 1000 mL). After standing at 25° C. for 16 hours, the mixture was filtered by suction. The filtrate was diluted 2-fold with methanol/water = 1/1 (V/V), and the concentration in the filtrate was measured using HPLC or LC/MS/MS by the absolute calibration curve method.
(result)
Figure JPOXMLDOC01-appb-T000034

試験例8:光毒性試験
 In vitro光毒性試験として、生体膜に対する作用及び光過酸化を指標とした評価法である赤血球光溶血試験(Wolfgang J.W. Pepe et al., ATLA29, 145-162, 2001)を行った。本法では、ジメチルスホキシドを媒体とした本発明に係る化合物の調製液に対し2.5%(v/v)のヒツジ赤血球液を添加した混合液(濃度:0.1~0.0008%)を用いた。この混合液を添加したマイクロプレートを二つ用意し、一つのマイクロプレートに紫外線蛍光ランプ(GL20SEランプ、三共電気及びFL20S-BLBランプ、パナソニック)を用いてUVA及びUVB領域での光照射(10J/cm、290~400nm)を行い、光照射を行わなかったマイクロプレートと共に遠心操作を加えた後、上清の吸光度(540nm又は630nm)を測定した。光毒性の判定に用いる二つの指標(生体膜に対する作用及び光過酸化)を求めるために、本発明に係る化合物から得られた吸光度について光照射及び未照射のそれぞれで媒体での吸光度を差し引いた値を算出し、以降の計算に供した。生体膜に対する作用に関しては光照射と未照射の吸光度(540nm)の差から光溶血率を求め、光過酸化に対しては光照射と未照射の吸光度(630nm)の変化量を求めた。なお光溶血率の計算においては蒸留水を用いて強制溶血させた2.5%(v/v)のヒツジ赤血球液から得られた吸光度(540nm)を光溶血率100%の基準とした。光溶血率が10%未満であり、630nmでの吸光度の変化量が0.05未満の場合を(-)とし、光溶血率が10%以上であり、630nmでの吸光度の変化量が0.05以上の場合を(+)とした。
(結果)
化合物I-34:(-)
Test Example 8: Phototoxicity test As an in vitro phototoxicity test, a red blood cell photohemolysis test (Wolfgang JW Pepe et al., ATLA29, 145-162, 2001), which is an evaluation method using the effect on biological membranes and photoperoxidation as indicators, was performed. In this method, a mixed solution (concentration: 0.1 to 0.0008%) was used in which 2.5% (v/v) sheep red blood cell solution was added to a preparation solution of the compound according to the present invention using dimethyl sulfoxide as a medium. Two microplates containing this mixture were prepared, and one microplate was irradiated with UVA and UVB light (10 J/cm 2 , 290-400 nm) using an ultraviolet fluorescent lamp (GL20SE lamp, Sankyo Electric and FL20S-BLB lamp, Panasonic), and centrifuged together with the microplate that was not irradiated, and the absorbance of the supernatant (540 nm or 630 nm) was measured. In order to determine two indices used in determining phototoxicity (action on biomembranes and photoperoxidation), the absorbance obtained from the compound according to the present invention was subtracted from the absorbance in the medium for each of the light-irradiated and non-irradiated cases, and the values were used in the following calculations. Regarding the action on biomembranes, the photohemolysis rate was calculated from the difference between the absorbance (540 nm) of the light-irradiated and non-irradiated cases, and regarding photoperoxidation, the change in absorbance (630 nm) of the light-irradiated and non-irradiated cases was calculated. In calculating the rate of photohemolysis, the absorbance (540 nm) obtained from a 2.5% (v/v) sheep red blood cell solution subjected to forced hemolysis using distilled water was used as the standard for the rate of photohemolysis of 100%. When the rate of photohemolysis was less than 10% and the change in absorbance at 630 nm was less than 0.05, it was marked as (-), and when the rate of photohemolysis was 10% or more and the change in absorbance at 630 nm was 0.05 or more, it was marked as (+).
(result)
Compound I-34: (-)

試験例9:細胞障害性試験
 細胞イメージアナライザーであるToxinsight(Thermofisher Scientific社)を用いて、化合物曝露後の細胞数を自動測定し、本発明に係る化合物の細胞障害性を評価した。
 HepG2細胞(ヒト肝がん細胞由来)を384ウェルプレートに60000cells/mLとなるように播種し、24時間後に各ウェルに化合物溶液を添加した。化合物溶液としては、本発明化合物を含むDMSO溶液(最高濃度を50μmol/Lに設定し2倍公比で5段階希釈、最低濃度は約3.1μmol/L)、陰性対照としてDMSOのみの溶液、陽性対照としてカンプトテシン溶液を用いた。本発明に係る化合物のDMSO溶液、陰性対照溶液、又は陽性対照溶液を各ウェルに添加した。71時間後に、最終濃度1μg/mLになるようにダルベッコリン酸緩衝液(D-PBS)で希釈したHoechst 33342溶液を各ウェルに添加し、37℃、5%COインキュベーター内で1時間核の染色を行った。染色後、4%パラフォルムアルデヒドを用いて20分間、37℃のCOインキュベーター内で固定した。最後に、D-PBSで3回洗浄後、Toxinsight(Thermofisher Scientific社)を用いて、ウェル毎の蛍光発色した核の数を計測した。1濃度あたり4ウェルを設け、4ウェル中の核の数(障害性がみられなかった細胞数)の平均値とばらつき(SD)算出した。陰性対照群と比較し、平均値が陰性対照の平均値から50%以上減少する化合物曝露濃度(IC50)を算出する。IC50の値が小さいほど細胞障害性のリスクが高いと判断した。
Test Example 9: Cytotoxicity test Using a cell image analyzer Toxinsight (Thermofisher Scientific), the number of cells after exposure to the compound was automatically measured to evaluate the cytotoxicity of the compound according to the present invention.
HepG2 cells (derived from human hepatoma cells) were seeded in a 384-well plate at 60,000 cells/mL, and after 24 hours, a compound solution was added to each well. As the compound solution, a DMSO solution containing the compound of the present invention (maximum concentration set at 50 μmol/L, diluted five times with a common ratio of 2 times, minimum concentration was about 3.1 μmol/L), a solution of DMSO only as a negative control, and a camptothecin solution as a positive control were used. A DMSO solution of the compound of the present invention, a negative control solution, or a positive control solution was added to each well. After 71 hours, a Hoechst 33342 solution diluted with Dulbecco's phosphate buffer (D-PBS) to a final concentration of 1 μg/mL was added to each well, and the nuclei were stained for 1 hour in a 37°C, 5% CO2 incubator. After staining, the cells were fixed with 4% paraformaldehyde for 20 minutes in a 37°C CO2 incubator. Finally, after washing three times with D-PBS, the number of fluorescent nuclei per well was counted using Toxinsight (Thermofisher Scientific). Four wells were set up per concentration, and the average and variance (SD) of the number of nuclei (number of cells in which no damage was observed) in the four wells were calculated. The compound exposure concentration (IC50) at which the average value was reduced by 50% or more from the average value of the negative control was calculated in comparison with the negative control group. It was determined that the smaller the IC50 value, the higher the risk of cytotoxicity.

試験例10:CYP阻害試験
 市販のプールドヒト肝ミクロソームを用いて、ヒト主要CYP5分子種(CYP1A2、2C9、2C19、2D6、3A4)の典型的基質代謝反応として7-エトキシレゾルフィンのO-脱エチル化(CYP1A2)、トルブタミドのメチル-水酸化(CYP2C9)、メフェニトインの4’-水酸化(CYP2C19)、デキストロメトルファンのO脱メチル化(CYP2D6)、テルフェナジンの水酸化(CYP3A4)を指標とし、それぞれの代謝物生成量が本発明に係る化合物によって阻害される程度を評価した。
 反応条件は以下のとおり:基質、0.5μmol/L エトキシレゾルフィン(CYP1A2)、100μmol/L トルブタミド(CYP2C9)、50μmol/L S-メフェニトイン(CYP2C19)、5μmol/L デキストロメトルファン(CYP2D6)、1μmol/L テルフェナジン(CYP3A4);反応時間、15分;反応温度、37℃;酵素、プールドヒト肝ミクロソーム0.2mg タンパク質/mL;本発明に係る化合物濃度、1、5、10、20μmol/L(4点)。
 96穴プレートに反応溶液として、50mmol/L Hepes緩衝液中に各5種の基質、ヒト肝ミクロソーム、本発明に係る化合物を上記組成で加え、補酵素であるNADPHを添加して、指標とする代謝反応を開始した。37℃、15分間反応した後、メタノール/アセトニトリル=1/1(V/V)溶液を添加することで反応を停止した。3000rpm、15分間の遠心後、遠心上清中のレゾルフィン(CYP1A2代謝物)を蛍光マルチラベルカウンタ又はLC/MS/MSで定量し、トルブタミド水酸化体(CYP2C9代謝物)、メフェニトイン4’水酸化体(CYP2C19代謝物)、デキストロルファン(CYP2D6代謝物)、テルフェナジンアルコール体(CYP3A4代謝物)をLC/MS/MSで定量した。
 薬物を溶解した溶媒であるDMSOのみを反応系に添加したものをコントロール(100%)とし、残存活性(%)を算出し、濃度と抑制率を用いて、ロジスティックモデルによる逆推定によりIC50を算出した。
(結果)
化合物II-103:5種 >20μmol/L
Test Example 10: CYP inhibition test Using commercially available pooled human liver microsomes, the extent to which the production amount of each metabolite was inhibited by the compound of the present invention was evaluated using as indicators the O-deethylation of 7-ethoxyresorufin (CYP1A2), methyl-hydroxylation of tolbutamide (CYP2C9), 4'-hydroxylation of mephenytoin (CYP2C19), O-demethylation of dextromethorphan (CYP2D6), and hydroxylation of terfenadine (CYP3A4) as typical substrate metabolic reactions of human major CYP5 molecular species (CYP1A2, 2C9, 2C19, 2D6, 3A4).
The reaction conditions were as follows: substrate, 0.5 μmol/L ethoxyresorufin (CYP1A2), 100 μmol/L tolbutamide (CYP2C9), 50 μmol/L S-mephenytoin (CYP2C19), 5 μmol/L dextromethorphan (CYP2D6), 1 μmol/L terfenadine (CYP3A4); reaction time, 15 minutes; reaction temperature, 37° C.; enzyme, pooled human liver microsome, 0.2 mg protein/mL; compound of the present invention concentration, 1, 5, 10, 20 μmol/L (4 points).
Five kinds of substrates, human liver microsomes, and the compound according to the present invention were added to a 50 mmol/L Hepes buffer solution in a 96-well plate in the above composition as a reaction solution, and NADPH, a coenzyme, was added to start the metabolic reaction as an indicator. After reacting at 37°C for 15 minutes, the reaction was stopped by adding a methanol/acetonitrile = 1/1 (V/V) solution. After centrifugation at 3000 rpm for 15 minutes, resorufin (CYP1A2 metabolite) in the centrifugal supernatant was quantified by a fluorescent multi-label counter or LC/MS/MS, and tolbutamide hydroxylated form (CYP2C9 metabolite), mephenytoin 4' hydroxylated form (CYP2C19 metabolite), dextrorphan (CYP2D6 metabolite), and terfenadine alcohol form (CYP3A4 metabolite) were quantified by LC/MS/MS.
A reaction system in which only DMSO, the solvent in which the drug was dissolved, was added was used as a control (100%). The remaining activity (%) was calculated, and the IC50 was calculated by inverse estimation using a logistic model using the concentration and inhibition rate.
(result)
Compound II-103: 5 species >20 μmol/L

試験例11:BA試験
経口吸収性の検討実験材料と方法
(1)使用動物:マウスあるいはSDラットを使用した。
(2)飼育条件:マウスあるいはSDラットは、固形飼料及び滅菌水道水を自由摂取させた。
(3)投与量、群分けの設定:経口投与、静脈内投与を所定の投与量により投与した。以下のように群を設定した。(化合物ごとで投与量は変更有)
 経口投与 1~30mg/kg(n=2~3)
 静脈内投与 0.5~10mg/kg(n=2~3)
(4)投与液の調製:経口投与は溶液又は懸濁液として投与した。静脈内投与は可溶化して投与した。
(5)投与方法:経口投与は、経口ゾンデにより強制的に胃内に投与した。静脈内投与は、注射針を付けたシリンジにより尾静脈又は大腿静脈から投与した。
(6)評価項目:経時的に採血し、血漿中本発明に係る化合物濃度をLC/MS/MSを用いて測定した。
(7)統計解析:血漿中本発明に係る化合物濃度推移について、非線形最小二乗法プログラムWinNonlin(登録商標)を用いて血漿中濃度‐時間曲線下面積(AUC)を算出し、経口投与群と静脈内投与群のAUCから本発明に係る化合物のバイオアベイラビリティ(BA)を算出した。
Test Example 11: BA Test: Examination of oral absorbability Experimental materials and methods (1) Animals used: Mice or SD rats were used.
(2) Rearing conditions: Mice or SD rats were allowed to freely consume solid food and sterilized tap water.
(3) Dosage and grouping: Oral and intravenous administration was performed at a predetermined dose. The groups were set up as follows. (Dosages were changed for each compound.)
Oral administration: 1-30 mg/kg (n=2-3)
Intravenous administration: 0.5-10 mg/kg (n=2-3)
(4) Preparation of administration liquid: For oral administration, the compound was administered as a solution or suspension. For intravenous administration, the compound was administered in a solubilized form.
(5) Method of administration: Oral administration was performed by forcibly administering the compound into the stomach using an oral probe, whereas intravenous administration was performed by administering the compound into the tail vein or femoral vein using a syringe with an injection needle.
(6) Evaluation item: Blood samples were collected over time, and the plasma concentration of the compound according to the present invention was measured using LC/MS/MS.
(7) Statistical analysis: The area under the plasma concentration-time curve (AUC) of the compound according to the present invention was calculated using the nonlinear least squares program WinNonlin (registered trademark) for the plasma concentration transition of the compound according to the present invention, and the bioavailability (BA) of the compound according to the present invention was calculated from the AUC of the oral administration group and the intravenous administration group.

試験例12:CYP3A4(MDZ)MBI試験
 本発明化合物のCYP3A4阻害に関して代謝反応による増強からMechanism based inhibition(MBI)能を評価する試験である。プールドヒト肝ミクロソームを用いてミダゾラム(MDZ)の1-水酸化反応を指標としてCYP3A4阻害を評価した。
 反応条件は以下のとおり:基質、10μmol/L MDZ;プレ反応時間、0又は30分;反応時間、2分;反応温度、37℃;プールドヒト肝ミクロソーム、プレ反応時0.5mg/mL、反応時0.05mg/mL(10倍希釈時);本発明に係る化合物プレ反応時の濃度、1、5、10、20μmol/L(4点)。
 96穴プレートにプレ反応液としてK-Pi緩衝液(pH7.4)中にプールドヒト肝ミクロソーム、本発明に係る化合物溶液を上記のプレ反応の組成で加え、別の96穴プレートに基質とK-Pi緩衝液で1/10希釈されるようにその一部を移行し、補酵素であるNADPHを添加して指標とする反応を開始し(プレ反応無)、所定の時間反応後、メタノール/アセトニトリル=1/1(V/V)溶液を加えることによって反応を停止した。また残りのプレ反応液にもNADPHを添加しプレ反応を開始し(プレ反応有)、所定時間プレ反応後、別のプレートに基質とK-Pi緩衝液で1/10希釈されるように一部を移行し指標とする反応を開始した。所定の時間反応後、メタノール/アセトニトリル=1/1(V/V)溶液を加えることによって反応を停止する。それぞれの指標反応を行ったプレートを3000rpm、15分間の遠心後、遠心上清中の1-水酸化ミダゾラムをLC/MS/MSで定量した。
 本発明に係る化合物を溶解した溶媒であるDMSOのみを反応系に添加したものをコントロール(100%)とし、本発明に係る化合物をそれぞれの濃度添加したときの残存活性(%)を算出し、濃度と阻害率を用いて、ロジスティックモデルによる逆推定によりICを算出した。Preincubataion 0minのIC/Preincubataion 30minのICをShifted IC値とし、Shifted ICが1.5以上であればPositive、Shifted ICが1.0以下であればNegativeとした。
(結果)
化合物II-103:Negative
Test Example 12: CYP3A4 (MDZ) MBI Test This is a test to evaluate the mechanism based inhibition (MBI) ability of the compound of the present invention from the enhancement by metabolic reaction regarding CYP3A4 inhibition. CYP3A4 inhibition was evaluated using pooled human liver microsomes with the 1-hydroxylation reaction of midazolam (MDZ) as an index.
The reaction conditions were as follows: substrate, 10 μmol/L MDZ; pre-reaction time, 0 or 30 minutes; reaction time, 2 minutes; reaction temperature, 37° C.; pooled human liver microsomes, 0.5 mg/mL at pre-reaction and 0.05 mg/mL (at 10-fold dilution) at reaction; concentration of the compound according to the present invention at pre-reaction, 1, 5, 10, and 20 μmol/L (4 points).
Pooled human liver microsomes and a compound solution according to the present invention were added to a 96-well plate as a pre-reaction solution in K-Pi buffer (pH 7.4) in the above pre-reaction composition, and a portion of the solution was transferred to another 96-well plate so as to be diluted 1/10 with the substrate and K-Pi buffer, and a coenzyme, NADPH, was added to start the reaction using the indicator (no pre-reaction). After a predetermined reaction time, the reaction was stopped by adding a methanol/acetonitrile = 1/1 (V/V) solution. NADPH was also added to the remaining pre-reaction solution to start the pre-reaction (with pre-reaction). After a predetermined reaction time, a portion of the solution was transferred to another plate so as to be diluted 1/10 with the substrate and K-Pi buffer, and a reaction using the indicator was started. After a predetermined reaction time, the reaction was stopped by adding a methanol/acetonitrile = 1/1 (V/V) solution. The plates in which the respective indicator reactions were performed were centrifuged at 3000 rpm for 15 minutes, and 1-hydroxymidazolam in the centrifugal supernatant was quantified by LC/MS/MS.
The reaction system was treated as a control (100%) with only DMSO, the solvent in which the compound according to the present invention was dissolved, and the residual activity (%) was calculated when the compound according to the present invention was added at each concentration. The IC was calculated by inverse estimation using a logistic model using the concentration and inhibition rate. The IC at 0 min preincubation/IC at 30 min preincubation was used as the Shifted IC value, and if the Shifted IC was 1.5 or more, it was considered positive, and if the Shifted IC was 1.0 or less, it was considered negative.
(result)
Compound II-103: Negative

試験例13:粉末溶解度試験
 適当な容器に本発明に係る化合物を適量入れ、各容器にJP-1液(塩化ナトリウム2.0g、塩酸7.0mLに水を加えて1000mLとする)、JP-2液(pH6.8のリン酸塩緩衝液500mLに水500mLを加える)、20mmol/L タウロコール酸ナトリウム(TCA)/JP-2液(TCA1.08gにJP-2液を加え100mLとする)を200μLずつ添加した。試験液添加後に全量溶解した場合には、適宜、本発明に係る化合物を追加した。密閉して37℃で1時間振とう後に濾過し、各濾液100μLにメタノール100μLを添加して2倍希釈を行う。希釈倍率は、必要に応じて変更する。気泡及び析出物がないかを確認し、密閉して振とうした。絶対検量線法によりHPLCを用いて本発明に係る化合物を定量した。
Test Example 13: Powder solubility test An appropriate amount of the compound according to the present invention was placed in an appropriate container, and 200 μL of JP-1 solution (2.0 g of sodium chloride, 7.0 mL of hydrochloric acid, and water added to make 1000 mL), JP-2 solution (500 mL of water added to 500 mL of phosphate buffer solution at pH 6.8), and 20 mmol/L sodium taurocholate (TCA)/JP-2 solution (1.08 g of TCA and JP-2 solution added to make 100 mL) were added to each container. If the entire amount was dissolved after the addition of the test solution, the compound according to the present invention was appropriately added. The container was sealed and shaken at 37° C. for 1 hour, and then filtered, and 100 μL of methanol was added to 100 μL of each filtrate to perform 2-fold dilution. The dilution ratio was changed as necessary. The container was checked for the presence of bubbles and precipitates, and then sealed and shaken. The compound according to the present invention was quantified using HPLC by the absolute calibration curve method.

試験例14:Fluctuation Ames試験
 本発明に係る化合物の変異原性を評価した。
 凍結保存しているネズミチフス菌(Salmonella typhimurium TA98株、TA100株)20μLを10mL液体栄養培地(2.5% Oxoid nutrient broth No.2)に接種し37℃にて10時間、振盪前培養した。TA98株は8.0mLの菌液を遠心(2000×g、10分間)して培養液を除去した。8.0mLのMicro F緩衝液(KHPO:3.5g/L、KHPO:1g/L、(NHSO:1g/L、クエン酸三ナトリウム二水和物:0.25g/L、MgSO・7H0:0.1g/L)に菌を懸濁し、120mLのExposure培地(ビオチン:8μg/mL、ヒスチジン:0.2μg/mL、グルコース:8mg/mLを含むMicroF緩衝液)に添加した。TA100株は3.1mL菌液に対しExposure培地120mLに添加し試験菌液を調製した。本発明に係る化合物DMSO溶液(最高用量50mg/mLから2~3倍公比で数段階希釈)、陰性対照としてDMSO、陽性対照として非代謝活性化条件ではTA98株に対しては50μg/mLの4-ニトロキノリン-1-オキシドDMSO溶液、TA100株に対しては0.25μg/mLの2-(2-フリル)-3-(5-ニトロ-2-フリル)アクリルアミドDMSO溶液、代謝活性化条件ではTA98株に対して40μg/mLの2-アミノアントラセンDMSO溶液、TA100株に対しては20μg/mLの2-アミノアントラセンDMSO溶液それぞれ12μLと試験菌液588μL(代謝活性化条件では試験菌液498μLとS9 mix 90μLの混合液)を混和し、37℃にて90分間、振盪培養した。本発明に係る化合物を暴露した菌液230μLを、Indicator培地(ビオチン:8μg/mL、ヒスチジン:0.2μg/mL、グルコース:8mg/mL、ブロモクレゾールパープル:37.5μg/mLを含むMicroF緩衝液)1150μLに混和し、50μLずつマイクロプレート48ウェル/用量に分注し、37℃にて3日間、静置培養した。アミノ酸(ヒスチジン)合成酵素遺伝子の突然変異によって増殖能を獲得した菌を含むウェルは、pH変化により紫色から黄色に変色するため、1用量あたり48ウェル中の黄色に変色した菌増殖ウェルを計数し、陰性対照群と比較して評価した。変異原性が陰性のものを(-)、陽性のものを(+)として示す。
Test Example 14: Fluctuation Ames test The mutagenicity of the compound according to the present invention was evaluated.
20 μL of frozen Salmonella typhimurium (Salmonella typhimurium TA98 strain, TA100 strain) was inoculated into 10 mL of liquid nutrient medium (2.5% Oxoid nutrient broth No. 2) and pre-cultured with shaking for 10 hours at 37° C. For the TA98 strain, 8.0 mL of the bacterial solution was centrifuged (2000×g, 10 minutes) to remove the culture medium. The bacteria were suspended in 8.0 mL of Micro F buffer (K 2 HPO 4 : 3.5 g / L, KH 2 PO 4 : 1 g / L, (NH 4 ) 2 SO 4 : 1 g / L, trisodium citrate dihydrate: 0.25 g / L, MgSO 4 · 7H 2 0: 0.1 g / L) and added to 120 mL of Exposure medium (Micro F buffer containing biotin: 8 μg / mL, histidine: 0.2 μg / mL, glucose: 8 mg / mL). TA100 strain was added to 120 mL of Exposure medium per 3.1 mL of bacterial solution to prepare a test bacterial solution. The compound DMSO solution according to the present invention (diluted in several stages at a common ratio of 2 to 3 times from the maximum dose of 50 mg / mL), DMSO as a negative control, 50 μg / mL 4-nitroquinoline-1-oxide DMSO solution for the TA98 strain as a positive control under non-metabolic activation conditions, 0.25 μg / mL 2- (2-furyl) -3- (5-nitro-2-furyl) acrylamide DMSO solution for the TA100 strain, 40 μg / mL 2-aminoanthracene DMSO solution for the TA98 strain under metabolic activation conditions, 20 μg / mL 2-aminoanthracene DMSO solution for the TA100 strain were mixed with 12 μL each and 588 μL of test bacteria solution (a mixture of 498 μL of test bacteria solution and 90 μL of S9 mix under metabolic activation conditions), and cultured with shaking at 37 ° C. for 90 minutes. 230 μL of the bacterial solution exposed to the compound according to the present invention was mixed with 1150 μL of Indicator medium (MicroF buffer containing biotin: 8 μg/mL, histidine: 0.2 μg/mL, glucose: 8 mg/mL, bromocresol purple: 37.5 μg/mL), and 50 μL was dispensed into 48 wells/dose of a microplate, and cultured at 37° C. for 3 days. Since wells containing bacteria that have acquired the ability to grow due to mutation of the amino acid (histidine) synthase gene change color from purple to yellow due to pH change, the number of wells that have grown yellow in the 48 wells per dose were counted and evaluated in comparison with the negative control group. Negative mutagenicity is indicated as (-), and positive mutagenicity is indicated as (+).

試験例15:hERG試験
 本発明に係る化合物の心電図QT間隔延長リスク評価を目的として、human ether-a-go-go related gene (hERG)チャネルを発現させたCHO細胞を用いて、心室再分極過程に重要な役割を果たす遅延整流K電流(IKr)への本発明化合物の作用を検討した。
 全自動パッチクランプシステム(QPatch;Sophion Bioscience A/S)を用い、ホールセルパッチクランプ法により、細胞を-80mVの膜電位に保持し、-50mVのリーク電位を与えた後、+20mVの脱分極刺激を2秒間、さらに-50mVの再分極刺激を2秒間与えた際に誘発されるIKrを記録した。発生する電流が安定した後、本発明に係る化合物を目的の濃度で溶解させた細胞外液(NaCl:145 mmol/L、KCl:4 mmol/L、CaCl:2 mmol/L、MgCl:1 mmol/L、グルコース:10 mmol/L、HEPES(4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid、4-(2-ヒドロキシエチル)-1-ピペラジンエタンスルホン酸):10mmol/L、pH=7.4)を室温条件下で、10分間細胞に適用させる。得られたIKrから、解析ソフト(Falster Patch;Sophion Bioscience A/S)を使用して、保持膜電位における電流値を基準に最大テール電流の絶対値を計測した。さらに、本発明に係る化合物適用前の最大テール電流に対する阻害率を算出し、本発明化合物のIKrへの影響を評価した。
(結果)化合物濃度10μmol/Lでの阻害率を示す。
化合物I-253:6.9%
Test Example 15: hERG Test For the purpose of evaluating the risk of electrocardiogram QT interval prolongation of the compound according to the present invention, the effect of the compound according to the present invention on delayed rectifier K + current (I Kr ), which plays an important role in the ventricular repolarization process, was investigated using CHO cells expressing human ether-a-go-go related gene (hERG) channel.
Using a fully automated patch clamp system (QPatch; Sophion Bioscience A/S), the cells were held at a membrane potential of -80 mV by the whole-cell patch clamp method, and a leak potential of -50 mV was applied, followed by a depolarizing stimulus of +20 mV for 2 seconds and a repolarizing stimulus of -50 mV for 2 seconds. The I Kr evoked by this was recorded. After the generated current became stable, an extracellular solution containing the compound according to the present invention dissolved at a desired concentration (NaCl: 145 mmol/L, KCl: 4 mmol/L, CaCl 2 : 2 mmol/L, MgCl 2 : 1 mmol/L, glucose: 10 mmol/L, HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid): 10 mmol/L, pH=7.4) was applied to the cells at room temperature for 10 minutes. From the obtained I Kr , the absolute value of the maximum tail current was measured based on the current value at the resting membrane potential using analysis software (Falster Patch; Sophion Bioscience A/S). Furthermore, the inhibition rate relative to the maximum tail current before application of the compound according to the present invention was calculated to evaluate the effect of the compound according to the present invention on I Kr .
(Results) The inhibition rate at a compound concentration of 10 μmol/L is shown.
Compound I-253: 6.9%

試験例16:生殖発生毒性試験
 本発明に係る化合物の胚・胎児発生毒性評価をCrl:CD(SD)ラットにおいて検討した。
 10~14週齢の雌性Crl:CD(SD)ラットに妊娠6~17日の12日間、1日1回、本発明に係る化合物の強制経ロ投与を実施し、妊娠21日に帝王切開した。母動物の一般状態観察、体重及び摂餌量測定、剖検、及び黄体数検査を行った。また、胚・胎児の生存性及び性別確認、体重及び胎盤重量測定、外表・胎盤肉眼検査、内部器官・骨格検査を行った。
(結果)
 II-203投与群では、1000mg/kg/dayの用量まで毒性は認められなかった。
Test Example 16: Reproductive and developmental toxicity test The embryo-fetal developmental toxicity of the compound according to the present invention was evaluated in Crl:CD (SD) rats.
The compound of the present invention was administered by gavage once a day to 10-14 week old female Crl:CD (SD) rats for 12 days from 6 to 17 days of pregnancy, and then cesarean section was performed on 21 days of pregnancy. The general condition of the dams was observed, and the weight and food intake were measured, and necropsy and luteal count were examined. In addition, the viability and sex of the embryos and fetuses were confirmed, and the weight and placenta were measured, and external and placental visual examinations were performed, and internal organs and skeletons were examined.
(result)
In the II-203 administration group, no toxicity was observed up to a dose of 1000 mg/kg/day.

 以下に示す製剤例は例示にすぎないものであり、発明の範囲を何ら限定することを意図するものではない。
 本発明の化合物は、任意の従来の経路により、特に、経腸、例えば、経口で、例えば、錠剤又はカプセル剤の形態で、又は非経口で、例えば注射液剤又は懸濁剤の形態で、局所で、例えば、ローション剤、ゲル剤、軟膏剤又はクリーム剤の形態で、又は経鼻形態又は座剤形態で医薬組成物として投与することができる。少なくとも1種の薬学的に許容される担体又は希釈剤と一緒にして、遊離形態又は薬学的に許容される塩の形態の本発明の化合物を含む医薬組成物は、従来の方法で、混合、造粒又はコーティング法によって製造することができる。例えば、経口用組成物としては、賦形剤、崩壊剤、結合剤、滑沢剤等及び有効成分等を含有する錠剤、顆粒剤、カプセル剤とすることができる。また、注射用組成物としては、溶液剤又は懸濁剤とすることができ、滅菌されていてもよく、また、保存剤、安定化剤、緩衝化剤等を含有してもよい。
The formulation examples shown below are merely illustrative and are not intended to limit the scope of the invention in any way.
The compounds of the present invention can be administered as pharmaceutical compositions by any conventional route, in particular enterally, e.g., orally, e.g., in the form of tablets or capsules, or parenterally, e.g., in the form of injection solutions or suspensions, topically, e.g., in the form of lotions, gels, ointments or creams, or in the form of intranasal or suppositories. Pharmaceutical compositions containing the compounds of the present invention in free form or in the form of a pharma-ceutically acceptable salt together with at least one pharma-ceutically acceptable carrier or diluent can be prepared by conventional mixing, granulation or coating methods. For example, oral compositions can be tablets, granules, or capsules containing excipients, disintegrants, binders, lubricants, etc., and active ingredients, etc. In addition, injectable compositions can be solutions or suspensions, which may be sterilized and may contain preservatives, stabilizers, buffers, etc.

 本発明の非アルコール性脂肪肝疾患の治療及び/又は予防方法、及びそれに用いる治療用医薬組成物は、活性成分の式(I)で示される化合物又はその製薬上許容される塩の所定量を、非アルコール性脂肪肝疾患の患者に対して投与することにより、優れた治療効果を示すと考えられる。また、式(I)で示される化合物又はその製薬上許容される塩の投与による止血異常、肝障害、腎障害又は胚致死性などの副作用は認められず、極めて安全に適用することができ、長期間の投与にも適していることから、本発明の治療及び/又は予防方法及び治療用医薬組成物は、きわめて優れた治療及び/又は予防方法及び治療用医薬組成物である。 The method for treating and/or preventing non-alcoholic fatty liver disease of the present invention and the pharmaceutical composition for treatment therewith are believed to show excellent therapeutic effects by administering a predetermined amount of the active ingredient, the compound represented by formula (I) or a pharma- ceutical acceptable salt thereof, to a patient with non-alcoholic fatty liver disease. Furthermore, the administration of the compound represented by formula (I) or a pharma-ceutical acceptable salt thereof does not cause side effects such as hemostatic abnormalities, liver damage, kidney damage, or embryonic lethality, and can be applied extremely safely and is suitable for long-term administration, so the method for treating and/or preventing and the pharmaceutical composition for treatment of the present invention are extremely excellent.

Claims (13)

 式:
Figure JPOXMLDOC01-appb-C000001

(式中、
 R2a及びR2bは、隣接する炭素原子と一緒になって、環Bを形成し;
 環Bは、式:
Figure JPOXMLDOC01-appb-C000002

(式中、Rは、それぞれ独立して、ハロゲン、ハロアルキル、アルキルオキシ、ハロアルキルオキシ、又はシクロプロパニルである)であり;
 R4aは、式:
Figure JPOXMLDOC01-appb-C000003

(式中、
 Lは、アルキレンであり、
 Rは、アルキルスルフォニルである)であり;
 R4bは、置換基群αで置換されていてもよいアルキル、置換基群βで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり;
 置換基群αは、ハロゲン、ハロアルキルオキシ、及びシクロプロパニルであり、
 置換基群βは、ハロゲン、シアノ、アルキル、ハロアルキル、及びシクロプロパニルである)で示される化合物又はその製薬上許容される塩を含有する、脂肪性肝疾患を治療及び/又は予防するための、医薬組成物。
formula:
Figure JPOXMLDOC01-appb-C000001

(In the formula,
R 2a and R 2b together with adjacent carbon atoms form ring B;
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000002

wherein each R6 is independently halogen, haloalkyl, alkyloxy, haloalkyloxy, or cyclopropanyl;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000003

(In the formula,
L3 is alkylene;
R 7 is alkylsulfonyl;
R 4b is an alkyl group optionally substituted with substituent group α, a phenyl group optionally substituted with substituent group β, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β;
Substituent group α is halogen, haloalkyloxy, and cyclopropanyl;
A pharmaceutical composition for treating and/or preventing fatty liver disease, comprising a compound represented by the formula (I) or a pharma- ceutically acceptable salt thereof, wherein substituent group β is halogen, cyano, alkyl, haloalkyl, and cyclopropanyl.
 Rが、それぞれ独立して、ハロゲン、ハロアルキル、又はハロアルキルオキシであり、
 R4aが、式:
Figure JPOXMLDOC01-appb-C000004

であり、
 R4bが、ハロアルキルオキシアルキル、ハロゲンで置換されていてもよいフェニル基、又は置換基群βで置換されていてもよい5~6員の芳香族複素環式基であり、
 置換基群βは、ハロゲン及びアルキルである、請求項1記載の医薬組成物。
R 6 is independently halogen, haloalkyl, or haloalkyloxy;
R 4a is a group represented by the formula:
Figure JPOXMLDOC01-appb-C000004

and
R 4b is a haloalkyloxyalkyl, a phenyl group optionally substituted with halogen, or a 5- to 6-membered aromatic heterocyclic group optionally substituted with substituent group β,
The pharmaceutical composition according to claim 1 , wherein the substituent group β is halogen and alkyl.
 環Bが、式:
Figure JPOXMLDOC01-appb-C000005

(式中、各記号は請求項1又は2と同意義)
である、請求項1又は2記載の医薬組成物。
Ring B is of the formula:
Figure JPOXMLDOC01-appb-C000005

(In the formula, each symbol has the same meaning as in claim 1 or 2.)
The pharmaceutical composition according to claim 1 or 2,
 化合物I-8、I-23、I-34、I-190、I-212、I-236、I-253、I-275、I-276、II-93、II-103、II-121、II-151、II-168、II-174、II-203、II-225、II-233及びII-295からなる群から選択される化合物又はその製薬上許容される塩を含有する、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, comprising a compound selected from the group consisting of compounds I-8, I-23, I-34, I-190, I-212, I-236, I-253, I-275, I-276, II-93, II-103, II-121, II-151, II-168, II-174, II-203, II-225, II-233 and II-295, or a pharma- ceutical acceptable salt thereof.  化合物I-236、I-253、I-275、II-103、II-121、II-174、II-203、II-225及びII-233からなる群から選択される化合物又はその製薬上許容される塩を含有する、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, which contains a compound selected from the group consisting of compounds I-236, I-253, I-275, II-103, II-121, II-174, II-203, II-225 and II-233, or a pharma- ceutical acceptable salt thereof.  前記脂肪性肝疾患が非アルコール性脂肪肝疾患(NAFLD)である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is nonalcoholic fatty liver disease (NAFLD).  前記脂肪性肝疾患が代謝機能障害関連脂肪性肝疾患(MASLD)、成因不明脂肪性肝疾患(Cryptogenic SLD)又は特定成因脂肪性肝疾患(Specific aetiology SLD)である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is metabolic dysfunction-associated fatty liver disease (MASLD), cryptogenic fatty liver disease (Cryptogenic SLD), or specific aetiology fatty liver disease (Specific Aetiology SLD).  前記脂肪性肝疾患が非アルコール性脂肪肝炎(NASH)である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is nonalcoholic steatohepatitis (NASH).  前記脂肪性肝疾患が代謝機能障害関連脂肪肝炎(MASH)である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is metabolic dysfunction-associated steatohepatitis (MASH).  前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝線維症である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is hepatic fibrosis caused by NASH or MASH.  前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝硬変である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is cirrhosis caused by NASH or MASH.  前記脂肪性肝疾患がNASH又はMASHによって引き起こされる肝細胞癌(HCC)である、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein the fatty liver disease is hepatocellular carcinoma (HCC) caused by NASH or MASH.  前記医薬組成物の投与によって、止血異常、肝障害、腎障害及び胚致死性の少なくとも1つを伴わない、請求項1記載の医薬組成物。 The pharmaceutical composition according to claim 1, wherein administration of the pharmaceutical composition does not result in at least one of hemostatic abnormalities, liver damage, kidney damage, and embryonic lethality.
PCT/JP2024/043962 2023-12-13 2024-12-12 Medicine for treating steatotic liver diseases Pending WO2025127097A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2023-210184 2023-12-13
JP2023210184 2023-12-13

Publications (1)

Publication Number Publication Date
WO2025127097A1 true WO2025127097A1 (en) 2025-06-19

Family

ID=96057350

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2024/043962 Pending WO2025127097A1 (en) 2023-12-13 2024-12-12 Medicine for treating steatotic liver diseases

Country Status (1)

Country Link
WO (1) WO2025127097A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008093464A1 (en) * 2007-01-31 2008-08-07 Banyu Pharmaceutical Co., Ltd. Animal having modification in mgat2 gene
JP2022068347A (en) * 2017-07-14 2022-05-09 塩野義製薬株式会社 Condensation ring derivative with MGAT2 inhibitory activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008093464A1 (en) * 2007-01-31 2008-08-07 Banyu Pharmaceutical Co., Ltd. Animal having modification in mgat2 gene
JP2022068347A (en) * 2017-07-14 2022-05-09 塩野義製薬株式会社 Condensation ring derivative with MGAT2 inhibitory activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHENG DONG, ZINKER BRADLEY A., LUO YI, SHIPKOVA PETIA, DE OLIVEIRA CLAUDIA H., KRISHNA GOPAL, BROWN ELIZABETH A., BOEHM STEPHANIE : "MGAT2 inhibitor decreases liver fibrosis and inflammation in murine NASH models and reduces body weight in human adults with obesity", CELL METABOLISM, CELL PRESS, UNITED STATES, vol. 34, no. 11, 1 November 2022 (2022-11-01), United States , pages 1732 - 1748.e5, XP093323507, ISSN: 1550-4131, DOI: 10.1016/j.cmet.2022.10.007 *
TAKE KAZUMI, MOCHIDA TAISUKE, MAKI TOSHIYUKI, SATOMI YOSHINORI, HIRAYAMA MEGUMI, NAKAKARIYA MASANORI, AMANO NOBUYUKI, ADACHI RYUTA: "Pharmacological Inhibition of Monoacylglycerol O-Acyltransferase 2 Improves Hyperlipidemia, Obesity, and Diabetes by Change in Intestinal Fat Utilization", PLOS ONE, PUBLIC LIBRARY OF SCIENCE, US, vol. 11, no. 3, 3 March 2016 (2016-03-03), US , pages e0150976, XP093323524, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0150976 *
TURDI HUJI, CHAO HANNGUANG, HANGELAND JON J., AHMAD SALEEM, MENG WEI, BRIGANCE ROBERT, ZHAO GUOHUA, WANG WEI, MOORE FANG, YE XIANG: "Screening Hit to Clinical Candidate: Discovery of BMS-963272, a Potent, Selective MGAT2 Inhibitor for the Treatment of Metabolic Disorders", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 64, no. 19, 14 October 2021 (2021-10-14), US , pages 14773 - 14792, XP093323525, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.1c01356 *

Similar Documents

Publication Publication Date Title
JP7493635B2 (en) Fused ring derivatives having MGAT2 inhibitory activity
EP4400105A1 (en) Medicine for prevention and treatment of diseases linked to anti-obesity activity
EA013214B1 (en) APPLICATION OF THE PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF DIABETES, METABOLIC SYNDROME, NEURODEGENERATIVE DISEASES AND OBESITY
JP7068743B2 (en) A pharmaceutical composition containing a condensed ring derivative having MGAT2 inhibitory activity.
TW200843778A (en) Pterin analogs
WO2024063143A1 (en) Fused ring compound having glp-1 receptor agonist effect
WO2024063140A1 (en) Monocyclic compound having glp-1 receptor agonist activity
JP7608166B2 (en) Dihydropyrazolopyrazinone derivatives having MGAT2 inhibitory activity
WO2025127097A1 (en) Medicine for treating steatotic liver diseases
WO2017110841A1 (en) Non-aromatic heterocyclic derivative having mgat2 inhibitory activity
KR20230013126A (en) Medicines for the treatment of fatty liver disease
WO2018235785A1 (en) Dihydropyridone derivatives having MGAT2 inhibitory activity
HK40112712A (en) Medicine for prevention and treatment of diseases linked to anti-obesity activity
US20250041279A1 (en) Therapeutic medicine for heart disease or skeletal muscle disease
HK40059861A (en) Fused ring derivative having mgat-2 inhibitory activity
EP1609783A1 (en) Marine compounds with calcium channel blocking properties for Alzheimer's disease treatment
JP2024045832A (en) Pharmaceutical characterized by combining glp-1 receptor agonist and agent for treating diseases associated with anti-obesity actions
HK40059861B (en) Fused ring derivative having mgat-2 inhibitory activity

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: 24903750

Country of ref document: EP

Kind code of ref document: A1