CN109232538B - 1,2, 4-triazole compound - Google Patents

1,2, 4-triazole compound Download PDF

Info

Publication number
CN109232538B
CN109232538B CN201811159093.7A CN201811159093A CN109232538B CN 109232538 B CN109232538 B CN 109232538B CN 201811159093 A CN201811159093 A CN 201811159093A CN 109232538 B CN109232538 B CN 109232538B
Authority
CN
China
Prior art keywords
compound
deuterium
disease
pharmaceutically acceptable
compounds
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.)
Active
Application number
CN201811159093.7A
Other languages
Chinese (zh)
Other versions
CN109232538A (en
Inventor
王义汉
李焕银
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.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Priority to CN202010554939.8A priority Critical patent/CN111518080A/en
Publication of CN109232538A publication Critical patent/CN109232538A/en
Application granted granted Critical
Publication of CN109232538B publication Critical patent/CN109232538B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Obesity (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Transplantation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a 1,2, 4-triazole compound shown in a formula (I) and pharmaceutically acceptable salts thereof. The compounds have activity as inhibitors of apoptosis signal-regulated kinase 1 ("ASK 1"), and are therefore useful in the treatment of ASK1 mediated conditions including chronic liver disease, cardiovascular disease, metabolic disorders, respiratory disorders, gastrointestinal disorders, and neurodegenerative diseases. The invention also provides a pharmaceutical composition containing the compound and application thereof.

Description

1,2, 4-triazole compound
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a 1,2, 4-triazole compound, a composition containing the compound and application of the compound. In particular, the present invention relates to certain deuterium substituted 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methyl-N- [6- (4-isopropyl-4H-1, 2,4, -triazol-3-yl) -2-pyridine ] -benzamides, which are useful in the treatment of ASK1 mediated diseases and have superior pharmacokinetic properties.
Background
Many current drugs suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties, which prevents their wider use or limits their use for certain indications. Poor ADME properties are also a significant cause of drug candidate failure in clinical trials. One such problem is rapid metabolism, which results in the removal of many drugs from the body too rapidly, which would otherwise be highly effective in the treatment of disease. A common solution for rapid drug clearance is frequent or high dose administration to achieve sufficiently high plasma drug levels. However, this poses a number of potential therapeutic problems, such as poor patient compliance with the dosing regimen, side effects becoming more acute at higher doses, and increased treatment costs. Rapidly metabolized drugs may also expose patients to undesirable toxic or reactive metabolites.
Another limitation of ADME that affects many drugs is the formation of toxic or bioreactive metabolites. Thus, some patients receiving the drug may suffer from toxicity, or the safe dose of such a drug may be limited such that the patient receives a sub-optimal amount of the active agent. In some cases, varying the dosing interval or formulation method may help to reduce clinical adverse effects, but the formation of such undesirable metabolites is often inherent in the metabolism of the compound.
One potentially attractive strategy to improve the metabolic performance of drugs is deuterium modification. In this approach, attempts have been made to slow drug metabolism or reduce the formation of undesirable metabolites by replacing one or more hydrogen atoms with deuterium atoms. Deuterium is a safe, stable, non-radioactive isotope of hydrogen. Deuterium forms a stronger bond with carbon than hydrogen. In selected cases, the increased bond strength imparted by deuterium can positively affect the ADME properties of a drug, thereby creating the potential to improve drug efficacy, safety, and/or tolerability. Also, because the size and shape of deuterium is substantially the same as the size and shape of hydrogen, replacement of hydrogen by deuterium would not be expected to affect the biochemical potency and selectivity of the drug compared to the original chemical entity containing only hydrogen.
During the past 35 years, the effect of deuterium substitution on metabolic rate was reported for a very small percentage of approved drugs (see, e.g., Foster, AB, Adv Drug Res, 1985, 14:1-40 ("Foster"); Fisher, MB et al, Curr Opindrug Discov Devel, 2006, 9: 101-09 ("Fisher)). The results are variable and unpredictable. For some compounds, deuteration causes a decrease in metabolic clearance in vivo. For other compounds, there was no metabolic change. Still other compounds exhibit increased metabolic clearance. Variability in the deuterium effect has also led the skilled person to suspect or abandon a viable drug design strategy for deuterium modification as an inhibitor of adverse metabolism (see pages 35 of Foster and 101 of Fisher).
The effect of deuterium modification on the metabolic properties of a drug is not predictable, even in the case of deuterium atom penetration into known metabolic sites. One can only determine if and how the metabolic rate differs from its non-deuterated counterpart by actually preparing and testing a deuterated drug. Many drugs have multiple sites where metabolism can occur. The sites where deuterium substitution is required and the effect on metabolism, if any, seen, the degree of deuteration necessary will vary for each drug.
The present invention relates to novel derivatives of Selonsertib and pharmaceutically acceptable salts thereof. The invention also provides compositions comprising the compounds of the invention and the use of such compositions in methods of treatment of diseases and disorders beneficially treated by administration of an inhibitor of ASK1 (apoptosis signal-regulating kinase 1).
Seloserteib, also known as GS-4997 and chemically known as 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methyl-N- [6- (4-isopropyl-4H-1, 2,4, -triazol-3-yl) -2-pyridine ] -benzamide (having the structure shown below) is a small molecule inhibitor developed by Gilide pharmaceutical companies and can effectively reduce the pathological function of ASK 1. Results of a second phase clinical trial with Selonsertib in the treatment of nonalcoholic steatohepatitis (NASH) showed that anti-fibrotic effects were achieved in NASH patients only after 24 weeks. Currently, the study of Selonsertib for the treatment of NASH is in the third clinical stage, and the study for the treatment of alcoholic hepatitis is in the second clinical stage.
Figure GDA0002426852640000021
ASK1 activation and signaling have been reported to play an important role in a wide range of diseases including neurodegenerative, cardiovascular, inflammatory and metabolic disorders, in addition, ASK1 has been implicated in organ damage following mediation of ischemia and reperfusion of the heart, brain and kidneys (Watanabe et al (2005) BBRC 333, 562-567; Zhang et al (2003) L ifeSci 74-37-43; Terada et al (2007) BBRC 364: 1043-49).
Even though Selonsertib is already present, there is still a need for effective compounds with improved pharmacokinetic and/or pharmacodynamic behaviour in the treatment of diseases related to the activation of ASK 1.
Disclosure of Invention
Aiming at the technical problems, the invention discloses a 1,2, 4-triazole compound, a composition containing the compound and application thereof, wherein the compound has stronger ASK1 inhibitor activity.
In contrast, the invention adopts the following technical scheme:
in a first aspect, the present invention relates to a 1,2, 4-triazole compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure GDA0002426852640000031
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、Y3、Y4、Y5and Y6Each independently is from hydrogen or deuterium;
X1、X2and X3Each independently is CH3、CH2D、CHD2Or CD3
Provided that if X is1、X2And X3Each is CH3Then R1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、 Y3、Y4、Y5And Y6Is deuterium.
As a preferred embodiment of the present invention, the compound of formula (I) contains at least one deuterium atom, more preferably two deuterium atoms, more preferably three deuterium atoms, more preferably four deuterium atoms, more preferably six deuterium atoms, more preferably seven deuterium atoms, more preferably nine deuterium atoms.
As a preferred embodiment of the present invention, the deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
Specifically, in the present invention R1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、Y3、Y4、Y5、Y6、X1、 X2And X3The deuterium isotope content in each deuterated position is at least 5%, preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
In another embodiment, R of the compound of formula (I)1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、Y3、 Y4、Y5、Y6、X1、X2And X3At least one of them contains deuterium, preferably two contain deuterium, more preferably three contain deuteriumPreferably four deuterium containing groups, more preferably five deuterium containing groups, more preferably six deuterium containing groups, more preferably seven deuterium containing groups, more preferably eight deuterium containing groups, more preferably nine deuterium containing groups, more preferably ten deuterium containing groups, more preferably eleven deuterium containing groups, more preferably twelve deuterium containing groups, more preferably thirteen deuterium containing groups, more preferably fourteen deuterium containing groups, more preferably fifteen deuterium containing groups, more preferably sixteen deuterium containing groups, more preferably seventeen deuterium containing groups, more preferably eighteen deuterium containing groups, more preferably nineteen deuterium containing groups, more preferably twenty-one deuterium containing groups, more preferably twenty-two deuterium containing groups, more preferably twenty-three deuterium containing groups. In particular, the compounds of formula (I) contain at least one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three deuterium atoms.
As a preferred embodiment of the present invention, R1、R2、R3、R4、R5、R6、R7And R8Each independently from hydrogen or deuterium.
In another preferred embodiment, R1、R3And R4Is deuterium.
In another preferred embodiment, R5Is deuterium.
In another preferred embodiment, R1、R3、R4And R5Is deuterium.
As a preferred embodiment of the present invention, Y1、Y2、Y3、Y4、Y5And Y6Each independently from hydrogen or deuterium.
In another preferred embodiment, Y1Is deuterium.
In another preferred embodiment, Y2、Y3、Y4、Y5And Y6Is deuterium.
As a preferred embodiment of the present invention, X1、X2And X3Each independently is CH3、CH2D、CHD2Or CD3
In another preferred embodiment, X1、X2Is a CD3
In another preferred embodiment, X3Is a CD3
As a preferred embodiment of the present invention, the compound is selected from the group consisting of:
Figure GDA0002426852640000041
Figure GDA0002426852640000051
Figure GDA0002426852640000061
in another preferred embodiment, the compound does not include non-deuterated compounds.
In a second aspect of the present invention, the present invention also discloses a pharmaceutical composition comprising a pharmaceutically acceptable excipient and the 1,2, 4-triazole compound described above, or a pharmaceutically acceptable salt thereof.
In a third aspect of the present invention, the present invention also discloses a preparation method of the pharmaceutical composition as described above, comprising the following steps: mixing a pharmaceutically acceptable excipient with a 1,2, 4-triazole-type compound, or a pharmaceutically acceptable salt thereof, as described above, to form a pharmaceutical composition.
In another preferred embodiment, the pharmaceutical composition is an injection, a sachet, a tablet, a pill, a powder or a granule.
In another preferred embodiment, the pharmaceutical composition further comprises an additional therapeutic agent, wherein the additional therapeutic agent is an agent for treating cancer, cardiovascular disease, inflammation, infection, immune disease, metabolic disease or organ transplantation.
In a fourth aspect of the invention, there is also provided a method of treating a disease mediated at least in part by ASK1 in a patient in need thereof, comprising administering to the patient in need thereof a therapeutically effective amount of a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In particular embodiments, the diseases treatable by the compounds of the invention are selected from diabetes, diabetic nephropathy, renal disease, renal fibrosis, pulmonary fibrosis, Idiopathic Pulmonary Fibrosis (IPF), hepatic fibrosis, pulmonary hypertension, nonalcoholic steatohepatitis, liver disease, alcoholic liver disease, inflammatory disorders, autoimmune diseases, proliferative diseases, transplant rejection, diseases involving impairment of cartilage metabolism, congenital cartilage malformations, or diseases associated with hypersecretion of I L6.
In another preferred embodiment, the treatable disease is non-alcoholic steatohepatitis or alcoholic liver disease.
In another preferred embodiment, the treatable disease is pulmonary hypertension or pulmonary fibrosis.
In another preferred embodiment, the treatable disease is diabetic nephropathy, renal disease or renal fibrosis.
It is understood that within the scope of the present invention, each of the above-described technical features, embodiments of the present invention, and each of the technical features specifically described below (e.g., examples) may be combined with each other to constitute a new or preferred technical solution. Not to be reiterated herein, but to the extent of space.
Detailed Description
Compared with the non-deuterated compound, the compound of the deuterated ASK1 inhibitor and the pharmaceutically acceptable salt thereof have better pharmacokinetic and/or pharmacodynamic properties, so that the compound is more suitable to be used as an ASK1 inhibitor compound and is further more suitable to be used for preparing medicines for treating ASK 1-mediated related diseases. The present invention has been completed based on this finding.
Definition of
Herein, "deuterated", unless otherwise specified, means that one or more hydrogens of a compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
Herein, unless otherwise specified, "non-deuterated compound" means a compound containing deuterium at an atomic ratio of deuterium not higher than the natural deuterium isotope content (0.015%).
The invention also includes isotopically-labeled compounds, equivalent to those disclosed herein as the original compound. Examples of isotopes that can be listed as compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, respectively2H,3H,13C,14C,15N,17O,18O,31P,32P,35S,18F and36and (4) Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing such compounds are within the scope of the present invention. Certain isotopically-labelled compounds of the invention, e.g.3H and14among these, the radioactive isotope of C is useful in tissue distribution experiments of drugs and substrates. Tritium, i.e.3H and carbon-14, i.e.14C, their preparation and detection are relatively easy, and are the first choice among isotopes. Isotopically labeled compounds can be prepared by conventional methods by substituting readily available isotopically labeled reagents for non-isotopically labeled reagents using the protocols set forth in the examples.
Compound (I)
The present invention provides compounds of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof:
Figure GDA0002426852640000071
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、Y3、Y4、Y5、Y6each independently is from hydrogen or deuterium;
X1、X2、X3each independently is CH3、CH2D、CHD2Or CD3
Provided that if X is1、X2And X3Each is CH3Then R1、R2、R3、R4、R5、R6、R7、R8、Y1、Y2、 Y3、Y4、Y5And Y6Is deuterium.
As a preferred embodiment of the present invention, the deuterium isotope content of deuterium at the deuterium position is at least 0.015% greater than the natural isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
In a particular embodiment of formula (I), "R1、R2、R3、R4、R5、R6、R7And R8Each independently from hydrogen or deuterium "comprising R1Selected from hydrogen or deuterium, R2Selected from hydrogen or deuterium, R3Selected from hydrogen or deuterium, and so on, up to R8Selected from hydrogen or deuterium. More specifically, includes R1Is hydrogen or R1Is deuterium, R2Is hydrogen or R2Is deuterium, R3Is hydrogen or R3Deuterium, and so on, until R8Is hydrogen or R8Is a technical scheme of deuterium.
In another embodiment of formula (I), "Y1、Y2、Y3、Y4、Y5And Y6Each independently from hydrogen or deuterium "comprising Y1Selected from hydrogen or deuterium, Y2Selected from hydrogen or deuterium, Y3Selected from hydrogen or deuterium, and so on, up to Y6Selected from hydrogen or deuterium. More specifically, including Y1Is hydrogen or Y1Is deuterium, Y2Is hydrogen or Y2Is deuterium, Y3Is hydrogen or Y3Deuterium, and so on, up to Y6Is hydrogen or Y6Is a technical scheme of deuterium.
In another embodiment of formula (I), "X1、X2、X3Each independently is CH3、CH2D、CHD2Or CD3"includes X1Is selected from CH3、CH2D、CHD2Or CD3,X2Is selected from CH3、CH2D、CHD2Or CD3,X3Is selected from CH3、CH2D、 CHD2Or CD3The technical scheme of (1). More specifically, including X1Is CH3、X1Is CH2D、X1Is CHD2Or X1Is a CD3,X2Is CH3、X2Is CH2D、X2Is CHD2Or X2Is a CD3,X3Is CH3、X3Is CH2D、X3Is CHD2Or X3Is a CD3The technical scheme of (1).
In a preferred embodiment, the present invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate or solvate, crystal form, stereoisomer or isotopic variant thereof, wherein R is7And R8、Y2-Y6Is hydrogen, R1-R8And Y1Each independently of the other being hydrogen or deuterium, X1、X2And X3Each independently is CH3、CH2D、CHD2Or CD3With the proviso that said compound contains at least one deuterium atom.
In another preferred embodiment, R2And R6Is hydrogen.
In another preferred embodiment, R1、R3And R4Each independently selected from hydrogen or deuterium. In another more preferred embodiment, R1、R3And R4Is hydrogen. In another more preferred embodiment, R1、R3And R4Is deuterium.
In another preferred embodimentIn the scheme, R5Is hydrogen or deuterium. In another more preferred embodiment, R5Is hydrogen. In another more preferred embodiment, R5Is deuterium.
In another preferred embodiment, X3Is CH3
In another preferred embodiment, X1And X2Each independently selected from CH3Or CD3. In another more preferred embodiment, X1And X2Is CH3. In another more preferred embodiment, X1And X2Is a CD3
In another preferred embodiment, Y1Is hydrogen or deuterium. In another more preferred embodiment, Y1Is hydrogen. In another more preferred embodiment, Y1Is deuterium.
The term "pharmaceutically acceptable salts" refers, inter alia, to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977)66: 1-19. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Salts formed using methods conventional in the art, e.g., ion exchange methods, are also included. Other pharmaceutically acceptable salts include: adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cypionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoseAcid salts, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, lauryl sulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoate, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, valerates, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like. Other pharmaceutically acceptable salts include, if appropriate, non-toxic ammonium, quaternary ammonium and amine cations formed with counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by coordination of a compound of the present invention with water.
The term "prodrug" includes a class of compounds which may be biologically active or inactive in nature and which, when administered by an appropriate method, undergo a metabolic or chemical reaction in the human body to convert it to a compound of formula (I), or a salt or solution of a compound of formula (I). The prodrug includes (but is not limited to) carboxylate, carbonate, phosphate, nitrate, sulfate, sulfone ester, sulfoxide ester, amino compound, carbamate, azo compound, phosphoramide, glucoside, ether, acetal and other forms of the compound.
Pharmaceutical compositions and methods of administration
The compound has excellent activity of inhibiting ASK1 kinase, so the compound and various crystal forms, pharmaceutically acceptable salts, hydrates or solvates thereof, and a pharmaceutical composition containing the compound as a main active ingredient can be used for treating, preventing and relieving ASK1 mediated diseases. According to the prior art, the compounds of the invention are useful for the treatment of the following diseases: chronic liver diseases, cardiovascular diseases, metabolic disorders, respiratory disorders, gastrointestinal disorders, neurodegenerative diseases, and the like.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 0.5-2000mg of a compound of the invention per dose, more preferably, 1-500mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable excipient" refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compounds formulated together. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, duodenal, rectal, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or solubilizers, for example, starch, lactose, sucrose, glucose, mannitol, and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, e.g., paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, for example, cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert release agents conventionally employed in the art, such as water or other solvents, solubilizing agents and emulsifiers, for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, and oils, especially cottonseed, groundnut, corn germ, olive, castor and sesame oils, or mixtures of these materials.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 0.5-2000mg, preferably 1-500 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
Methods of treating diseases
Therapeutic agents useful as inhibitors of ASK1 signaling have the following potential: treating or improving the life of neurodegenerative, cardiovascular, inflammatory, autoimmune and metabolic disorders in a patient in need of treatment of the disease or disorder. In particular, ASK1 inhibitors have the following potential: treating cardiorenal diseases, including kidney diseases, diabetic nephropathy, chronic kidney diseases, fibrotic diseases (including lung and kidney fibrosis), dilated cardiomyopathy, respiratory diseases (including Chronic Obstructive Pulmonary Disease (COPD) and acute lung injury), and acute and chronic liver diseases (such as nonalcoholic steatohepatitis and alcoholic hepatitis).
Various assays for identifying the ability of compounds to inhibit ASK1 kinase activity and their effectiveness as ASK1 inhibitors are known in the art and are described, for example, in U.S. patent No. 8,742,126.
Some embodiments described herein relate to the use of a form of a compound described herein or a pharmaceutical composition described herein to treat a disease in a patient in need of treatment with an ASK1 inhibitor.
Some embodiments described herein are methods of treating diabetic nephropathy or diabetic complications comprising administering a therapeutically effective amount of a compound of formula (I) or a pharmaceutical composition described herein. In some embodiments, diabetes includes type 1 and type 2 diabetes, gestational diabetes, prediabetes, insulin resistance, metabolic syndrome, impaired fasting glucose, and impaired glucose tolerance. Type 1 diabetes is also known as Insulin Dependent Diabetes Mellitus (IDDM). Type 2 is also known as non-insulin-dependent diabetes mellitus (NIDDM).
Another embodiment relates to a method of treating kidney disease or diabetic nephropathy comprising administering a therapeutically effective amount of a compound of formula (I) as described herein or a pharmaceutical composition as described herein.
Another embodiment relates to a method of treating renal fibrosis, pulmonary fibrosis or Idiopathic Pulmonary Fibrosis (IPF) comprising administering a therapeutically effective amount of a form of compound I described herein or a pharmaceutical composition described herein.
Another embodiment relates to a method of treating diabetic nephropathy, renal fibrosis, liver fibrosis or lung fibrosis comprising administering a therapeutically effective amount of a crystalline form of compound I described herein or a pharmaceutical composition described herein.
Disclosed herein are methods of treating and/or preventing liver disease in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a form of compound I described herein, or a composition thereof, optionally in combination with a therapeutically effective amount of a L OX L inhibitor the presence of active liver disease can be detected by the presence of elevated enzyme levels in the blood, in particular, blood levels of alanine aminotransferase (a L T) and aspartate Aminotransferase (AST) known to be above the clinically acceptable normal range are indicative of ongoing liver injury.
In general, chronic liver disease can be caused by viruses (e.g., hepatitis B, hepatitis C, Cytomegalovirus (CMV), or Epstein Barr Virus (EBV)), toxic agents or drugs (e.g., alcohol, methotrexate, or nitrofurantoin), metabolic diseases (e.g., non-alcoholic steatohepatitis (NAF L D), non-alcoholic steatohepatitis (NASH), hemochromatosis, or Wilson's disease), autoimmune diseases (e.g., autoimmune chronic hepatitis, primary biliary cirrhosis, or primary sclerosing cholangitis), or other causes (e.g., right heart failure).
In one embodiment, the liver disease is non-alcoholic fatty liver disease (NAF L D). NAF L D is associated with insulin resistance and metabolic syndrome (obesity, mixed hyperlipidemia, diabetes (type II), and hypertension). NAF L D is believed to encompass a range of disease activities and begins with fat accumulation in the liver (hepatic steatosis).
The compounds of the present invention have a number of advantages over the non-deuterated compounds known in the prior art. The advantages of the invention include: firstly, the compound adopting the technical scheme of the invention has excellent inhibitory property on ASK1 protein kinase. Second, the metabolism of the compound in the organism is improved, giving the compound better pharmacokinetic parameters. In this case, the dosage can be varied and a long acting formulation formed, improving the applicability. Thirdly, the medicine concentration of the compound in the animal body is improved, and the medicine curative effect is improved. Fourth, certain metabolites are inhibited, increasing the safety of the compounds.
Examples
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Parts and percentages are parts and percentages by weight unless otherwise indicated.
In general, in the preparative schemes, each reaction is usually carried out in an inert solvent at a temperature ranging from room temperature to reflux temperature (e.g., from 0 ℃ to 100 ℃, preferably from 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Example 15- (4-cyclopropyl-1H-imidazolyl-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazole-3- 2Group-5-d) preparation of pyridin-2-yl-3, 4-d) -2-fluoro-4-methylbenzamide (Compound T-1).
Figure GDA0002426852640000131
The specific synthesis steps are as follows:
Figure GDA0002426852640000132
step 1 synthesis of compound 2.
Compound 1(5.0g, 32.86mmol) and methanol (60M L) are added to a 100M L single-neck flask equipped with magnetic stirring in sequence, stirred to dissolve, hydrazine hydrate (3.29g, 65.72mmol) is slowly added dropwise, after dropping, the reaction mixture is heated under reflux for 3 hours, then cooled to room temperature, a large amount of white solid is precipitated, filtered, the filter cake is washed with cold methanol, and dried to obtain 3.5g of the white solid with a yield of 70%. L C-MS (APCI): M/z is 153.2(M +1)+.1H NMR(DMSO-d6,300MHz)(/ppm):9.14(s,1H),7.51(t,J=5.7Hz, 1H),7.11(d,J=5.7Hz,1H),6.61(d,J=6.0Hz,1H),6.08(s,2H),4.48(s,2H).
Step 2 synthesis of compound 3.
Sequentially burning the mixture into a single port of 100m L equipped with magnetic stirringA flask was charged with Compound 2(3.5g, 23mmol), toluene (Tol, 50m L), isopropylamine (9.8g, 166mmol) and N, N-dimethylformamide-dipropylacetal (DMF-DPA, 10.89g, 62mmol), and acetic acid (2.1g, 35mmol) was added slowly dropwise with stirring, after addition, N2Refluxing the reaction mixture under reflux for 24 hr, cooling to room temperature, evaporating under reduced pressure to remove solvent, adding water (40M L), stirring to precipitate a large amount of solid, filtering, washing the filter cake with isopropanol to obtain 3.1g of white solid with yield of 66.3%. L C-MS (APCI): M/z is 204.2(M +1)+.1H NMR(CDCl3,300MHz)(/ppm): 8.31(s,1H),7.58-7.54(m,2H),6.56(dd,J1=5.4Hz,J2=1.5Hz,1H),5.64-5.57(m,1H),4.45(s,2H),1.52(d, J=4.8Hz,6H).
Step 3 synthesis of compound 4.
Adding compound 3(200mg, 984umol), heavy water (10M L) and Pd/C (50 mg, 10%) into a 50M L sealed tube with magnetic stirring, bubbling with hydrogen for 5min under stirring, sealing, heating to 110 ℃, keeping the temperature and stirring for reaction overnight, cooling to room temperature, adding dichloromethane (20M L), filtering, layering, extracting with aqueous dichloromethane (20M L x2), combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating, separating by a silica gel column to obtain 120mg of white solid, yield 59.1%, L C-MS (APCI), M/z 207.3(M +1)+.1H NMR(CDCl3,300MHz)(/ppm):7.56(s,1H),5.62-5.59(m,1H),4.50(s,1H),1.51(d,J=5.4Hz,6H).
Step 4 synthesis of compound 7.
Compound 6(5.0g, 24.5mmol), toluene (50m L) were added to a 100m L single neck flask equipped with magnetic stirring in succession, the mixture was dissolved by stirring, compound 5(4.4g, 27.0mmol) and N, N-diisopropylethylamine (DIPEA,8.5m L, 51.5mmol) were added dropwise, the reaction mixture was heated under reflux for 2 hours, cooled to room temperature, water (50m L) was added, the layers were separated, and the organic phase was successively treated with saturated NH in succession4Cl water solution (20m L), saturated NaHCO3Washing water solution (20m L) and saturated saline (20m L), drying with anhydrous sodium sulfate, filtering, concentrating, recrystallizing the residue in n-hexane, filtering, and oven drying to obtain white solid (3.1g, yield 44.1%. L C-MS (APCI): m/z 286.2&288.2(M+1)+.1H NMR(DMSO-d6,300MHz)(/ppm):7.07(d,J=6.9Hz,1H), 6.52(d,J=4.5Hz,1H),5.29(t,J=4.2Hz,1H),4.18(d,J=4.2Hz,2H),4.52-4.51(m,1H),2.11(s,3H), 0.98-0.88(m,4H).
Step 5 synthesis of compound 8.
Compound 7(3.1g, 10.8mmol) and dichloromethane (DCM, 15m L) were added sequentially to a 50m L one-neck flask equipped with magnetic stirring, stirred to dissolve and cool to 0 deg.C, and formic acid (15m L) and acetic anhydride (Ac) were added slowly dropwise2O, 4.1m L, 43.3mmol), reaction solution N2Stirring the reaction at 0 ℃ for 3 hours under an atmosphere, adding water (20m L) to quench the reaction, adjusting the pH to 9 with 40% aqueous NaOH, separating the organic phase, extracting the aqueous phase with dichloromethane (30m L x2), combining the organic phases, drying over anhydrous sodium sulfate, filtering, and using the concentrated solution directly in the next step, L C-MS (APCI): m/z 314.1&316.1(M+1)+.1H NMR(DMSO-d6,300MHz)(/ppm): 8.15(d,J=9.6Hz,1H),7.61(d,J=5.4Hz,1H),7.41(d,J=7.2Hz,1H),4.68(d,2H),2.50(s,3H), 2.12-1.98(m,1H),0.96-0.85(m,4H).
Step 6 synthesis of compound 9.
A50 m L one-neck flask equipped with magnetic stirring was charged with Compound 8(3.39g, 10.8mmol) and glacial acetic acid (30m L) in sequence, the mixture was dissolved by stirring, and ammonium acetate (2.57g, 42.1mmol), N2The reaction mixture was heated under reflux overnight under ambient temperature, the acetic acid was evaporated under reduced pressure, water (20M L), 40% aqueous NaOH was added to adjust the pH to 9, dichloromethane was extracted (30M L X3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated and the residue was isolated on a silica gel column to give 1.7g of a white solid with a yield of 53.4% L C-MS (APCI) where M/z is 295.1 and 297.1(M +1)+.1H NMR(CDCl3,300MHz)(/ppm):7.41(d,J=4.8Hz,1H), 7.37(s,1H),7.07(d,J=6.6Hz,1H),6.72(s,1H),2.14(s,3H),1.90-1.85(m,1H),0.90-0.78(m,4H).
Step 7 synthesis of compound 10.
To a 50m L three-necked flask equipped with magnetic stirring was added compound 9(1.7g, 5.8mmol), vacuum was applied and N was added2Substitution 3 times, N2Anhydrous THF (30m L) was added dropwise under an atmosphere, the solution was stirred clear, cooled to 0 ℃, isopropyl magnesium chloride (4.3m L, 8.6mmol,2M), stirring and reacting for 1 hour under heat preservation, and then filling with CO2Slowly introducing CO into the reaction liquid by a balloon of gas2Reacting for 1 hour, adding water (20M L) to quench the reaction, adjusting pH to 5 with 6M HCl aqueous solution, extracting with ethyl acetate (30M L x3), combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating, recrystallizing the residue with diethyl ether to obtain 600mg of white solid with a yield of 40.0%, L C-MS (APCI) M/z 261.1(M +1)+.1H NMR(DMSO-d6,300MHz)(/ppm):9.17(s,1H),7.95(d,J=4.8Hz, 1H),7.70(s,1H),7.52(d,J=8.4Hz,1H),2.24(s,3H),2.05-1.91(m,1H),1.01-0.85(m,4H).
Step 8 Synthesis of Compound T-1.
To a 50m L three-necked flask equipped with magnetic stirring was added compound 10(100mg, 0.384mmol), vacuum was applied and N was added2Substitution of N2Dropping dried dichloromethane (2M L) and DMF (2M L) via syringe under the atmosphere, stirring for dissolving, cooling the reaction solution to 0 deg.C, slowly dropping oxalyl chloride (0.33M L, 0.653mmol, 2M dichloromethane solution), removing ice bath, stirring at room temperature for 1 hr, evaporating under reduced pressure to remove solvent, adding dried dichloromethane (10M L), N2Stirring the solution under an atmosphere, cooling to 0 ℃, slowly dropwise adding a dichloromethane solution (2M L) of compound 4(94mg, 0.461mmol), then dropwise adding DIPEA (0.19M L, 1.15mmol), removing the ice bath, stirring at room temperature for 2 hours, adding water (20M L) to quench the reaction, separating an organic layer, extracting an aqueous phase with dichloromethane (20M L x2), combining the organic phases, drying with anhydrous sodium sulfate, filtering, concentrating, separating the residue by a silica gel column to obtain 60mg of an off-white solid, wherein the yield is 35.1%. L C-MS (APCI): M/z is 446.4(M +1)+.1H NMR(CDCl3,300MHz)(/ppm): 9.06(d,J=6.0Hz,1H),8.40-8.36(m,2H),8.07-8.05(m,2H),7.91(t,J=6.0Hz,1H),7.45(s,1H),7.18(d, J=9.0Hz,1H),6.78(s,1H),5.51-5.44(m,1H),2.28(s,3H),1.91-1.88(m,1H),1.59(d,J=5.1Hz,6H), 0.90-0.81(m,4H).
Example 2 5- (4-cyclopropyl-1H-imidazolyl-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazole-3-) Yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide-6-d (Compound T-2).
Figure GDA0002426852640000161
The specific synthesis steps are as follows:
Figure GDA0002426852640000162
step 1 synthesis of compound 12.
Adding compound 6(2.0g, 9.8mmol) and heavy water (10M L) into a 20M L microwave tube equipped with magnetic stirring, slowly adding DCl heavy water solution (0.817M L, 9.8mmol, 12M) under stirring, heating the reaction mixture to 160 deg.C under microwave, reacting for 1.5 hr, cooling to room temperature, and adding saturated NaHCO3Adjusting pH to 10, extracting with dichloromethane (20M L X3), combining the organic phases, drying over anhydrous sodium sulfate, filtering, and concentrating to obtain a brown solid 1.59g, yield 79.1%. L C-MS (APCI): M/z ═ 205.1(M +1)+.1H NMR(DMSO-d6,300MHz)(/ppm):6.93(d,J=7.2Hz,1H),4.94(s,2H),1.99(s,3H).
Step 2 synthesis of compound 13.
Compound 12(1.59g, 7.75mmol), toluene (30m L) were added to a 100m L single neck flask equipped with magnetic stirring in sequence, the mixture was dissolved by stirring, compound 5(1.39g, 8.53mmol) and DIPEA (2.7m L, 16.28mmol) were added dropwise, the reaction was heated under reflux for 2 hours, cooled to room temperature, water (30m L) was added, the layers were separated, and the organic phase was successively treated with saturated NH4Cl water solution (10m L), saturated NaHCO3Washing water solution (10m L) and saturated saline (10m L), drying with anhydrous sodium sulfate, filtering, concentrating, recrystallizing the residue in n-hexane, filtering, and oven drying to obtain white solid 1.5g, with yield 67.4%. L C-MS (APCI) m/z 287.2&289.2(M+1)+.
Step 3 synthesis of compound 14.
Compound 13(1.5g, 5.22mmol) and dichloromethane (10m L) were added sequentially to a 50m L one-neck flask equipped with magnetic stirring, stirred to dissolve and cool to 0 deg.C, formic acid (15m L) and acetic anhydride (2.1m L, 20.9mmol) were added slowly dropwise, reaction N2Stirring the mixture at 0 ℃ under an atmosphere for 3 hours, adding water (10m L) to quench the reaction, and mixing the solution to obtain a mixtureAdjusting pH to 9 with NaOH aqueous solution, separating organic phase, extracting aqueous phase with dichloromethane (20m L X2), combining organic phases, drying with anhydrous sodium sulfate, filtering, and concentrating directly for next reaction L C-MS (APCI): m/z 315.1&317.1(M+1)+.
Step 4 synthesis of compound 15.
Add Compound 14(1.65g, 5.24mmol) and glacial acetic acid (15m L) sequentially to a 50m L single neck flask equipped with magnetic stirring, stir to dissolve, add ammonium acetate (1.25g, 20.5mmol), N2The reaction was refluxed overnight under an atmosphere, cooled to room temperature, acetic acid was evaporated under reduced pressure, water (10M L) was added, the pH was adjusted to 9 with 40% aqueous NaOH, dichloromethane was extracted (20M L X3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the residue was separated on a silica gel column to give 1.7g of a white solid with a yield of 53.4% L C-MS (APCI) where M/z is 296.1 and 298.1(M +1)+.1H NMR(CDCl3,300MHz)(/ppm):7.38(s,1H),7.09(d,J=6.9Hz,1H),6.73(s,1H), 2.15(s,3H),1.91-1.87(m,1H),0.91-0.78(m,4H).
Step 5 synthesis of compound 16.
To a 50m L three-necked flask equipped with magnetic stirring was added compound 15(820mg, 2.77mmol), vacuum was applied and N was added2Substitution 3 times, N2Anhydrous THF (15M L) was added dropwise under an atmosphere, the solution stirred to clear, cooled to 0 deg.C, isopropyl magnesium chloride (2.77M L, 5.54mmol, 2M) was added slowly dropwise, the reaction was stirred at constant temperature for 1 hour, and then the reaction mixture was passed through a CO-flood2Slowly introducing CO into the reaction liquid by a balloon of gas2After 1 hour of reaction, water (20M L) was added to quench the reaction, the pH was adjusted to 5 with 6M aqueous HCl, ethyl acetate was extracted (30M L X3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated, and the residue was recrystallized from ether to give 200mg of a white solid, yield 27.6%. L C-MS (APCI): M/z ═ 262.1(M +1)+.1H NMR(DMSO-d6,300MHz)(/ppm):8.69(s,1H),7.53(s,1H), 7.50(d,J=8.7Hz,1H),2.24(s,3H),1.96-1.93(m,1H),0.96-0.93(m,2H),0.80-0.77(m,2H).
Step 6 Synthesis of Compound T-2.
To a 50m L three-necked flask equipped with magnetic stirring, compound 16(130mg, 0.497mmol) was added, vacuum pulled, and N2Substitution three times, N2Dropping dried dichloromethane (2M L) and DMF (2M L) via syringe, stirring, cooling to 0 deg.C, slowly dropping oxalyl chloride (0.42M L, 0.846mmol, 2M dichloromethane solution), removing ice bath, stirring at room temperature for 1 hr, evaporating under reduced pressure to remove solvent, adding dried dichloromethane (10M L), N, and adding N2Stirring the solution to clear under an atmosphere, cooling to 0 ℃, slowly dropwise adding a dichloromethane solution (2M L) of compound 3(122mg, 0.597mmol), then dropwise adding DIPEA (0.25M L, 1.49mmol), removing the ice bath, stirring at room temperature for 2 hours, adding water (20M L) to quench the reaction, separating an organic layer, extracting an aqueous phase with dichloromethane (20M L x2), combining the organic phases, drying over anhydrous sodium sulfate, filtering, concentrating, separating the residue by a silica gel column to obtain 60mg of an off-white solid, wherein the yield is 35.1%. L C-MS (APCI): M/z is 447.4(M +1)+.1H NMR(CDCl3,300MHz)(/ppm): 9.06(d,J=12.3Hz,1H),8.41-8.38(m,2H),8.07(d,J=5.7Hz,1H),7.93(t,J=6.0Hz,1H),7.49(s,1H),7.20(d, J=9.6Hz,1H),6.78(s,1H),5.52-5.46(m,1H),2.29(s,3H),2.00-1.88(m,1H),1.59(d,J=5.1Hz,6H), 0.91-0.81(m,4H).
6Example 35- (4-cyclopropyl-1H-imidazolyl-1-yl) -N- (6- (4- (prop-2-yl-1, 1,1,3,3,3-d) Preparation of 4H-1,2, 4-triazol-3-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide (Compound T-3).
Figure GDA0002426852640000181
The specific synthesis steps are as follows:
Figure GDA0002426852640000182
step 1 synthesis of compound 18.
To a 50m L single neck flask equipped with magnetic stirring was added ammonium acetate (6.32g, 78mmol) and MeOD (50m L), N2Heating and refluxing for 3 h under the atmosphere, concentrating under reduced pressure to dryness, adding MeOD (10m L), adding acetone-d under stirring6(1.0g, 15.6mmol) and NaBH3CN(980mg,15.6mmol),N2Under the atmosphereThe reaction was stirred at room temperature overnight, quenched by addition of water (10M L), pH adjusted to 2 with 6M hydrochloric acid, extracted with ethyl acetate (20M L X2), adjusted to 12 with aqueous 6M NaOH, extracted with dichloromethane (20M L X3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, adjusted to pH 2 with 2M methanolic hydrochloric acid, concentrated to dryness to give compound 18 hydrochloride, which was used directly in the next step.
Step 2 synthesis of compound 19.
Compound 2(300mg, 1.97mmol), toluene (10m L), compound 18 hydrochloride (642mg, 9.86mmol) and N, N-dimethylformamide-dipropyl acetal (933mg, 5.32mmol) were added successively to a 50m L single-neck flask equipped with magnetic stirring, and acetic acid (296mg, 4.93mmol) was slowly added dropwise with stirring, after addition, N2The reaction mixture was refluxed for 3 hours under an atmosphere, cooled to room temperature, the solvent was distilled off under reduced pressure, and the white solid was obtained as 170mg by passing through a silica gel column in 41.2% yield L C-MS (APCI) where M/z is 210.2(M +1)+.1H NMR(CDCl3,300MHz)(/ppm):8.32(s,1H),7.62-7.56(m,2H),6.58(dd,J1=5.1Hz,J2=1.2Hz,1H), 5.59(s,1H),4.51(s,2H).
Step 3 Synthesis of Compound T-3.
To a 50m L three-necked flask equipped with magnetic stirring, compound 10(130mg, 0.497mmol) was added, vacuum was applied and N was added2Substitution of N2Dropping dried dichloromethane (2M L) and DMF (2M L) via syringe, stirring to dissolve, cooling the reaction solution to 0 deg.C, slowly dropping oxalyl chloride (0.42M L, 0.846mmol, 2M dichloromethane solution), removing ice bath, stirring at room temperature for 1 hr, evaporating under reduced pressure to remove solvent, adding dried dichloromethane (10M L), and adding N2Stirring the solution to clear under an atmosphere, cooling to 0 ℃, slowly dropwise adding a dichloromethane solution (2M L) of compound 19(122mg, 0.597mmol), then dropwise adding DIPEA (0.25M L, 1.49mmol), removing the ice bath, stirring at room temperature for 2 hours, adding water (20M L) to quench the reaction, separating an organic layer, extracting an aqueous phase with dichloromethane (20M L x2), combining the organic phases, drying over anhydrous sodium sulfate, filtering, concentrating, separating the residue by a silica gel column to obtain 60mg of an off-white solid, wherein the yield is 35.1%. L C-MS (APCI): M/z is 452.4(M +1)+.1H NMR(CDCl3,300MHz)(/ppm): 9.05(d,J=11.7Hz,1H),8.39-8.35(m,2H),8.07-8.04(m,2H),7.91(t,J=6.6Hz,1H),7.45(s,1H),7.18(d, J=9.0Hz,1H),6.77(s,1H),5.43(s,1H),2.27(s,3H),1.94-1.90(m,1H),0.90-0.80(m,4H).
7Example 45- (4-cyclopropyl-1H-imidazolyl-1-yl) -N- (6- (4- (prop-2-yl-d) -4H-1,2, 4-tris Preparation of oxazol-3-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide (compound T-4).
Figure GDA0002426852640000191
The specific synthesis steps are as follows:
Figure GDA0002426852640000192
step 1 synthesis of compound 20.
To a 50m L single neck flask equipped with magnetic stirring was added ammonium acetate (6.32g, 78mmol) and MeOD (50m L), N2Heating and refluxing for 3 h under the atmosphere, concentrating under reduced pressure to dryness, adding MeOD (10m L), adding acetone-d under stirring6(1.0g, 15.6mmol) and NaBD3CN(980mg,15.6mmol),N2The reaction was stirred at room temperature overnight under an atmosphere, quenched by addition of water (10M L), pH adjusted to 2 with 6M hydrochloric acid, extracted with ethyl acetate (20M L x2), pH adjusted to 12 with aqueous 6M NaOH, extracted with dichloromethane (20M L x3), the organic phases combined, dried over anhydrous sodium sulfate, filtered, pH adjusted to 2 with 2M methanol hcl, concentrated to dryness to give compound 20 hydrochloride, which was used directly in the next step.
Step 2 synthesis of compound 21.
Compound 2(300mg, 1.97mmol), toluene (10m L), compound 20 hydrochloride (642mg, 9.86mmol) and N, N-dimethylformamide-dipropyl acetal (933mg, 5.32mmol) were added successively to a 50m L single-neck flask equipped with magnetic stirring, and acetic acid (296mg, 4.93mmol) was slowly added dropwise with stirring, after addition, N2The reaction mixture was refluxed for 3 hours under an atmosphere, cooled to room temperature, the solvent was distilled off under reduced pressure, and the white solid was passed through a silica gel column to give 170mg of a 41.2% yield L C-MS (APCI) where M/z is 211.2(M +1)+.
Step 3 Synthesis of Compound T-4.
To a 50m L three-necked flask equipped with magnetic stirring, compound 10(130mg, 0.497mmol) was added, vacuum was applied and N was added2Substitution of N2Dropping dried dichloromethane (2M L) and DMF (2M L) via syringe, stirring to dissolve, cooling the reaction solution to 0 deg.C, slowly dropping oxalyl chloride (0.42M L, 0.846mmol, 2M dichloromethane solution), removing ice bath, stirring at room temperature for 1 hr, evaporating under reduced pressure to remove solvent, adding dried dichloromethane (10M L), and adding N2The solution was stirred clear under atmosphere, cooled to 0 ℃ and a solution of compound 21(122mg, 0.597mmol) in dichloromethane (2M L) was slowly added dropwise, then DIPEA (0.25M L, 1.49mmol) was added dropwise, the ice bath was removed and the reaction was stirred at room temperature for 2 hours, water (20M L) was added to quench the reaction, the organic layer was separated, the aqueous phase was extracted with dichloromethane (20M L x2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, concentrated and the residue was separated on a silica gel column to give 60mg of an off-white solid in 35.1%. L C-ms (apci): M/z 453.4(M +1)+.1H NMR(CDCl3,300MHz)(/ppm): 9.06(d,J=12.0Hz,1H),8.41(d,J=6.9Hz,1H),8.38(s,1H),8.09(d,J=5.7Hz,2H),7.94(t,J=6.0Hz,1H), 7.47(s,1H),7.21(d,J=9.3Hz,1H),6.80(s,1H),2.31(s,3H),1.98-1.88(m,1H),0.92-0.84(m,4H).
And (4) testing the biological activity.
(1) Kinase inhibition
Reagents and consumables:
ASK1(Invitrogen catalog number PV3809), ATP (Sigma, catalog number A7699), DMSO (Sigma, catalog number D8418-1L), 384 well plates (Greiner, catalog number 784076), HTRF KinEASE-STK Kit (Cisbio, catalog number PV3809), 5 xkinase buffer A (L if Technologies, catalog number PV3186), kinase tracer 199 (L if Technologies, catalog number PV5830),
Figure GDA0002426852640000211
Eu-anti-GST antibody (L if Technologies, Cat. No. PV 5594).
The specific experimental method comprises the following steps:
compound preparation: test compounds were dissolved in DMSO to make 20mM stock. Then, the cells were diluted ten times in DMSO with a gradient of 3-fold. When adding medicine, the medicine is diluted by 10 times by using buffer solution.
ASK1 kinase assay: ASK1 kinase was mixed with pre-diluted formulations of compounds at different concentrations for 10 minutes in 5x kinase buffer a, each concentration being duplicate wells. Adding corresponding substrate and ATP, reacting for 20 minutes at room temperature (wherein negative and positive controls: negative is blank control, and positive is erlotinib). Adding a detection reagent (reagent in an HTRF KinEASE-STK Kit) after the reaction is finished, incubating for 30 minutes at room temperature, detecting by an Evnvision microplate reader, determining the enzyme activity of the compound in the presence of each concentration, calculating the inhibitory activity of the compound with different concentrations on the enzyme activity, fitting the inhibitory activity of the compound with different concentrations on the enzyme activity according to a four-parameter equation and Graphpad 5.0 software, and calculating IC50The value is obtained.
The compounds of the invention and the non-deuterated compound, Selonsertib, were tested in the kinase inhibition assay described above and found to have more potent or comparable activity against ASK1 kinase. The results of the inhibition of kinases by representative example compounds are summarized in table 1 below.
TABLE 1
EXAMPLES Compounds ASK1 IC50(nM)
Selonsertib 1.10
T-1 1.00
T-2 1.12
T-3 1.12
T-4 1.03
(2) Metabolic stability evaluation
Microsome assay human liver microsome 0.5mg/m L, Xenotech, rat liver microsome 0.5mg/m L, Xenotech, mouse liver microsome 0.5mg/m L, Xenotech, coenzyme (NADPH/NADH) 1mM, Sigma L ifeCIENCE, magnesium chloride 5mM, 100mM phosphate buffer (pH 7.4).
Preparing a stock solution: an amount of the compound of example was weighed out finely and dissolved in DMSO to 5mM each.
Phosphate buffer (100mM, pH7.4) was prepared by mixing 150M L M of 0.5M potassium dihydrogenphosphate and 700M L M of 0.5M dipotassium hydrogenphosphate, adjusting the pH of the mixture to 7.4 with 0.5M dipotassium hydrogenphosphate solution, diluting 5-fold with ultrapure water before use, and adding magnesium chloride to obtain 100mM phosphate buffer (100mM) containing 100mM potassium phosphate and 3.3mM magnesium chloride and having a pH of 7.4.
NADPH regenerating system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U/m L G-6-P D, 3.3mM magnesium chloride) was prepared and placed on wet ice before use.
Preparing a stop solution, namely adding acetonitrile solution containing 50ng/m L propranolol hydrochloride and 200ng/m L tolbutamide (internal standard) into a 25057.5 mu L phosphate buffer solution (pH7.4) to a 50m L centrifuge tube, respectively adding 812.5 mu L human, rat and mouse liver microsomes, and uniformly mixing to obtain a liver microsome diluent with a protein concentration of 0.625mg/m L, incubating a sample, namely respectively diluting a stock solution of a corresponding compound to 0.25mM by using an aqueous solution containing 70% acetonitrile to serve as a working solution, respectively adding 398 mu L human liver microsomes, rat liver microsomes or mouse liver microsome diluent into a 96-well incubation plate (N is 2), respectively adding 2 mu L0.25 to 0.25mM working solution, and uniformly mixing.
Adding 300 mu L precooled stop solution into each well of a 96-well deep-well plate, placing the plate on ice to serve as a stop plate, placing a 96-well incubation plate and an NADPH regeneration system in a 37 ℃ water bath box, oscillating at 100 rpm, pre-incubating for 5min, taking 80 mu L incubation solution out of each well of the incubation plate, adding the stop plate, uniformly mixing, supplementing 20 mu L NADPH regeneration system solution to serve as a 0min sample, adding 80 mu L NADPH regeneration system solution into each well of the incubation plate, starting reaction, starting timing, taking the reaction concentration of a corresponding compound to be 1 mu M and the protein concentration to be 0.5mg/M L, respectively taking 100 mu L reaction solution when the reactions are 10, 30 and 90min, adding the 100 mu L reaction solution into the stop plate, vortexing for 3min to stop the reaction, centrifuging for 10min at 4 ℃, taking 100 mu L supernatant to 96 in which 100 mu L distilled water is added in advance, uniformly mixing, and analyzing the sample by adopting L C-MS.
Data analysis by detecting peak areas of the corresponding compound and internal standard by L C-MS/MS system, calculating the peak area ratio of the compound to the internal standard, measuring the slope by plotting the natural logarithm of the percentage of the remaining amount of the compound against time, and calculating t according to the following formula1/2And C LintWhere V/M is equal to 1/protein concentration.
Figure GDA0002426852640000221
t1/2(min);CLint(μL/min/mg)。
The compounds of the present invention and the non-deuterated compound Selonsertib were simultaneously tested and compared to evaluate their metabolic stability in human, rat and mouse liver microsomes. The half-life and intrinsic hepatic clearance as indicators of metabolic stability are shown in table 2. As shown in table 2, the compounds of the present invention can significantly improve metabolic stability by comparison with the non-deuterated compound Selonsertib in human, rat and mouse liver microsome experiments.
TABLE 2
Figure GDA0002426852640000231
(3) Pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 per group and compared for pharmacokinetic differences by intravenous or oral administration of a single dose of compound (10 mg/kg oral).
Rats were fed with standard feed and given water. Fasting began 16 hours prior to the experiment. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L blood samples were collected from the orbit in test tubes containing 30 μ L1% heparin salt solution before use, the tubes were dried overnight at 60 ℃ before use.
After blood collection, the tubes were gently inverted at least 5 times immediately to ensure adequate mixing and placed on ice, the blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate plasma from erythrocytes, 100 μ L plasma was pipetted into a clean plastic centrifuge tube indicating the name of the compound and the time point, the plasma was stored at-80 ℃ prior to analysis, the concentration of the compound of the invention in the plasma was determined using L C-MS/MS.
Experiments show that the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and treatment effect.
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (10)

1. A compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
Figure FDA0002469442850000011
2. a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of claim 1 or a pharmaceutically acceptable salt thereof.
3. A method of preparing the pharmaceutical composition of claim 2, comprising: mixing a pharmaceutically acceptable excipient with the compound of claim 1, or a pharmaceutically acceptable salt thereof, to form a pharmaceutical composition.
4. Use of a compound of claim 1 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition of claim 2 or 3 in the manufacture of a medicament for the treatment of a disease mediated at least in part by ASK 1.
5. The use of claim 4, further comprising administering another therapeutic agent.
6. The use of claim 5, wherein the other therapeutic agent is L OX L2 inhibitor.
7. The use according to any one of claims 4 to 6, wherein the disease is diabetes, renal disease, pulmonary fibrosis, pulmonary hypertension, liver disease, autoimmune disease, proliferative disease, transplant rejection, a disease involving impairment of cartilage metabolism, congenital cartilage malformations, or a disease associated with hypersecretion of I L6.
8. The use according to claim 7, wherein the liver disease is nonalcoholic steatohepatitis, alcoholic liver disease, or liver fibrosis.
9. The use of claim 7, wherein the disease is pulmonary hypertension or pulmonary fibrosis.
10. The use of claim 7, wherein the renal disease is diabetic nephropathy or renal fibrosis.
CN201811159093.7A 2017-10-12 2018-09-30 1,2, 4-triazole compound Active CN109232538B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010554939.8A CN111518080A (en) 2017-10-12 2018-09-30 1,2, 4-triazole compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2017109453180 2017-10-12
CN201710945318 2017-10-12

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202010554939.8A Division CN111518080A (en) 2017-10-12 2018-09-30 1,2, 4-triazole compound

Publications (2)

Publication Number Publication Date
CN109232538A CN109232538A (en) 2019-01-18
CN109232538B true CN109232538B (en) 2020-07-24

Family

ID=65054891

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201811159093.7A Active CN109232538B (en) 2017-10-12 2018-09-30 1,2, 4-triazole compound
CN202010554939.8A Pending CN111518080A (en) 2017-10-12 2018-09-30 1,2, 4-triazole compound

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202010554939.8A Pending CN111518080A (en) 2017-10-12 2018-09-30 1,2, 4-triazole compound

Country Status (2)

Country Link
CN (2) CN109232538B (en)
WO (1) WO2019072130A1 (en)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI491606B (en) * 2009-07-13 2015-07-11 Gilead Sciences Inc Apoptosis signal-regulating kinase inhibitors
JP5705314B2 (en) * 2010-07-02 2015-04-22 ギリアード サイエンシーズ, インコーポレイテッド Apoptosis signal-regulating kinase inhibitor
UY34573A (en) * 2012-01-27 2013-06-28 Gilead Sciences Inc QUINASE INHIBITOR REGULATING THE APOPTOSIS SIGNAL
NZ720660A (en) * 2013-12-20 2018-01-26 Gilead Sciences Inc Apoptosis signal-regulating kinase inhibitors
MA41252A (en) * 2014-12-23 2017-10-31 Gilead Sciences Inc SOLID FORMS OF AN ASK 1 INHIBITOR
CA2972192C (en) * 2014-12-23 2021-07-06 Gilead Sciences, Inc. Processes for preparing ask1 inhibitors
CN109983007B (en) * 2017-03-03 2021-11-05 江苏豪森药业集团有限公司 Amide derivative inhibitor and preparation method and application thereof

Also Published As

Publication number Publication date
WO2019072130A1 (en) 2019-04-18
CN109232538A (en) 2019-01-18
CN111518080A (en) 2020-08-11

Similar Documents

Publication Publication Date Title
JP5244179B2 (en) ((1S) -1-(((2S) -2- (5- (4 ′-(2-((2S) -1-((2S) -2-((methoxycarbonyl) amino) -3-methyl Butanoyl) -2-pyrrolidinyl) -1H-imidazol-5-yl) -4-biphenylyl) -1H-imidazol-2-yl) -1-pyrrolidinyl) carbonyl) -2-methylpropyl) carbamate methyl dihydrochloride Crystal form
CN113784963B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN115175908A (en) Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
RU2633694C2 (en) Dyetherned phenylaminopyrimidine and pharmaceutical composition containing such connection
CN112694475A (en) Cycloalkyl and heterocycloalkyl inhibitors, and preparation method and application thereof
WO2007013673A1 (en) Fused heterocycles as lck inhibitors
TWI321566B (en) Pyrido[2,3-d]pyrimidine derivatives, preparation thereof, therapeutic use thereof
CN115109078A (en) Pyrimidopyridine inhibitor and preparation method and application thereof
JP7249508B2 (en) Carbazole compound and its use
CN111592528A (en) Deuterated pyridazinone, derivatives thereof and pharmaceutical compositions
TWI786303B (en) Crystal forms and applications of active compounds inhibiting CDK4/6
CA3203698A1 (en) 2-pyridone derivative, and preparation method therefor and pharmaceutical application thereof
CN112707892A (en) Pyridazinone or pyridazine compound and derivative and pharmaceutical composition thereof
WO2007039580A1 (en) Imidazolyl-substituted benzophenone compounds
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
WO2020221275A1 (en) Solid form of diaminopyrimidine compound or hydrate thereof, preparation method therefor, and application thereof
JP7464613B2 (en) Diarylthiohydantoin compound crystals
JP2023512471A (en) Crystals of PDE3/PDE4 dual inhibitors and uses thereof
AU2019339994B2 (en) Furo[3,4-b]pyrrole-containing BTK inhibitor
CN109232538B (en) 1,2, 4-triazole compound
JP5781537B2 (en) Aminoalkylpyrimidine derivatives as histamine H4 receptor antagonists
WO2017143011A1 (en) Histone demethylase inhibitors
WO2022268218A1 (en) Preparation method for heterocycloalkyl compound, and intermediate and application thereof heterocycloalkyl compound
CN117043163A (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical application thereof
JP2023525933A (en) Compounds as RET kinase inhibitors and their applications

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant