CN109265443A - Heterocyclic compound and its application as ASK inhibitor - Google Patents

Heterocyclic compound and its application as ASK inhibitor Download PDF

Info

Publication number
CN109265443A
CN109265443A CN201810780971.0A CN201810780971A CN109265443A CN 109265443 A CN109265443 A CN 109265443A CN 201810780971 A CN201810780971 A CN 201810780971A CN 109265443 A CN109265443 A CN 109265443A
Authority
CN
China
Prior art keywords
alkyl
alkoxy
hydroxy
radical
amino
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.)
Granted
Application number
CN201810780971.0A
Other languages
Chinese (zh)
Other versions
CN109265443B (en
Inventor
王勇
赵立文
葛崇勋
黄奕强
曹陈
李晴晴
江宏
韩伟
张着伟
张亚楠
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.)
Nanjing Sanhome Pharmaceutical Co Ltd
Original Assignee
Nanjing Sanhome Pharmaceutical 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 Nanjing Sanhome Pharmaceutical Co Ltd filed Critical Nanjing Sanhome Pharmaceutical Co Ltd
Publication of CN109265443A publication Critical patent/CN109265443A/en
Application granted granted Critical
Publication of CN109265443B publication Critical patent/CN109265443B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • 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
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • 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
    • 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
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • 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
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Cardiology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Immunology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Pulmonology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

The invention belongs to medicinal chemistry arts, it is related to a kind of heterocyclic compound as ASK inhibitor and its application, specifically, the present invention provides Formulas I compound represented or its isomers, pharmaceutically acceptable salt, solvate, crystallization or prodrug, their preparation method and pharmaceutical composition containing these compounds and these compound or compositions are for treating and/or preventing the purposes of the relevant disease of Apoptosis signal-regulating kinase 1.The compound of the present invention has excellent Apoptosis signal-regulating kinase 1 inhibitory activity, promises to be very much the therapeutic agent of Apoptosis signal-regulating kinase 1 related disease.

Description

Heterocyclic compounds as ASK inhibitors and uses thereof
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to heterocyclic compounds serving as ASK inhibitors or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, preparation methods of the heterocyclic compounds, pharmaceutical compositions containing the heterocyclic compounds, and application of the heterocyclic compounds or the pharmaceutical compositions in treating and/or preventing diseases related to apoptosis signal-regulated kinase 1.
Background
Organisms in nature are often exposed to various physical conditionsUnder the chemical pressure, the response of cells to external stimuli has a positive effect on maintaining the physiological homeostasis of cells, and is the most fundamental means for realizing self-renewal and self-protection of organisms after long-term development and evolution. The signaling pathway of the mitogen-activated protein kinase (MAPK) cascade is considered to be the most important pathway for regulating cellular oxidative stress, and its biological effects include regulation of cell growth, differentiation, inflammation and apoptosis. Apoptosis signal-regulating kinase 1(ASK1) is one of the major members of the mitogen-activated protein kinase (MAP 3Ks) family. ASK1 balances and integrates many endogenous and exogenous stimuli, allowing cells to respond appropriately to different stimuli. Under non-stressed conditions, ASK1 renders ASK1 inactive by binding thioredoxin (Trx). Stress injury (such as oxidative stress, ROS, endoplasmic reticulum stress, calcium ion influx, etc.) and proinflammatory factors (H) occur when organism appears2O2TNF- α, etc.) which in turn activates ASK1, activated ASK1 in turn activates the downstream JNK and p38-MAPK kinase signaling pathways, responding to stress by regulating cell growth, differentiation, inflammation and apoptosis ASK2 is a member of the MAP3K family with 45% homology to ASK1, ASK2 is highly similar to ASK1 in the kinase domain, ASK1 is widely distributed in a variety of tissues, and ASK2 is only specifically distributed in tissues exposed to the body surface, such as skin, lung and gastrointestinal tract, and ASK1 and ASK2 are generally believed to interact to form an endogenous ASK1-ASK2 complex to exert physiological effects.
Oxidative stress refers to a process in which excessive production of Reactive Oxygen Species (ROS) and/or a weakened antioxidant defense function of the body causes damage and death of cells, resulting in repair of tissue damage. During normal physiological processes, the production of reactive oxygen species, ROS, is at a low level and can be rapidly degraded. During the process of disease development, such as inflammation, ischemia reperfusion and neurodegenerative diseases, due to the continuous stimulation of pathogenic factors, the generation of ROS is increased, the body is in an oxidative stress state, and the ROS promotes ASK1 to be activated and abnormally activated for a long time after Trx and ASK1 are separated, so that the apoptosis and injury response are regulated. Due to the important role of ASK1 in the process of apoptosis signal transduction, ASK1 is closely related to the occurrence and development of various diseases such as inflammation, metabolic syndrome, neurodegenerative diseases, cardiovascular diseases, tumors and the like.
Oxidative stress plays an important role in the development of hepatic fibrosis (hepatic fibrosis, HF) is an essential process and a common pathway for various liver diseases to progress to cirrhosis, liver damage occurs when chronic diseases such as viral hepatitis, alcoholic liver disease and steatohepatitis occur, liver injury and inflammation induce massive production, cytokine increase such as TNF- α and TGF- β 1 are caused, and ROS and TNF- α and the like can activate JNK 1, activate downstream hepatic pathway k and p38, induce MAPK signaling and MAPK and promote apoptosis, and on the other hand, liver necrosis and ECM apoptosis occur in liver tissues.
In recent years, a plurality of researches find that the ASK1 inhibitor has certain research and application in the aspects of chronic kidney diseases (diabetic nephropathy, end-stage nephropathy, renal fibrosis and the like), cardiovascular diseases (heart failure and the like), neurodegenerative diseases (such as Alzheimer's disease and Parkinson's disease) and tumors, and particularly, the research of medicines taking ASK1 as targets in the aspects of chronic liver diseases such as non-alcoholic steatohepatitis (NASH), hepatic fibrosis and the like has important clinical significance and obtains certain clinical curative effect. However, there is still a need to develop more ASK1 inhibitors, so that the drugs have more excellent properties, such as better therapeutic effects and less side effects, and thus are better used for preventing or treating ASK 1-related diseases.
Disclosure of Invention
The invention aims to provide a compound with apoptosis signal regulating activity shown in a general formula A, or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof,
wherein,
ring a is selected from cycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, aryl, and heteroaryl are optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring B is selected from monocyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl, wherein said monocyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl are optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring C is a monocyclic heteroaryl, wherein said heteroaryl is optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or heteroaryl;
ring D is selected fromAnd tetrazole, wherein the groups are optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or heteroaryl;
Z1is acyl, alkylene acyl, or absent;
Z2is-CONH-, -NHCO-or is absent;
R1selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; and
provided that Z is1When absent, ring a is heteroaryl and is optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, or oxo groups;
and when the ring B is monocyclic aryl, the ring D is tetrazole.
It is another object of the present invention to provide a method for preparing the compound of the general formula a of the present invention or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug.
It is a further object of the present invention to provide a composition comprising a compound of formula a of the present invention or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug and a pharmaceutically acceptable carrier, and a composition comprising a compound of formula a of the present invention or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug and another drug or drugs.
Still another object of the present invention is to provide a method for treating and/or preventing apoptosis signal-regulating kinase 1-associated diseases by using the compound of the general formula a or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug of the present invention, and the use of the compound of the general formula a or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug of the present invention in the preparation of drugs for treating and/or preventing apoptosis signal-regulating kinase 1-associated diseases.
Aiming at the above purpose, the invention provides the following technical scheme:
in a first aspect, the present invention provides a compound represented by the general formula A or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof,
wherein,
ring a is selected from cycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, aryl, and heteroaryl are optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring B is selected from monocyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl, wherein said monocyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl are optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring C is a monocyclic heteroaryl, wherein said heteroaryl is optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or heteroaryl;
ring D is selected fromAnd tetrazole, wherein the groups are optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or heteroaryl;
Z1is acyl, alkylene acyl, or absent;
Z2is-CONH-, -NHCO-or is absent;
R1selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoalkyl, dialkylamino, carboxyalkyl, amino, carboxyalkyl, carboxya,Alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; and
provided that Z is1When absent, ring a is heteroaryl and is optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, or oxo groups;
and when the ring B is monocyclic aryl, the ring D is tetrazole.
In some preferred embodiments, the present invention provides a compound represented by formula a, or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, wherein formula a has the structure of the following formula I,
wherein,
ring a is selected from cycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, aryl, and heteroaryl are optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring B is selected from the group consisting of bicyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl, wherein said bicyclic aryl, bicyclic heteroaryl, and bicyclic heterocyclyl are optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamido, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, or oxo groups;
ring C is a monocyclic heteroaryl, wherein said heteroaryl is optionally substituted with one or more halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or heteroaryl;
Z1is acyl, alkylene acyl, or absent;
Z2is-CONH-, -NHCO-or is absent;
R1selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; and
provided that Z is1When absent, ring a is heteroaryl and is optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, or oxo groups.
In some preferred embodiments, the compound of the present invention is a compound of formula a or formula I or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
ring A is selected from C3-12Cycloalkyl radical, C6-18Aryl and 5-20 membered heteroaryl, wherein said cycloalkyl, aryl and heteroaryl are optionally substituted with one or more halogen, hydroxyBase, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
further preferably, ring A is selected from C3-8Cycloalkyl radical, C6-12Aryl and 5-12 membered heteroaryl, wherein said cycloalkyl, aryl and heteroaryl are optionally substituted with one or more halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
still further preferably, ring a is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, naphthyl, furyl, pyrrolyl, thienyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, pyridyl, pyrimidinyl, pyrazinyl and pyridazinyl, wherein said groups are optionally substituted with one or more halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkyl acyl radicalAmino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-10 membered aryl, 5-10 membered heteroaryl, or oxo.
In some preferred embodiments, the compound of the present invention is a compound of formula a or formula I or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
ring B is selected from the group consisting of 10-18 membered bicyclic aryl, 9-or 10-membered bicyclic heteroaryl, 9-or 10-membered bicyclic heterocyclyl, wherein said bicyclic aryl, bicyclic heteroaryl and bicyclic heterocyclyl are optionally substituted with one or more halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
further preferably, ring B is selected from naphthyl, 9-or 10-membered benzazepine, 9-or 10-membered benzooxacyclo, 9-or 10-membered benzothiepinyl, 9-or 10-membered benzodiazepine, 9-or 10-membered benzoxazepine, 9-or 10-membered benzothiepin, wherein said groups are optionally substituted with one or more halogen, hydroxyl, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkyl acylAmino radical, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
still more preferably, ring B is selected from naphthyl, indolyl, isoindolyl, indolinyl, isoindolinyl, benzofuranyl, dihydrobenzofuranyl, benzopyranyl, dihydrobenzopyranyl, benzothienyl, dihydrobenzothienyl, benzoxazolonyl, benzopyrazolyl, dihydrobenzopyrazolyl, benzimidazolyl, dihydrobenzimidazolyl, benzopyrazolyl, dihydrobenzopyrazolyl, benzoxazolyl, dihydrobenzoxazolyl, benzothiazolyl, dihydrobenzothiazolyl, benzisoxazolyl, dihydrobenzisoxazolyl, benzisothiazolyl, dihydrobenzisothiazolyl, quinolinyl, tetrahydroquinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, dihydroquinolinyl, dihydroisoquinolinyl, quinazolinyl, tetrahydroquinazolinyl, dihydroquinazolinyl, cinnolinyl, dihydrocinnolinyl, tetrahydrocinnolinyl, isoindolinyl, benzisothiazolyl, and benz, Quinoxalinyl, dihydroquinoxalinyl, tetrahydroquinoxalinyl, benzoxazinyl, dihydrobenzoxazinyl, benzothiazinyl, dihydrobenzothiazinyl, benzodioxanyl, benzomorpholinyl, benzoxazothiohexyl and benzodioxolyl, wherein said radicals are optionally substituted by one or more halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-10 membered aryl, 5-10 membered heteroaryl, or oxo.
In some preferred embodiments, the compound of the present invention is a compound of formula a or formula I or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
ring C is a 5-12 membered monocyclic heteroaryl, wherein said heteroaryl is optionally substituted with one or more halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl or 5-12 membered heteroaryl;
further preferably, ring C is a 5-6 membered monocyclic heteroaryl, wherein said heteroaryl is optionally substituted with one or more halogens, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl, 3-6 membered heterocyclyl or 5-10 membered heteroaryl;
even more preferably, ring C is selected from furyl, pyrrolyl, thienyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl and 1,3, 5-triazinyl, wherein said groups are optionally substituted with one or more halogens, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy radicalNitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl, 3-6 membered heterocyclyl or 5-10 membered heteroaryl.
In some preferred embodiments, the compound of the present invention is a compound of formula a or formula I or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
Z1is acyl, imino C1-6Alkyl acyl or absent;
further preferably, Z1Is acyl, imino C1-6Alkyl acyl or absent;
even more preferably, Z1Is composed ofOr is absent.
In some preferred embodiments, the compound of the present invention is a compound of formula a or formula I or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
R1selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl and 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl;
further preferably, R1Is selected fromHydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl;
even more preferably, R1Selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, halogenated C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl.
In some preferred embodiments, the compounds of the present invention are compounds of general formula a or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
ring D is selected from tetrazole, optionally substituted with one or more R2Substitution;
further preferably, ring D is selected from the groupSaid group being optionally substituted by one or more R2Substitution;
even more preferably, ring D is selected from the group
Wherein R is2One or more groups selected from the group consisting of halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, hydroxyhaloalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo.
In some preferred embodiments, the present invention provides a compound of formula Ia or an isomer, a pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein,
y is selected from alkylene, alkenylene and cycloalkylene, wherein the alkylene, alkenylene and cycloalkylene are optionally substituted with one or more alkyl, haloalkyl, hydroxy, hydroxyalkyl, halogen, oxo, alkoxy, carboxy, cyano, amino, monoalkylamino or dialkylamino groups, or when Y is alkylene and is substituted with two alkyl groups, the two alkyl groups may together with the C atom to which they are attached form a cycloalkyl group;
w is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
m is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; and
R1as defined above.
In some preferred embodiments, the compounds of the present invention are compounds of formula Ia or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
y is selected from sub-C1-6Alkyl radical, C2-10Alkenylene and C3-10Cycloalkyl, wherein said alkylene is1-6Alkyl radical, C2-10Alkenylene and C3-10Cycloalkylene radicals may be substituted by one or more C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, halogen, oxo, C1-6Alkoxy, carboxyl, cyano, amino, mono C1-6Alkylamino or di-C1-6Substituted by alkylamino, or when Y is C-ylene1-6When an alkyl group is substituted with two alkyl groups, the two alkyl groups may form C together with the C atom to which they are attached3-8A cycloalkyl group;
further preferably, Y is selected from the group consisting of sub-C1-3Alkyl radical, C2-6Alkenylene and C3-6Cycloalkylene, wherein said alkylene is C1-3Alkyl radical, C2-6Alkenylene and C3-6Cycloalkylene radicals may be substituted by one or more C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl, halogen, oxo, C1-3Alkoxy, carboxyl, cyano, amino, mono C1-3Alkylamino or di-C1-3Substituted by alkylamino, or when Y is C-ylene1-3When an alkyl group is substituted with two alkyl groups, the two alkyl groups may form C together with the C atom to which they are attached3-6A cycloalkyl group;
even more preferably, Y is selected from the group consisting of methylene, ethylene, propylene, ethenylene, propenylene and cyclopropylene, wherein said methylene, ethylene, propyleneOptionally substituted by one or more halogen, methyl, ethyl, propyl, isopropyl, oxo groups, or when Y is methylene and is substituted by two alkyl groups, the two alkyl groups may form together with the C atom to which they are attached a C3-C6A cycloalkyl group.
In some preferred embodiments, the compounds of the present invention are compounds of formula Ia or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
m is selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl;
further preferably, M is selected from hydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl.
In some particular embodiments, the compounds of formula Ia according to the present invention or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein the structureIs selected from
In some particular embodiments, the compounds of formula Ia according to the present invention or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof, wherein M is hydrogen,
In some preferred embodiments, the compounds of the present invention are compounds of formula Ia or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
w is selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl;
further preferably, W is selected from hydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl;
even more preferably, W is selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-8 membered aryl or 5-8 membered heteroaryl.
In some particular embodiments, a compound according to formula Ia of the present invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
structure of the productIs selected from
M is hydrogen,
W is selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl radicalAcyl, aminoacyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl; and
R1selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl and 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl.
In some preferred embodiments, the present invention provides a compound of formula Ib, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein, W, R1、Z1、Z2Ring B and ring C are as defined above.
In some preferred embodiments, the present invention provides a compound of formula Ic or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein, W, R1Y and Ring C are as defined above, X1、X2Are each independently selected from C (R)2) And N, wherein R2Selected from hydrogen, halogen, hydroxy, alkaneA group selected from the group consisting of alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxyl, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamido, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
In some preferred embodiments, the compounds of the present invention are of formula Ic or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
R2selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl;
further preferably, R2Selected from hydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl;
even more preferably, R2Selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, halogenated C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-8 membered aryl, and 5-8 membered heteroaryl.
In some embodiments, the present invention provides a compound of the general formula Id or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
wherein,
ring B is selected from monocyclic aryl andoptionally substituted with one or more groups selected from halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamido, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo;
R2one or more groups selected from the group consisting of halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, hydroxyhaloalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo; and
W、Z2as defined above for formula A, I, Ia, Ib or Ic.
In some embodiments, a compound of formula Id or an isomer, a pharmaceutically acceptable salt, solvate, crystal or prodrug thereof according to the present invention, wherein ring B is selected from phenyl andoptionally substituted by one or more groups selected from halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl radical, C3-6Heterocyclyl, aryl, heteroaryl and oxo.
In some particular embodiments, the compound of formula Ib according to the invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein ring B is selected from phenyl andoptionally substituted by one or more groups selected from halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-6Cycloalkyl radical, C3-6Heterocyclyl, aryl, heteroaryl and oxo.
The present invention provides the following specific compounds or isomers, pharmaceutically acceptable salts, solvates, crystals or prodrugs thereof:
in another aspect, the invention provides a process for the preparation of a compound of the general formula (la) of the invention, for example:
1) the process of the present invention for the preparation of compounds of the general formula Ia can be prepared by the following steps:
a) reacting the compound shown in the formula 1 with hydrazine hydrate to obtain a compound shown in a formula 2;
b) reacting the compound of formula 2 with N, N-dimethylformamide dimethyl acetal to obtain a compound of formula 3;
c) reacting the compound of formula 3 with the compound of formula 4 to obtain a compound of formula 5;
d) reacting the compound of formula 5 with the compound of formula 6 to obtain a compound of formula 7;
e) reacting the compound of formula 7 with the compound of formula 8 produces the compound of formula Ia.
Wherein X is halogen, preferably chlorine or bromine, R1W, Y, M are as defined above for formula Ia.
2) When Z is2The process for the preparation of the compounds of formula Id according to the invention, chosen from-CONH-, can be carried out by reacting a compound of formula Id-1 with a compound of formula Id-2The preparation method comprises the following steps:
wherein X is halogen, preferably chlorine or bromine, R2、W、Z2Ring B is as defined above for formula Id.
3) The compounds of formula A, I, Ib or Ic according to the invention can be prepared by reference to the preparation of formula Ia or by other techniques customary to those skilled in the art.
In a third aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention or an isomer, a pharmaceutically acceptable salt, solvate, crystal or prodrug thereof.
In some embodiments, the present invention provides pharmaceutical compositions comprising a compound of the present invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, further comprising one or more agents selected from the group consisting of: one or more Angiotensin Converting Enzyme (ACE) inhibitors such as enalapril, captopril, ramipril, lisinopril, and quinapril; or angiotensin II receptor blockers (ARBs), such as losartan, olmesartan and irbesartan; or antihypertensive agents such as amlodipine, nifedipine, felodipine, and the like; antibiotics, analgesics, antidepressants and/or anxiolytics, and the like.
In some embodiments, the present invention provides a compound of the present invention or an isomer, a pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutical composition comprising a compound of the present invention or an isomer, a pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, for use in the treatment and/or prevention of an apoptosis signal-regulating kinase 1-associated disease.
In some embodiments, the present invention provides a pharmaceutical composition comprising a compound of the present invention, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
The compound of the present invention or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug can be mixed with a pharmaceutically acceptable carrier, diluent or excipient to prepare a pharmaceutical preparation suitable for oral or parenteral administration. Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, and oral routes. The formulations may be administered by any route, for example by infusion or bolus injection, by a route of absorption through epithelial or cutaneous mucosa (e.g. oral mucosa or rectum, etc.). Administration may be systemic or local. Examples of the formulation for oral administration include solid or liquid dosage forms, specifically, tablets, pills, granules, powders, capsules, syrups, emulsions, suspensions and the like. The formulations may be prepared by methods known in the art and include carriers, diluents or excipients conventionally used in the art of pharmaceutical formulation.
In a fourth aspect, the present invention provides a use of a compound represented by general formula a, I, Ia, Ib, Ic or Id of the present invention, or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, or a pharmaceutical composition comprising the same, for the preparation of a medicament for treating and/or preventing an apoptosis signal-regulated kinase 1-associated disease, wherein the apoptosis signal-regulated kinase 1-associated disease or condition includes, but is not limited to: neurodegenerative diseases, cardiovascular diseases, inflammation, autoimmune diseases, tumors, metabolic disorders, and the like, and particularly, diseases for which ASK1 inhibitors are used for treatment include: kidney diseases (e.g., diabetic nephropathy, end-stage renal disease, chronic kidney disease, etc.), fibrotic diseases (e.g., lung, renal fibrosis, etc.), cardiovascular diseases (e.g., heart failure, etc.), respiratory diseases (e.g., Chronic Obstructive Pulmonary Disease (COPD), pulmonary hypertension, acute lung injury, etc.), acute and chronic liver diseases (e.g., nonalcoholic steatohepatitis (NASH), Alcoholic Hepatitis (AH), liver fibrosis, etc.), neurodegenerative diseases (e.g., alzheimer's disease, parkinson's disease, etc.), and tumors.
Description of the terms
Unless stated to the contrary, terms used in the specification and claims have the following meanings.
The "hydrogen", "carbon" and "oxygen" in the compounds of the present invention include all isotopes thereof. Isotopes are understood to include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include protium, tritium, and deuterium, and isotopes of carbon include12C、13C and14c, isotopes of oxygen including16O and18o, and the like.
The "halogen" in the present invention means fluorine, chlorine, bromine and iodine. "halo" in the context of the present invention means substituted by fluorine, chlorine, bromine or iodine.
"alkyl" in the present invention means a straight-chain or branched saturated aliphatic hydrocarbon group, preferably a straight-chain or branched group having 1 to 6 carbon atoms, further preferably a straight-chain or branched group having 1 to 3 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl and the like. The alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be at any available point of attachment.
"alkylene" in the context of the present invention refers to a radical of an alkyl group which is formally left after removal of one hydrogen atom, such as methylene (-CH)2-, ethylene (-CH)2-CH2-, propylene (-CH)2-CH2-CH2-) etc., as used herein, said "C-ene1-6Alkyl "means C1-6Alkyl radicals formally leaving one hydrogen atom removed, said "C-ene1-3Alkyl "means C1-3Alkyl formally removes a hydrogen atom from the remaining radical. The alkylene group may be substituted or unsubstituted, and when substituted, the substituent may be at any available point of attachment.
The "alkenylene group" in the present invention means a group obtained by formally removing one hydrogen atom from an alkenyl group, such as an ethenylene group (-CH-), an propenylene group (-CH-), and the like2-or-CH2-CH ═ CH —), etc., and in the present context, said "sub-C —"2-10Alkenyl "means C2-10Alkenyl radicals formally removed from one hydrogen atom to leave a radical, said "C-ene2-6Alkenyl "means C2-6Alkenyl groups formally remove one hydrogen atom from the remaining group.
"Cycloalkylene" in the context of the present invention refers to a cycloalkyl group which is formally interrupted by a hydrogen atom, e.g. cyclopropyleneCyclo-butylene groupEtc., as used herein, the term "sub-C" refers to3-10Cycloalkyl "means C3-10Cycloalkyl radicals formally leaving one hydrogen atom removed, said "C being3-6Cycloalkyl "means C3-6Cycloalkyl formally removes a hydrogen atom from the remaining radical.
The "alkylidenyl group" in the present invention means a group in which an alkylacyl group is formally left by removing one hydrogen atom, such as a methyleneacyl group (-CH)2CO-), ethyleneamino (-CH)2CH2CO-), propyleneamino (-CH)2CH2CH2CO-) and the like.
The "acyl" in the present invention means-CO-.
"haloalkyl" in the context of the present invention means an alkyl group substituted with at least one halogen.
"hydroxyalkyl" in the context of the present invention means an alkyl group substituted with at least one hydroxyl group.
"alkoxy" in the context of the present invention means-O-alkyl. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, n-propoxy, isopropoxy, isobutoxy, sec-butoxy and the like. An alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent may be at any available point of attachment.
The "cycloalkyl group" in the present invention means a cyclic saturated hydrocarbon group. Suitable cycloalkyl groups may be substituted or unsubstituted monocyclic, bicyclic or tricyclic saturated hydrocarbon groups having 3 to 12 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
"heterocyclyl" in the present invention refers to a group of a 3-to 12-membered non-aromatic ring system having 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus and silicon ("3-12 membered heterocyclyl"). In heterocyclyl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. The heterocyclyl group can either be monocyclic ("monocyclic heterocyclyl") or a fused, bridged or spiro ring system (e.g., a bicyclic system ("bicyclic heterocyclyl")) and can be saturated or can be partially unsaturated. The heterocyclic bicyclic ring system may include one or more heteroatoms in one or both rings. "heterocyclyl" also includes ring systems in which a heterocycle, as defined above, is fused to one or more carbocyclyl groups (in which the point of attachment is on the carbocyclyl or on the heterocycle), or in which a heterocycle, as defined above, is fused to one or more aryl or heteroaryl groups (in which the point of attachment is on the heterocycle), and in such cases the number of ring members continues to be referred to as the number of ring members in the heterocyclic system. Unless otherwise specified, each instance of a heterocyclyl is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heterocyclyl") or substituted (a "substituted heterocyclyl") with one or more substituents, such as substituted or unsubstituted piperidinyl, substituted or unsubstituted bridged morpholinyl, and the like. In certain embodiments, the heterocyclyl group is a substituted 3-10 membered heterocyclyl. Fusion to C6Exemplary 5-membered heterocyclyl groups for aryl rings (also referred to herein as 5, 6-bicyclic heterocycles) include, but are not limited to, indolinyl, isoindolinyl, indolyl, and a pharmaceutically acceptable salt thereof,Dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolonyl, and the like. Exemplary 6-membered heterocyclyl groups fused to aryl rings (also referred to herein as 6, 6-bicyclic heterocycles) include, but are not limited to, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
"aryl" as used herein refers to an aromatic system which may comprise a single ring or fused polycyclic ring, preferably a single ring or fused bicyclic ring, having from 6 to 18 carbon atoms, preferably from about 6 to about 12 carbon atoms. Suitable aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, tetrahydronaphthyl, fluorenyl, indanyl. Aryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents may be at any available point of attachment.
The "heteroaryl group" of the present invention means an aryl group having at least one carbon atom replaced with a heteroatom, which is composed of 5 to 20 atoms (5-20 membered heteroaryl group), and more preferably consists of 5 to 12 atoms (5-12 membered heteroaryl group), and the heteroatom is O, S, N, including but not limited to, imidazolyl, benzimidazolyl, imidazopyridinyl, quinazolinone, pyrrolyl, imidazolonyl, furyl, thienyl, pyrazolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, pyrimidinyl, pyridyl, pyrazinyl, pyridazinyl, pyrimidopyrazolyl, pyrimidoimidazolyl, and the like. Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents may be at any available point of attachment.
"isomers" of the present invention are compounds having the same molecular formula but differing in nature or in the bond sequence of their atoms or in the spatial arrangement of their atoms. Stereoisomers are isomers whose atoms differ in their spatial arrangement. Stereoisomers that are not mirror images of each other are diastereomers and stereoisomers that are non-overlapping mirror images of each other are enantiomers. When the compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. Enantiomers are characterized by the absolute configuration of their asymmetric centers and are described and designated as dextrorotatory or levorotatory (i.e., as (+) or (-) -isomers, respectively) by the R-and S-sequencing rules of Cahn and Prelog, or by methods in which molecules rotate the plane of polarized light. The chiral compound may exist as a single enantiomer or a mixture thereof. Mixtures containing equal proportions of enantiomers are referred to as "racemic mixtures".
The term "pharmaceutically acceptable salt" as used herein refers to salts of the compounds of the present invention which are safe and effective for use in the body of a mammal and which possess the requisite biological activity.
"solvate" of the present invention refers in the conventional sense to a complex of a solute (e.g., active compound, salt of active compound) and a solvent (e.g., water) in combination. Solvent means a solvent known or readily determined by one skilled in the art. If water, the solvate is often referred to as a hydrate, e.g., a hemihydrate, monohydrate, dihydrate, trihydrate or a substitute amount thereof, and the like.
The in vivo effect of the compound of formula (a) may be exerted in part by one or more metabolites formed in the human or animal body after administration of the compound of formula (a). As mentioned above, the in vivo effect of the compound of formula (a) may also be exerted via metabolism of the precursor compound ("prodrug"). The "prodrug" of the present invention refers to a compound which is converted into a compound of the formula (a) by a reaction with an enzyme, gastric acid or the like under physiological conditions in a living body, that is, a compound which is converted into a compound of the formula (a) by oxidation, reduction, hydrolysis or the like by an enzyme, and/or a compound which is converted into a compound of the formula (a) by a hydrolysis reaction or the like by gastric acid or the like, and the like.
The "crystal" in the present invention is a solid whose internal structure is formed by repeating constituent atoms (or groups thereof) regularly in three dimensions, and is different from an amorphous solid having no such regular internal structure.
The "pharmaceutical composition" of the present invention is meant to comprise a mixture of any of the compounds described herein, including the corresponding isomers, prodrugs, solvates, pharmaceutically acceptable salts, or chemically protected forms thereof, and one or more pharmaceutically acceptable carriers and/or another drug or drugs. The purpose of the pharmaceutical composition is to facilitate the administration of the compound to an organism. The compositions are generally useful for the preparation of medicaments for the treatment and/or prevention of diseases mediated by one or more kinases.
The "pharmaceutically acceptable carrier" of the present invention means a carrier that does not cause significant irritation to an organism and does not interfere with the biological activity and properties of the administered compound, and includes all solvents, diluents or other excipients, dispersants, surfactant isotonicity agents, thickeners or emulsifiers, preservatives, solid binders, lubricants and the like. Unless any conventional carrier medium is incompatible with the compounds of the present invention. Some examples of carriers that may be pharmaceutically acceptable include, but are not limited to, sugars such as lactose, glucose, and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, and cellulose acetate; malt, gelatin, and the like.
"excipient" in the context of the present invention refers to an inert substance added to a pharmaceutical composition to further facilitate administration of the compound. Excipients may include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols.
Detailed Description
The following representative examples are intended to better illustrate the present invention and are not intended to limit the scope of the present invention. The materials used in the following examples are all commercially available unless otherwise specified.
Example 1: preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] imidazole-6-carboxamide
Step 1 preparation of 6-aminopyridine-2-carbohydrazide
In a 250mL single-neck flask, methyl 6-aminopyridinecarboxylate (15.0g, 98.7mmol) was added, dissolved in methanol (150mL), and hydrazine hydrate (12.0g, 198mmol) was added and refluxed at 70 ℃ for 5 h. After the reaction was complete, filtration was carried out, and the filter cake was washed with ethyl acetate and dried in vacuo to give the title compound. LC-MS M/z [ M + H ]]+=153.
Step 2: (E) preparation of (E) -N' - (6- (2- ((E) - (dimethylamino) methylene) hydrazine-1-carbonyl) pyridin-2-yl) -N, N-dimethylformamidine
6-aminopyridine-2-carbohydrazide (12.0g, 78.9mmol) obtained in step 1 was dissolved in 120mL of N-dimethylformamide dimethyl acetal in a 250mL single-neck flask and reacted for 4h under reflux. After completion of the reaction, the reaction solution was washed with water (100 mL. times.3), extracted with dichloromethane (100 mL. times.3), the organic phases were combined, dried and concentrated, and the residue was slurried with ethyl acetate to give the title compound. LC-MS M/z [ M + H ]]+=263.
And step 3: preparation of 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine
(E) -N' - (6- (2- ((E) - (dimethylamino) methylene) hydrazine-1-carbonyl) pyridin-2-yl) -N, N-dimethylformamidine (16.0g, 60.8mmol) from step 2 was placed in a 500mL single vial and acetonitrile (150mL), acetic acid (50mL) and isopropylamine (17.9g, 304mmol), 8Reflux at 0 ℃ overnight. After the reaction, the reaction mixture was concentrated under reduced pressure, dissolved in water (30mL), adjusted to neutral pH, extracted with ethyl acetate (300 mL. times.5), the organic phases combined and concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=204.
Step 4 preparation of 1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] imidazole-6-carboxylic acid methyl ester
In a 100mL single-necked flask, 2-bromo-1-phenyleth-1-one (1.00g, 5.00mmol) and methyl 1H-benzo [ d ] are added]Ethyl imidazole-6-carboxylate (798mg, 4.50mmol) was dissolved in N, N-dimethylformamide (25.0mL), followed by addition of potassium carbonate (1.00g, 7.50mmol) and reaction at room temperature for 4 hours, whereupon the reaction was terminated. The reaction was diluted with ethyl acetate (150mL), washed 3 times with water (450mL) to remove N, N-dimethylformamide, the combined organic layers were dried over anhydrous sodium sulfate, and the solvent was removed by evaporation to give the title compound. LC-MS M/z [ M + H ]]+=295.1。
And 5: preparation of 1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] imidazole-6-carboxylic acid
In a 100mL single-necked flask, 1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] is added]Imidazole-6-methyl formate (800mg, 2.72mmol) is dissolved by adding 10.0mL of ethanol, then 4M sodium hydroxide aqueous solution is added for 8mL, and the reaction is finished after 3 hours at room temperature; the reaction solution was adjusted to pH 1 with 1M hydrochloric acid and extracted with dichloromethane/isopropanol 3: 1. The organic phase was dried over anhydrous sodium sulfate and the solvent was removed by rotary evaporation to give the title compound. LC-MS M/z [ M + H ]]+=281.1.
Step 6: preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] imidazole-6-carboxamide
In a 100mL single-necked flask, 1- (2-oxo-2-phenylethyl) -1H-benzo [ d ] is added]Imidazole-6-carboxylic acid (910mg, 3.25mmol) and 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (600mg, 2.96mmol) were dissolved with o-xylene (15.0mL), refluxed at 150 ℃ for 5min, followed by addition of 5 drops of N, N-dimethylformamide and PCl5The reaction is carried out for 40min under the condition, and the reaction is finished; the reaction was quenched with water, adjusted to pH 12 with 2M aqueous sodium hydroxide and extracted with dichloromethane/methanol 3: 1. The organic phase was dried over anhydrous sodium sulfate and the solvent was removed by rotary evaporation to give the title compound.1H NMR(400MHz,DMSO-d6)δ10.64(s,1H),8.87(s,1H),8.44(s,1H),8.36(s,1H),8.22(d,1H),8.14(d,2H),8.00-8.08(m,1H),7.80-7.92(m,2H),7.67-7.78(m,2H),7.62-7.67(m,2H),6.09(s,2H),5.75-5.82(m,1H),1.43(d,6H).LC-MS m/z:[M+H]+=466.2.
Example 2- (Cyclopropanecarbonyl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxylic acid amide
Step 1 preparation of 3, 4-dibromomethyl-methyl benzoate
In a 250mL single neck flask, methyl 3, 4-dimethylbenzoate (6.00g, 36.6mmol), benzoyl peroxide (900mg, 3.66mmol), and N-bromosuccinimide (13.0g, 73.2mmol) were added, dissolved in chloroform (60mL), and refluxed for 20 h. After the reaction, insoluble substances were removed by suction filtration, the filtrate was concentrated under reduced pressure,the concentrated residue was dissolved in dichloromethane, washed with saturated aqueous sodium bicarbonate solution, dried over anhydrous sodium sulfate, and the solvent was removed by vortexing to give the title compound. LC-MS M/z [ M + H ]]+=321.
Step 2: preparation of methyl 2-benzylisoindoline-5-carboxylate
3, 4-Dibromomethyl-benzoate (12.0g, 37.5mmol) was dissolved in a single-neck flask containing 120mL of tetrahydrofuran, and benzylamine (4.00g, 37.5mmol) and triethylamine (7.57g, 75.0mmol) were added, and the mixture was stirred at room temperature overnight. After the reaction, the reaction mixture was washed with water (200mL), extracted with ethyl acetate (100 mL. times.3), and the organic phases were combined, concentrated under reduced pressure, purified by column chromatography, and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=268.
And step 3: preparation of 2-benzyl isoindoline-5-carboxylic acid
In a 100mL single-neck flask, methyl 2-benzylisoindoline-5-carboxylate (4.50g, 16.9mmol) was dissolved in methanol (40mL), water (10mL) and lithium hydroxide (2.00g, 84.3mmol) were added, and the mixture was stirred at room temperature for 5 h. After the reaction was complete, the pH was adjusted to neutral, extracted with dichloromethane (100 mL. times.3), and the organic phases were combined and spin dried to give the title compound. LC-MS M/z [ M + H ]]+=254.
And 4, step 4: preparation of 2-benzyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Placing the 2-benzyl isoindoline-5-formic acid (2.00g, 7.90mmol) prepared in the step 3 into a 100mL single-neck bottle, adding anhydrous dichloromethane (30mL), adding oxalyl chloride (1.60g, 12.6mmol) under cooling, stirring at room temperature for 2 hours, and after the reaction is finished, concentrating the reaction solution under reduced pressure to obtain a dark green solid.
6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (1.30g, 7.10mmol) obtained in step 3 of example 1 was placed in a 100mL single-necked flask, dissolved in anhydrous dichloromethane (30mL), and the above solid and N, N-diisopropylethylamine (2.00g, 15.8mmol) were added under cooling and stirred at room temperature for 3 hours. After completion of the reaction, the reaction mixture was washed with water (50mL), dried and concentrated to obtain a crude product, which was then subjected to column chromatography (ethyl acetate: ethanol ═ 0 to 5%) to isolate the title compound. LC-MS M/z [ M + H ]]+=439.
And 5: preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Pd/C (60.0mg, 10%) was placed in a 100mL single-neck flask, methanol (30mL) was added, then 2-benzyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide (600mg, 1.38mmol) prepared in step 4 was added, 3 drops of concentrated hydrochloric acid were added dropwise, hydrogen replaced three times, and stirred at room temperature. After the reaction was completed, filtration was carried out, and the filtrate was concentrated under reduced pressure to obtain the title compound. LC-MS M/z [ M + H ]]+=349.
Step 6: preparation of 2- (cyclopropanecarbonyl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Dissolving N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide (200mg, 0.570mmol) prepared in step 5 in dichloromethane, adding N, N-diisopropylethylamine (190mg, 1.44mmol) under cooling, stirring for 3minAfter that, cyclopropylcarbonyl chloride (71.0mg, 0.680mmol) was added thereto and stirred at room temperature. After completion of the reaction, the reaction mixture was diluted with water (50mL), extracted with dichloromethane: ethanol ═ 5:1(30mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was then subjected to preparative high performance liquid chromatography (acidic, formic acid) to give the title compound.1H NMR(400MHz,DMSO-d6)δ10.8(s,1H),8.88(s,1H),8.17-8.20(m,1H),8.01-8.05(m,1H),7.93-7.97(m,2H),7.87(d,1H),7.54(d,1H),5.69-5.75(m,1H),5.11(s,2H),4.73(s,2H),1.88-1.93(m,1H),1.44(d,6H),0.82-0.86(m,4H).LC-MS m/z:[M+H]+=417.
EXAMPLE 3 6- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -2-naphthamide
Step 1: preparation of methyl 6- (4-cyclopropyl-1H-imidazol-1-yl) -2-naphthoate
Methyl 6-bromo-2-naphthoate (491mg,1.85mmol) and 4-cyclopropyl-1H-imidazole (200mg, 1.85mmol) were charged into a 100mL single-neck flask, followed by addition of 8-hydroxyquinoline (27.0mg, 0.185mmol), potassium carbonate (511mg, 3.70mmol) and cuprous iodide (35.2mg, 0.185mmol), followed by dissolution in dimethyl sulfoxide (5mL), nitrogen substitution, and heating to 120 ℃ for 6H. And detecting the complete reaction of the raw materials by LC-MS. To the reaction solution was added water (20mL), extracted with ethyl acetate (50mL × 3), and the organic phase was filtered to remove the residue, followed by washing with a saturated sodium chloride solution, dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give a crude product, which was subjected to column chromatography (petroleum ether: ethyl acetate ═ 10:1) to give the title compound as pale yellow. LC-MS M/z [ M + H ]]+=293。
Step 2: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2-naphthoic acid
Methyl 6- (4-cyclopropyl-1H-imidazol-1-yl) -2-naphthoate (800mg, 2.74mmol) was dissolved in a mixed solution of tetrahydrofuran (20mL) and water (3mL), followed by addition of lithium hydroxide (132mg, 5.48mmol) and reaction at 25 ℃ for 3H, TLC indicated completion of the reaction. The reaction solution was adjusted to pH 3 with dilute hydrochloric acid (1N), extracted with 10% methanol in dichloromethane (30 mL. times.3), the organic phase was washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give the title compound. LC-MSm/z: [ M + H]+=279。
And step 3: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -2-naphthamide
6- (4-cyclopropyl-1H-imidazol-1-yl) -2-naphthoic acid (600mg, 2.16mmol) and 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (438mg, 2.16mmol) were dissolved in o-xylene (30mL) and N, N-dimethylformamide (10mL), then phosphorus pentachloride (225mg, 1.08mmol) was added, heated to 150 ℃ and reacted for 3H. LC-MS showed the reaction was complete, after removal of o-xylene under reduced pressure, ethyl acetate (50mL) and water (30mL) were added for dilution, the liquid was separated, the aqueous phase was extracted with ethyl acetate (50 mL. times.2), the organic phases were combined, dried and concentrated to give the crude product, which was prepared by preparative high performance liquid chromatography to give the title compound.1H NMR(400MHz,DMSO-d6)δ10.98(s,1H),8.89(s,1H),8.66(s,1H),8.18-8.35(m,4H),8.00-8.13(m,3H),7.93(d,1H),7.89(d,1H),7.70(s,1H),5.70-5.79(m,1H),1.78-1.91(m,1H),1.46(d,6H),0.79-0.85(m,2H)0.68-0.75(m,2H)。LC-MS m/z:[M+H]+=464.2。
Example 4:6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazol-4-yl) -isoindolin-1-one
Step 1 preparation of 4-bromo-1, 3-thiazole-2-carboxylic acid methyl ester
In a 100mL single-neck flask, 4-bromo-1, 3-thiazole-2-carboxylic acid (2.00g, 9.80mmol) was added, dissolved in methanol (20mL), added thionyl chloride (1.16g, 9.80mmol) in an ice-water bath, and refluxed at 70 ℃ for 3 h. After the reaction, the reaction mixture was concentrated under reduced pressure and dried in vacuo to give the title compound. LC-MS M/z [ M + H ]]+=222.
Step 2 preparation of 4-bromothiazole-2-formhydrazide
In a 100mL single-neck flask, 4-bromo-1, 3-thiazole-2-carboxylic acid methyl ester (2.00g, 9.01mmol) was added, dissolved in ethanol (20mL), added hydrazine hydrate (2.45g, 50.1mmol), and refluxed at 80 ℃ for 2 h. After the reaction was complete, filtration was carried out, the reaction solution was concentrated under reduced pressure, and the residue was washed with acetonitrile (10mL), filtered and dried in vacuo to give the title compound. LC-MS M/z [ M + H ]]+=222.
Step 3 preparation of N' - (4-bromothiazole-2-carbonyl) -N, N-dimethylformamidine
4-bromothiazole-2-formyl prepared in the step 2Hydrazine (1.50g, 6.76mmol) was dissolved in 20mL of dimethylformamide dimethyl acetal in a 100mL one-neck flask and stirred at 95 ℃ for 2 h. After completion of the reaction, the reaction solution was washed with water (50 mL. times.3), extracted with dichloromethane (50 mL. times.3), the organic phases were combined, dried and concentrated, and the residue was slurried with ethyl acetate to give the title compound. LC-MS M/z [ M + H ]]+=277.
Step 4 preparation of 4-bromo-2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazole
N' - (4-bromothiazole-2-carbonyl) -N, N-dimethylformamidine (1.80g, 6.52mmol) from step 3 was placed in a 100mL single-neck flask, acetonitrile (20mL), acetic acid (5mL) and isopropylamine (1.93g, 32.6mmol) were added, and the mixture was stirred at 95 ℃ for 3 h. After the reaction, the reaction mixture was concentrated under reduced pressure, dissolved in water (30mL), adjusted to neutral pH, extracted with ethyl acetate (300 mL. times.5), the organic phases combined and concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=273.
Step 5 preparation of 6-bromo-2- (2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazol-4-yl) isoindolin-1-one
4-bromo-2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazole (653mg, 2.40mmol), 6-bromoisoindolin-1-one (422mg, 2.00mmol), tris (dibenzylideneacetone) dipalladium (183mg, 0.200mmol), 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl (347mg, 0.600mmol) and potassium phosphate (84.8mg, 0.400mmol) prepared in step 4 were dissolved in a microwave tube containing toluene (15mL), and microwave reaction was carried out at 120 ℃ for 1H after nitrogen substitution. After completion of the reaction, filtration was carried out by suction, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was then slurried with petroleum ether and ethyl acetate (5:1) to give the title compound. LC-MS M/z [ M + H ]]+=404.
Step 6: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazol-4-yl) -isoindolin-1-one
6-bromo-2- (2- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) thiazol-4-yl) isoindolin-1-one (403mg, 1.00mmol), 4-cyclopropyl-1 (3) H-imidazole (119mg, 1.10mmol), cesium carbonate (652mg, 2.00mmol), cuprous iodide (19.0mg, 0.100mmol), and N, N-dimethylethylenediamine (88.0mg, 0.100mmol) prepared in step 5 were dissolved in dioxane (10mL), and after nitrogen substitution, they were reacted at 135 ℃ for 3 hours with a microwave. After the reaction is completed, the reaction solution is filtered, the filtrate is diluted by water (50mL), dichloromethane (50mL multiplied by 3) is used for extraction, organic phases are combined, dried and concentrated to obtain a crude product, and the crude product is prepared and separated by preparative high performance liquid chromatography to obtain the title compound.1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),8.27(d,1H),8.10(s,1H),8.04(d,1H),7.95-7.99(m,1H),7.84-7.88(m,1H),7.67(s,1H),5.44-5.53(m,1H),5.22(s,2H),1.81-1.91(m,1H),1.58(d,6H),0.80-0.86(m,2H),0.70-0.74(m,2H).LC-MSm/z:[M+H]+=432.
Example 5: 7- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 4-dihydroisoquinolin-1 (2H) -one
Step 1 preparation of 6-bromopyridine-2-formhydrazide
In a 250mL single-neck bottle, addMethyl 6-bromopicolinate (10.0g, 46.5mmol) was dissolved in methanol (150mL), hydrazine hydrate (4.60g, 93.0mmol) was added, and the mixture was refluxed at 70 ℃ for 5 hours. After the reaction was complete, filtration was carried out, and the filter cake was washed with ethyl acetate and dried in vacuo to give the title compound. LC-MS M/z [ M + H ]]+=216.
Step 2: preparation of N' - (6-bromo-1-carbonylpyridin-2-yl) -N, N-dimethylformamidine
6-bromopyridine-2-formylhydrazine (8.60g, 40.0mmol) obtained in step 1 was dissolved in a 250mL single-necked flask containing N, N-dimethylformamide dimethyl acetal (90mL), and the reaction was refluxed for 4 hours. After completion of the reaction, the reaction solution was washed with water (100 mL. times.3), extracted with dichloromethane (100 mL. times.3), the organic phases were combined, dried and concentrated, and the residue was slurried with ethyl acetate to give the title compound. LC-MS M/z [ M + H ]]+=271.
And step 3: preparation of 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine
N' - (6-bromo-1-carbonylpyridin-2-yl) -N, N-dimethylformamidine (8.60g, 31.9mmol) from step 2 was placed in a 250mL single-neck flask, acetonitrile (60mL), acetic acid (30mL), and isopropylamine (9.40g, 159mmol) were added, and the mixture was refluxed at 80 ℃ overnight. After the reaction, the reaction mixture was concentrated under reduced pressure, dissolved in water (30mL), adjusted to neutral pH with saturated aqueous sodium bicarbonate solution, extracted with ethyl acetate (300 mL. times.5), the organic phases combined and concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=267.
And 4, step 4: preparation of 7-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 4-dihydroisoquinolin-1 (2H) -one
2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (650mg, 2.44mmol), 7-bromo-3, 4-dihydroisoquinolin-1 (2H) -one (660mg, 2.93mmol), tris (dibenzylideneacetone) dipalladium (223mg, 0.244mmol), 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl (423mg, 0.732mmol), and cesium carbonate (954mg, 0.293mmol), which were prepared in step 3, were dissolved in a 100mL single-neck flask containing toluene (15mL), and nitrogen was purged 3 times and stirred at 100 ℃ for 6 hours. After completion of the reaction, the reaction mixture was filtered with suction, and the solvent was evaporated under reduced pressure to give a crude product, which was subjected to column chromatography (petroleum ether: ethyl acetate 1:3) to isolate the title compound. LC-MS M/z [ M + H ]]+=412.
And 5: preparation of 7- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 4-dihydroisoquinolin-1 (2H) -one
The 7-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 4-dihydroisoquinolin-1 (2H) -one (200mg, 0.480mmol) obtained in step 4, 4-cyclopropyl-1H-imidazole (60.0mg, 0.556mmol), cuprous iodide (20.0mg, 0.0240mmol), 8-hydroxyquinoline (7.00mg, 0.0480mmol), and cesium carbonate (320mg, 0.960mmol) were added to a reaction flask, dissolved in 1.4-dioxane (5mL), replaced with nitrogen three times, and reacted at 135 ℃ for 4H with microwave. After completion of the reaction, filtration was carried out by suction, 5mL of water was added to the filtrate, extraction was carried out with methylene chloride (10 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the title compound was obtained by preparative separation.1H NMR(400MHz,DMSO-d6)δ8.91(s,1H),8.16(s,1H),8.11(d,1H),7.95-8.07(m,3H),7.81-7.86(m,1H),7.54-7.58(m,2H),5.35-5.43(m,1H),4.23-4.30(m,2H),3.17-3.22(m,2H),1.82-1.91(m,1H),1.51(d,6H),0.78-0.84(m,2H),0.70-0.74(m,2H).LC-MS m/z:[M+H]+=440.
Example 6: 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
Step 1 preparation of 6-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
6-Bromoisoindoline-1-one (1.00g, 4.72mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (1.26g, 4.72mmol) and potassium phosphate (2.51g, 11.8mmol) were dissolved in dioxane (30mL), and 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (Xant-phos, 272mg, 0.472mmol) and tris (dibenzylideneacetone) dipalladium Pd were added2(dba)3(431mg, 0.472mmol), replaced with nitrogen, and reacted at 130 ℃ for 70min with a microwave. After completion of the reaction, the reaction mixture was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was slurried with petroleum ether and ethyl acetate (5:1) to give the title compound. LC-MS M/z [ M + H ]]+=398.
Step 2 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
6-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one (0.500g, 1.26mmol), 4-cyclopropyl-1 (3) H-imidazole (0.163g, 1.50mmol) and cesium carbonate (821mg, 2.52mmol) prepared in step 1 were dissolved in dioxane (8mL), cuprous iodide (24.0mg, 0.126mmol) and N, N-dimethyl were addedEthylenediamine (11.1mg, 0.126mmol) was reacted at 130 ℃ for 50min with a nitrogen-substituted microwave. After completion of the reaction, the reaction mixture was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was isolated by preparative thin layer chromatography (dichloromethane: methanol ═ 15:1) to give the title compound.1HNMR(300MHz,DMSO-d6)δ8.95(s,1H),8.66(d,1H),8.28(s,1H),8.06-8.13(m,2H),7.86-8.01(m,3H),7.68(s,1H),5.50-5.57(m,1H),5.20(s,2H),1.82-1.88(m,1H),1.59(d,6H),0.80-0.85(m,2H),0.70-0.74(m,2H).LC-MS m/z:[M+H]+=426.
Example 7:2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (4-methyl-1H-imidazol-1-yl) isoindolin-1-one
The preparation process was similar to that of example 6, except that 4-cyclopropyl-1 (3) H-imidazole as a starting material was replaced with 4-methyl-1 (3) H-imidazole to give the title compound.1HNMR(300MHz,DMSO-d6)δ8.95(s,1H),8.66(d,1H),8.33(s,1H),8.06-8.13(m,2H),7.87-7.99(m,3H),7.64(s,1H),5.48-5.57(m,1H),5.20(s,2H),2.19(s,3H),1.59(d,6H).LC-MS m/z:[M+H]+=400。
Example 8:6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 3-dimethylisoindol-1-one
Step 1 preparation of 6-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 3-dimethylisoindol-1-one
6-bromo-3, 3-dimethylisoindolin-1-one (1.13g, 4.72mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (1.26g, 4.72mmol) and potassium phosphate (2.51g, 11.8mmol) were dissolved in dioxane (10mL), Xant-phos (272mg, 0.472mmol) and Pd were added2(dba)3(431mg, 0.472mmol), replaced with nitrogen, and reacted at 130 ℃ for 70min with a microwave. After completion of the reaction, the reaction mixture was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was slurried with petroleum ether and ethyl acetate (5:1) to give the title compound. LC-MS M/z [ M + H ]]+=426.
Step 2 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 3-dimethylisoindol-1-one
6-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 3-dimethylisoindol-1-one (537mg, 1.26mmol), 4-cyclopropyl-1 (3) H-imidazole (0.163g, 1.50mmol) and cesium carbonate (821mg, 2.52mmol) obtained in step 1 were dissolved in dioxane (8mL), cuprous iodide (24.0mg, 0.126mmol) and N, N-dimethylethylenediamine (11.1mg, 0.126mmol) were added, and after nitrogen substitution, microwave reaction was performed at 130 ℃ for 90 min. After completion of the reaction, the reaction mixture was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was purified by preparative thin layer chromatography (dichloromethane: methanol ═ 15:1) to give the title compound.1H NMR(300MHz,DMSO-d6)δ8.97(s,1H),8.27(s,1H),8.09-8.17(m,2H),7.92-8.03(m,4H),7.67(s,1H),5.31-5.38(m,1H),1.84-1.87(m,1H),1.77(s,6H),1.45(d,6H),0.80-0.85(m,2H),0.71-0.74(m,2H).LC-MS m/z:[M+H]+=454.
Example 9: 2-benzoyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxylic acid amide
Step 1 preparation of 3, 4-dibromomethyl-benzoate
A250 mL single neck flask was charged with methyl 3, 4-dimethylbenzoate (6.00g, 36.6mmol), benzoyl peroxide (900mg, 3.66mmol) and N-bromosuccinimide (13.0g, 73.2mmol), dissolved in chloroform (60mL), and refluxed for 20 h. After the reaction, insoluble matter was removed by suction filtration, the filtrate was concentrated under reduced pressure, the concentrated residue was dissolved in dichloromethane, washed with saturated aqueous sodium bicarbonate solution and dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation to give the title compound. LC-MS M/z [ M + H ]]+=323.
Step 2: preparation of methyl 2-benzylisoindoline-5-carboxylate
3, 4-Dibromomethylbenzoate (12.0g, 37.5mmol) was dissolved in a single-neck flask containing 120mL of tetrahydrofuran, and benzylamine (4.00g, 37.5mmol) and triethylamine (7.57g, 75.0mmol) were added, and the mixture was stirred at room temperature overnight. After the reaction, the reaction mixture was washed with water (200mL), extracted with ethyl acetate (100 mL. times.3), and the organic phases were combined, concentrated under reduced pressure, purified by column chromatography, and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=268.
And step 3: preparation of 2-benzyl isoindoline-5-carboxylic acid
In a 100mL single-neck flask, methyl 2-benzylisoindoline-5-carboxylate (4.50g, 16.9mmol) was dissolved in methanol (40mL), water (10mL) and lithium hydroxide (2.00g, 84.3mmol) were added, and the mixture was stirred at room temperature for 5 h. After the reaction was complete, the pH was adjusted to neutral with hydrochloric acid (1M), extracted with dichloromethane (100 mL. times.3), and the organic phases were combined and spin-dried to give the title compound. LC-MSm/z: [ M + H]+254, step 4: preparation of 2-benzyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Placing the 2-benzyl isoindoline-5-formic acid (2.00g, 7.90mmol) prepared in the step 3 into a 100mL single-neck bottle, adding anhydrous dichloromethane (30mL), adding oxalyl chloride (1.60g, 12.6mmol) under cooling, stirring at room temperature for 2 hours, and after the reaction is finished, concentrating the reaction solution under reduced pressure to obtain a dark green solid.
6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (1.30g, 7.10mmol) was placed in a 100mL single-necked flask, dissolved in anhydrous dichloromethane (30mL), and the above solid and N, N-diisopropylethylamine (2.00g, 15.8mmol) were added under cooling and stirred at room temperature for 3H. After completion of the reaction, the reaction solution was washed with water (50mL), dried and concentrated to give a crude product, which was separated by column chromatography (ethyl acetate: ethanol ═ 0 to 5%) to give the title compound. LC-MS M/z [ M + H ]]+=439.
And 5: preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Pd/C (60.0mg, 10%) was placed in a 100mL single-neck flask, methanol (30mL) was added, and then2-benzyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide (600mg, 1.38mmol) obtained in step 4 and 3 drops of concentrated hydrochloric acid were added, replaced with hydrogen three times, and stirred at room temperature. After the reaction was completed, filtration was carried out, and the filtrate was concentrated under reduced pressure to obtain the title compound. LC-MS M/z [ M + H ]]+=349.
Step 6: preparation of 2-benzoyl-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide
Benzoic acid (50.0mg, 0.389mmol) was placed in a 25mL single neck flask and 2- (7-azobenzotriazol) -N, N' -tetramethyluronium hexafluorophosphate (246mg, 0.646mmol) was added, N-diisopropylethylamine (90.0mg, 0.646mmol) was added with cooling, stirred for 10min, and then N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindole-5-carboxamide (90.0mg, 0.258mmol) prepared in step 5 was added. After completion of the reaction, the reaction solution was diluted with water (50mL), extracted with dichloromethane: ethanol ═ 5:1(30mL × 3), the organic phases were combined, dried and concentrated to give a crude product, which was then subjected to preparative high performance liquid chromatography (acidic, formic acid) to give the title compound.1H NMR(400MHz,DMSO-d6)δ10.77(d,1H),8.88(d,1H),8.18(d,1H),7.85-8.05(m,4H),7.64-7.66(m,2H),7.45-7.60(m,4H),5.66-5.74(m,1H),4.96(s,2H),4.87(s,2H),1.42-1.46(m,6H).LC-MS m/z:[M+H]+=453.
Example 10: 5- (4-cyclopropyl-1H-imidazol-1-yl) -1-ethyl-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -indazol-3-one
Step 1 preparation of 5-bromo-1-ethylindazol-3-one
5-bromo-indazol-3-one (3.00g, 14.1mmol) was dissolved in N, N-dimethylformamide (30mL), followed by addition of potassium carbonate (3.89g, 28.2mmol), slow dropwise addition of iodoethane (1.25mL, 15.5mmol) at 0 deg.C, and reaction at room temperature for 3 hours after completion of the dropwise addition. After completion of the reaction, the reaction mixture was filtered by suction, 10mL of water was added, extraction was performed with ethyl acetate (20mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and column chromatography (petroleum ether: ethyl acetate ═ 10:1) was performed to isolate the title compound. LC-MSm/z: [ M + H]+=241。
Step 2: preparation of 5-bromo-1-ethyl-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -indazol-3-one
5-bromo-1-ethylindazol-3-one (0.450g, 1.87mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (0.599g, 2.25mmol) were dissolved in 15mL of 1, 4-dioxane, tris (dibenzylideneacetone) dipalladium (0.172g, 0.187mmol), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (0.326g, 0.561mmol) and potassium phosphate (0.596g, 2.81mmol) were added, nitrogen was substituted three times, and microwave reaction was carried out at 105 ℃ for 35 minutes. After completion of the reaction, filtration was performed under suction, 5mL of water was added to the filtrate, extraction was performed with dichloromethane (15mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and column chromatography (petroleum ether: ethyl acetate ═ 1:2) was performed to isolate the title compound. LC-MS M/z [ M + H ]]+=427。
And step 3: preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -1-ethyl-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -indazol-3-one
5-bromo-1-ethyl-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -indazol-3-one (410mg, 0.962mmol) and 4-cyclopropyl-1H-imidazole (125mg, 1.15mmol) were dissolved in 10mL of 1, 4-dioxane, and cuprous iodide (18.0mg, 0.0962mmol), 8-hydroxyquinoline (14.0mg, 0.0962mmol) and cesium carbonate (470mg, 1.44mmol) were added, the nitrogen was replaced three times, and the reaction was carried out at 135 ℃ for 4 hours by microwave. After completion of the reaction, filtration was carried out by suction, 5mL of water was added to the filtrate, extraction was carried out with methylene chloride (10 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the title compound was obtained by preparative separation.1H NMR(400MHz,DMSO-d6)δ8.65(s,1H),8.12(t,1H),8.04(s,1H),7.92(dd,2H),7.74(dd,1H),7.70(d,1H),7.46(s,1H),7.40(d,1H),4.46-4.52(m,1H),4.40-4.44(m,2H),1.77-1.81(m,1H),1.40(t,3H),0.75-0.77(m,2H),0.70(d,3H),0.69(d,3H),0.66-0.64(m,2H)。LC-MS m/z:[M+H]+=455。
Example 11:6- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-4-carboxamide
Step 1 preparation of 6-bromo-1-methyl-1H-indole-4-carboxylic acid methyl ester
In a 100mL single-neck flask, 6-bromo-1H-indole-4-carboxylic acid methyl ester (2.00g, 7.88mmol) was added, anhydrous tetrahydrofuran (25.0mL) was added for dissolution, NaH (350mg, 8.64mmol) was added under ice-bath conditions, stirring was carried out for 15min, and CH was added3I (1.23g, 8.64mmol), reacting for 1h at room temperature, and finishing the reaction; quenching the reaction solution with water, extracting the obtained water phase with ethyl acetate, drying the ethyl acetate with anhydrous sodium sulfate, and removing the solvent by rotary evaporationThe title compound was obtained. LC-MS M/z [ M + H ]]+=268.0.
Step 2: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -1-methyl-1H-indole-4-carboxylic acid methyl ester
In a 100mL single-neck flask, 6-bromo-1-methyl-1H-indole-4-carboxylic acid methyl ester (2.50g, 9.25mmol) and 4-cyclopropyl-1H-imidazole (1.00g, 9.25mmol) were added, DMSO (dimethyl sulfoxide, 30.0mL) was added to dissolve, 2-acetyl-cyclopropanone (125mg, 0.925mmol) and potassium carbonate (K)2CO32.55g, 18.5mmol) and cuprous iodide (175mg, 0.925mmol), pumping nitrogen for 3 times, reacting at 120 ℃ for 1h, and finishing the reaction; cooling to room temperature, suction filtering, adding ethyl acetate and water for extraction, taking an organic phase, drying the organic phase by using anhydrous sodium sulfate, and carrying out silica gel column chromatography after sand making to obtain the title compound. LC-MS M/z [ M + H ]]+=296.1.
And step 3: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -1-methyl-1H-indole-4-carboxylic acid
Adding 6- (4-cyclopropyl-1H-imidazol-1-yl) -1-methyl-1H-indole-4-carboxylic acid methyl ester (700mg, 2.38mmol) into a 100mL single-neck bottle, adding methanol (15.0mL) and water (3.0mL) for dissolving, adding sodium hydroxide (100mg, 2.38mmol), reacting at 60 ℃ for 40min, and finishing the reaction; the reaction solution was adjusted to pH 1 with 2M dilute hydrochloric acid and extracted with dichloromethane/methanol 3: 1. The organic phase was dried over anhydrous sodium sulfate and the solvent was removed by rotary evaporation to give the title compound. LC-MS M/z [ M + H ]]+=282.1.
Step 4 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-4-carboxamide
In a 100mL single-neck flask, 6- (4-cyclopropyl-1H-imidazol-1-yl) -1-methyl-1H-indole-4-carboxylic acid (260mg, 0.93mmol) was added, pyridine was dissolved in 10.0mL, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (267mg, 1.40mmol) was added under ice-bath conditions, stirring was continued for 20min, 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (207mg, 1.02mmol) was added, reaction was continued for 1H under ice-bath conditions, the reaction was complete, pyridine was removed by rotation, the residue was dissolved in water with dichloromethane and 1M HCl to pH 6, extracted with dichloromethane/methanol 3:1, the organic phase was dried over anhydrous sodium sulfate, the solvent was removed by rotary evaporation to give the crude product, which was then isolated by preparative separation to give the title compound.1H NMR(300MHz,DMSO-d6)δ10.69(s,1H),8.87(s,1H),8.26(d,1H),8.19(s,1H),8.35(t,1H),7.99(s,1H),7.81-7.89(m,2H),7.62(s,1H),7.52-7.61(m,1H),6.84(d,1H),5.67(t,1H),3.90(s,3H),1.85-1.91(m,1H),1.43(d,6H),0.81-0.84(m,2H),0.71-0.81(m,2H).LC-MS m/z:[M+H]+=467.2.
Example 12: 2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (4- (trifluoromethyl) -1H-imidazol-1-yl) -3, 4-dihydroisoquinolin-1 (2H) -one
7-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3, 4-dihydroisoquinolin-1-2H-one (200mg, 0.480mmol), 4- (trifluoromethyl) -1H-imidazole (72.0mg, 0.0530mmol), cuprous iodide (10.0mg,0.0530mmol), 8-hydroxyquinoline (1.77mg, 0.0122mmol), cesium carbonate (59.8mg, 0.183mmol) were added to a reaction flask, and 1, 4-dioxane (5mL) was dissolved, replaced with nitrogen three times, and reacted at 135 ℃ for 3H. After completion of the reaction, the reaction mixture was filtered with suction, the filtrate was extracted with water (5mL) and methylene chloride (10 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and evaporated under reduced pressureThe solvent was removed and the title compound was isolated by preparative separation.1H NMR(400MHz,DMSO-d6)δ8.92(s,1H),8.54-8.58(m,2H),8.26(d,1H),8.04-8.09(m,1H),7.93-8.02(m,3H),7.63(d,1H),5.36-5.44(m,1H),4.28(t,2H),3.23(t,2H),1.51(d,6H).LC-MS m/z:[M+H]+=468.
Example 13: 2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (4- (trifluoromethyl) -1H-imidazol-1-yl) isoquinolin-1 (2H) -one
Step 1: preparation of 7-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoquinolin-1 (2H) -one
7-Bromoisoquinolin-1 (2H) -one (0.600g, 2.68mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (641mg, 2.41mmol) were dissolved in 15mL of toluene, and cuprous iodide (50.9mg, 0.268mmol) and N were added1,N2Dimethylethylene-1, 2-diamine (23.6mg, 0.268mmol) and potassium phosphate (0.682g, 3.22mmol), nitrogen substitution three times, and reaction at 120 ℃ for 1 hour by microwave. After completion of the reaction, suction filtration was carried out, the solvent was distilled off from the filtrate under reduced pressure, and column chromatography (dichloromethane: methanol ═ 50:1) was carried out to isolate the title compound. LC-MS M/z [ M + H ]]+=410。
Step 2: preparation of 2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (4- (trifluoromethyl) -1H-imidazol-1-yl) isoquinolin-1 (2H) -one
Reacting 7-bromo-2- (6- (4-isopropyl)The yl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoquinolin-1 (2H) -one (50.0mg, 0.122mmol), 4- (trifluoromethyl) -1H-imidazole (20.0mg, 0.147mmol) were dissolved in 1, 4-dioxane (10mL), cuprous iodide (2.32mg, 0.0122mmol), 8-hydroxyquinoline (1.77mg, 0.0122mmol) and cesium carbonate (59.8mg, 0.183mmol) were added, replaced with nitrogen three times, and reacted at 135 ℃ for 3 hours. After completion of the reaction, filtration was carried out by suction, 5mL of water was added to the filtrate, extraction was carried out with methylene chloride (10 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was distilled off under reduced pressure, and the title compound was obtained by preparative separation.1H NMR(400MHz,DMSO-d6)δ8.95(s,1H),8.65-8.66(m,2H),8.55(d,1H),8.20-8.24(m,3H),7.97-8.01(m,2H),7.91(d,1H),6.92(d,1H),5.29-5.36(m,1H),1.50(d,3H),1.49(d,3H)。LC-MS m/z:[M+H]+=466。
According to the synthesis of examples 1 to 13 of the present invention, the compounds of examples 14 to 27 were synthesized using different commercially available starting materials, the characterization parameters of which are shown in Table 1:
table 1:
example 15: 3- (4-cyclopropyl-1H-imidazol-1-yl) -6- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6, 7-dihydro-5H-pyrrolo [3,4-b ] pyridin-5-one
Step 1 preparation of 3-bromo-5H-pyrrolo [3,4-b ] pyridine-5, 7(6H) -dione
5-bromopyridine-2, 3-dicarboxylic acid (5.00g, 20.3mmol) was dissolved in acetic anhydride (4.41mL, 46.7mmol) and reacted at 120 ℃ for 3 h. Concentrated under reduced pressure, and ammonium acetate (3.44g, 44.7mmol) was added to the reaction solution to react at 100 ℃ for 2.5 hours. After the reaction was complete, a solid precipitated, filtered off with suction, washed with water and dried to give the title compound. LC-MS M/z [ M + H ]]+=227.
Step 2 preparation of 3-bromo-7-hydroxy-6, 7-dihydro-5H-pyrrolo [3,4-b ] pyridin-5-one
Reacting 3-bromo-5H-pyrrolo [3,4-b ]]Pyridine-5, 7(6H) -dione (2.70g, 11.9mmol) was dissolved in a mixed solvent of 15mL methanol and 15mL dichloromethane, sodium borohydride (0.681g, 17.9mmol) was added in portions at-30 ℃ and after the addition was completed, the reaction was carried out at this temperature for 0.5H. After the reaction is completed, slowly dropwise adding 2N hydrochloric acid for quenching, continuously dropwise adding and adjusting the pH value to 3, stirring for 10min, slowly dropwise adding 1M sodium hydroxide to adjust the pH value to 9, separating out a solid, performing suction filtration, drying, and performing ethanol recrystallization to obtain the title compound. LC-MS M/z [ M + H ]]+=229.
Step 3 preparation of 3-bromo-6, 7-dihydro-5H-pyrrolo [3,4-b ] pyridin-5-one
3-bromo-7-hydroxy-6, 7-dihydro-5H-pyrrolo [3, 4-b)]Pyridin-5-one (740mg, 3.24mmol) was dissolved in 10mL of dichloromethane, and triethylsilane (753mg, 6.48mmol) and trifluoroacetic acid (3.05mL, 38.9mmol) were added to react at room temperature for 0.5 hour. After the reaction is completed, the reaction solution is reacted,concentrated under reduced pressure, 5mL of water was added, the pH was adjusted to 8 with saturated sodium bicarbonate, extracted with dichloromethane (20 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=213.
Step 4 preparation of 3-bromo-6- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6, 7-dihydro-5H-pyrrolo [3,4-b ] pyridin-5-one
3-bromo-6, 7-dihydro-5H-pyrrolo [3, 4-b)]Pyridin-5-one (510mg, 2.41mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (768mg, 2.89mmol) were dissolved in 10mL of 1, 4-dioxane, cuprous iodide (46.3mg, 0.241mmol), N, N' -dimethylethylenediamine (21.2mg, 0.241mmol), cesium carbonate (1.18g, 3.62mmol) were added, nitrogen was purged, and the reaction was carried out at 120 ℃ for 1 hour under microwave irradiation. After the reaction is completed, the reaction product is filtered, the filtrate is decompressed, the solvent is removed by evaporation, and the title compound is obtained by column chromatography separation. LC-MS M/z [ M + H ]]+=399.
And 5: preparation of 3- (4-cyclopropyl-1H-imidazol-1-yl) -6- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6, 7-dihydro-5H-pyrrolo [3,4-b ] pyridin-5-one
3-bromo-6- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6, 7-dihydro-5H-pyrrolo [3, 4-b)]Pyridin-5-one (230mg, 0.578mmol), 4-cyclopropyl-1H-imidazole (93.6mg, 0.867mmol) were dissolved in 5mL of 1, 4-dioxane, cuprous iodide (11.0mg, 0.0578mmol), 8-hydroxyquinoline (8.38mg, 0.0578mmol), cesium carbonate (377mg, 1.16mmol) were added, nitrogen was charged for protection, and the reaction was carried out at 130 ℃ for 80min with microwave. After the reaction is completed, the reaction solution is filtered by suction, and the filtrate is subjected to reduced pressure evaporation to remove the solvent, and then the title compound is obtained by preparative separation.1HNMR(400MHz,DMSO-d6)δ8.95(d,1H),8.74(dd,1H),8.43(s,1H),8.21(d,1H),8.20(dd,1H),8.00(t,1H),7.86(s,1H),7.15(d,1H),5.62-5.69(m,1H),5.20(s,2H),1.93-2.00(m,1H),1.67-1.68(m,6H),0.92-0.98(m,2H),0.86-0.90(m,2H).LC-MS m/z:[M+H]+=427.
EXAMPLE 28 5- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (5-isopropyl-1H-tetrazol-1-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide
Step 1 preparation of 5-amino-2-fluoro-4-methylbenzonitrile
5-bromo-4-fluoro-2-methylaniline (10.0g, 49mmol) was dissolved in N-methylpyrrolidone (50mL), and then cuprous cyanide (8.8g, 98mmol) was added to the solution, which was heated to 180 ℃ under argon and the reaction stirred for 3 h. After cooling to room temperature, water (100mL) and ammonia (150mL) were added to the reaction mixture, and the mixture was stirred for 0.5h, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered by suction, and purified by column chromatography to give the title compound. LC-MS M/z [ M + H ]]+=151。
Step 2 preparation of 5- ((2-cyclopropyl-2-oxoethyl) amino) -2-fluoro-4-methylbenzonitrile
Dissolving 5-amino-2-fluoro-4-methylbenzonitrile (3.5g, 23.3mmol) in N, N-dimethylformamide (50mL), adding potassium carbonate (3.8g, 28mmol) and potassium iodide (4.3g, 25.6mmol), stirring at room temperature for 5min, adding 1-cyclopropyl-2-bromoethanone (8.5g, 52.5mmol), heating to 60 deg.C, and reactingAnd 3 h. The reaction solution was concentrated under reduced pressure to remove N, N-dimethylformamide, water (100mL) and ethyl acetate (150mL) were added, the mixture was separated, the organic phase was dried over anhydrous sodium sulfate, filtered by suction, and purified by column chromatography to give the title compound. LC-MS M/z [ M + H ]]+=233。
Step 3 preparation of 5- (4-cyclopropyl-2-mercapto-1H-imidazol-1-yl) -2-fluoro-4-methylbenzonitrile
5- ((2-cyclopropyl-2-oxoethyl) amino) -2-fluoro-4-methylbenzonitrile (1.0g, 4.3mmol) was dissolved in glacial acetic acid (15mL), potassium thiocyanate (0.8g, 8.6mmol) was added and heated to 110 ℃ for 4 h. The reaction solution was concentrated under reduced pressure to remove glacial acetic acid, water (100mL) and dichloromethane (150mL) were added, the mixture was separated, the organic phase was dried over anhydrous sodium sulfate, filtered under suction, and purified by column chromatography to give the title compound. LC-MS M/z [ M + H ]]+=274。
Step 4 preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzonitrile
Hydrogen peroxide (1.4mL) was added to a mixed solvent of acetic acid (18mL) and water (3.5mL), the mixture was heated to 45 ℃,5- (4-cyclopropyl-2-mercapto-1H-imidazol-1-yl) -2-fluoro-4-methylbenzonitrile (1.1g, 4mmol) was added thereto, the internal temperature was maintained at not higher than 55 ℃, and after stirring at 45 ℃ for 0.5H, the mixture was cooled to room temperature. Adding 20% sodium sulfite solution, stirring for 0.5h until the color of the starch potassium iodide test paper is not changed, adding dichloromethane (100mL), extracting and separating liquid, drying an organic phase by using anhydrous sodium sulfate, performing suction filtration, and performing concentrated column chromatography purification to obtain the title compound. LC-MS M/z [ M + H ]]+=242。
Step 5 preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzamide
5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzonitrile (0.5g, 2mmol) was dissolved in dimethyl sulfoxide (5mL), and potassium carbonate (0.5g, 4mmol) and hydrogen peroxide (1mL) were added under cooling in an ice bath, and the mixture was stirred for 10 min. Water (20mL) was added and stirred, filtered, and the filter cake was washed with water (500 mL. times.2) and dried to give the title compound. LC-MS M/z [ M + H ]]+=260。
Step 6 preparation of N- (6-chloropyridin-2-yl) isobutyramide
Isobutyryl chloride (0.1g, 1.1mmol) was dissolved in dichloromethane (5mL), and the resulting solution was added dropwise to a solution of 6-chloro-2-aminopyridine (0.1g, 1.0mmol) and N, N-diisopropylethylamine (0.4g, 3.0mmol) in dichloromethane (10mL) and the reaction was stirred at room temperature for 0.5 h. The reaction solution is decompressed and concentrated to remove the solvent, and the crude product of the title compound is obtained by concentration and is directly put into the next reaction without purification. LC-MS M/z [ M + H ]]+=199。
Step 7 preparation of 2-chloro-6- (5-isopropyl-1H-tetrazol-1-yl) pyridine
Dissolving crude N- (6-chloropyridin-2-yl) isobutyramide (0.2g, 1.0mmol) and triphenylphosphine (0.5g, 2.0mmol) in acetonitrile (5mL), stirring under the protection of argon for reaction for 0.5h, adding carbon tetrachloride (0.3g, 2mmol), and heating under reflux for 4 h. After cooling to room temperature, trimethylsilyl azide (0.2g, 1.5mmol) was slowly added dropwise thereto, followed by heating and refluxing for 8 hours. The reaction was quenched with 5% aqueous sodium bicarbonate, water (50mL) and dichloromethane (50mL) were added, the layers were separated, the organic layer was dried over anhydrous sodium sulfate,and (5) carrying out suction filtration, and carrying out column chromatography purification by concentration to obtain a title compound. LC-MS M/z [ M + H ]]+=224。
Step 8 preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (5-isopropyl-1H-tetrazol-1-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide
5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzamide (0.1g, 0.5mmol), 2-chloro-6- (5-isopropyl-1H-tetrazol-1-yl) pyridine (0.1g, 0.5mmol), tris (dibenzylideneacetone) dipalladium (0.05g, 0.05mmol), 4, 5-bis diphenylphosphino-9, 9-dimethylxanthene (0.03g, 0.05mmol), cesium carbonate (0.3g, 1mmol) were added to dioxane (10mL) and then warmed to 110 ℃ under argon for 3H. Water (50mL) and dichloromethane (50mL) were added, the layers were separated, the organic phase was dried over anhydrous sodium sulfate, filtered with suction, and purified by column chromatography to give the title compound.1H NMR(400MHz,DMSO-d6)δ11.19(s,1H),8.33(d,J=8.3Hz,1H),8.23(d,J=7.9Hz,1H),7.68(dd,J=25.9,7.3Hz,3H),7.47(d,J=10.8Hz,1H),7.18(s,1H),3.98–3.80(m,1H),2.24(s,3H),1.94–1.79(m,1H),1.30(d,J=6.8Hz,6H),0.80(dd,J=5.4,2.7Hz,2H),0.69(d,J=2.8Hz,2H).LC-MS m/z[M+H]+=447.2。
Example 29 5- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (1-isopropyl-1H-tetrazol-5-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide
Step 1 preparation of 6-bromo-N-isopropylpicolinamide
6-bromo-2-pyridinecarboxylic acid (3.0g, 15mmol) was added to thionyl chloride (11mL), heated to 85 ℃ under argon protection, and the reaction was stirred for 2 h. The reaction mixture was cooled to room temperature, and the reaction mixture was concentrated under reduced pressure to remove thionyl chloride. To the resulting product was added dichloromethane (100mL), cooled to-10 deg.C, a solution of N-ethyldiisopropylamine (9.5g, 74mmol) and isopropylamine (2.6g, 44mmol) in dichloromethane (10mL) was added, and the reaction was stirred at room temperature for 0.5 h. Adding water (50mL) to quench the reaction, separating the liquid, drying the organic phase by using anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to obtain the 6-bromo-N-isopropyl picolinamide. LC-MS M/z [ M + H ]]+=243。
Step 2 preparation of 2-bromo-6- (1-isopropyl-1H-tetrazol-5-yl) pyridine
6-bromo-N-isopropylpicolinamide (0.5g, 2.0mmol) was dissolved in acetonitrile (5mL), cooled to-15 deg.C and trifluoromethanesulfonic anhydride (0.7g, 4mmol) was added. After stirring for 10min, trimethylsilyl azide (1.1mL, 8.3mmol) was slowly added dropwise, and the reaction was allowed to return to room temperature for 1.5 h. Adding 5% sodium bicarbonate aqueous solution to quench the reaction, adding water (50mL) and dichloromethane (50mL), separating, drying the organic phase with anhydrous sodium sulfate, filtering, concentrating, and purifying by column chromatography to obtain 2-bromo-6- (1-isopropyl-1H-tetrazol-5-yl) pyridine. LC-MS M/z [ M + H ]]+=268。
Step 3 preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (1-isopropyl-1H-tetrazol-5-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide
5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzamide (0.2g, 0.8mmol), 2-bromo-6- (1-isopropyl-1H-tetrazol-5-yl) pyridine (0.2g, 0.7mmol), tris (dibenzylidendene)Acetone) dipalladium (0.07g, 0.08mmol), 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene (0.04g, 0.07mmol), cesium carbonate (0.5g, 1.5mmol) were added to dioxane (10mL) and then heated to 110 ℃ under argon for 2h reaction. Water (50mL) and dichloromethane (50mL) were added, the organic phase was separated, dried over anhydrous sodium sulfate, filtered by suction, concentrated, and purified by column chromatography to give 5- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (1-isopropyl-1H-tetrazol-5-yl) pyridin-2-yl) -2-fluoro-4-methylbenzamide.1H NMR(400MHz,DMSO-d6)δ11.08(s,1H),8.29(d,J=8.3Hz,1H),8.13(t,J=8.0Hz,1H),8.00(d,J=7.5Hz,1H),7.70(s,1H),7.66(d,J=6.6Hz,1H),7.50(d,J=10.8Hz,1H),7.19(s,1H),6.07–5.91(m,1H),2.25(s,3H),1.91–1.79(m,1H),1.54(d,J=6.6Hz,6H),0.87–0.76(m,2H),0.74–0.65(m,2H).LC-MS m/z[M+H]+=447.2。
Example 30: 7- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) benzo [ d ] [1,3] dioxole-5-carboxamide
Step 1 preparation of 7-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) benzo [ d ] [1,3] dioxole-5-carboxamide
Reacting 7-bromobenzo [ d][1,3]Dioxole-5-carboxylic acid (300mg, 1.23mmol), 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (250mg, 1.23mmol) and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (934mg, 2.46mmol) were dissolved in 10mL of dichloromethane and N, N-diisopropylethylamine (397mg, 3.07mmol) was added and reacted at 25 ℃ for 2-3 hours. After completion of the reaction, the reaction mixture was washed with water (100mL) and dichloromethane (50 mLx)3) And (3) extracting, combining organic phases, drying and concentrating to obtain a crude product, and separating by using a column (ethyl acetate: petroleum ether is 30-100%) to obtain the title compound. LC-MS M/z [ M + H ]]+=430.
Step 2 preparation of 7- (4-cyclopropyl-1H-imidazol-1-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) benzo [ d ] [1,3] dioxole-5-carboxamide
The 7-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) benzo [ d ] prepared in the step 1][1,3]Dioxole-5-carboxamide (230mg, 0.535mmol), 4-cyclopropyl-1 (3) H-imidazole (0.163g, 1.50mmol), cesium carbonate (348mg, 1.07mmol) were dissolved in 1, 4-dioxane (8mL), CuI (24.0mg, 0.126mmol) and N, N-dimethylethylenediamine (11.1mg, 0.126mmol) were added, and after nitrogen substitution, the mixture was reacted at 130 ℃ for 90min with a microwave. After completion of the reaction, the reaction solution was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), the combined organic phases were dried and concentrated to give a crude product, which was isolated by prep-TLC (DCM: methanol ═ 15:1) to give the title compound.1H NMR(400MHz,DMSO-d6)δ10.73(s,1H),8.89(s,1H),8.16(d,2H),8.02-8.06(m,1H),7.84(d,2H),7.51-7.56(m,2H),6.30(s,2H),5.58-5.65(m,1H),1.86-1.93(m,1H),1.44(d,6H),0.82-0.86(m,2H),0.71-0.76(m,2H).LC-MS m/z:[M+H]+=458.
Example 31: 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methyl-N- (5-methyl-5, 6-dihydrobenzo [ f ] [1,2,4] triazolo [4,3-d ] [1,4] oxazepin-8-yl) benzamide
Step 1 preparation of methyl 2- (2- ((tert-butoxycarbonyl) amino) propoxy) -3-nitrobenzoate
In a 100mL single neck flask, N-Boc-DL-alanine (2.90g, 16.6mmol) was added, dissolved in tetrahydrofuran (30mL), and sodium hydride (540mg, 22.5mmol) was added under ice-water bath, followed by stirring for 10min, and 2-fluoro-3-nitrobenzyl ether (3.00g, 15.0mmol) was added, followed by stirring at room temperature. After the reaction was complete, it was washed with water (50mL), extracted with ethyl acetate (3X 50mL), and the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=355.
Step 2 preparation of (2- ((tert-butoxycarbonyl) amino) propoxy) -3-nitrobenzoic acid
Methyl 2- (2- ((tert-butoxycarbonyl) amino) propoxy) -3-nitrobenzoate (1.50g, 4.23mmol) obtained in step 1 was dissolved in a 50mL single-necked flask containing 20mL of methanol, and water (4mL) and lithium hydroxide (516mg, 21.6mmol) were added and the mixture was stirred at room temperature. After the reaction was completed, the pH was adjusted to neutral with 1N hydrochloric acid, dichloromethane/isopropanol (3X 50mL) was extracted, the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and dried under vacuum to give the title compound. LC-MS M/z [ M-H]-=339.
Step 3 preparation of 2- (2-aminopropoxy) -3-nitrobenzoic acid
(2- ((tert-butoxycarbonyl) amino) propoxy) -3-nitrobenzoic acid (1.50g, 4.42mmol) obtained in step 2 was dissolved in a 100mL single-necked flask containing 30mL of dichloromethane, and trifluoroacetic acid (10mL) was added and stirred at room temperature. After the reaction is completed, the reaction solution is decompressed and concentrated to obtainThe title compound was used directly in the next reaction. LC-MS M/z [ M + H ]]+=241.
Step 4 preparation of 3-methyl-9-nitro-3, 4-dihydrobenzo [ f ] [1,4] oxazepin-5 (2H) -one
The 2- (2-aminopropoxy) -3-nitrobenzoic acid (480mg, 2.00mmol) obtained in step 3 was placed in a 100mL single-neck flask, dissolved in dichloromethane (60mL), and 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (912mg, 2.40mmol) and N, N-diisopropylethylamine (516mg, 4.00mmol) were added and stirred at room temperature overnight. After the reaction was complete, it was washed with water (30mL), extracted with dichloromethane (5X 30mL), the organic phases were combined, concentrated under reduced pressure, and the crude product was slurried with ethyl acetate to give the title compound. LC-MS M/z [ M + H ]]+=223.
Step 5 preparation of 3-methyl-9-nitro-3, 4-dihydrobenzo [ f ] [1,4] oxazepine-5 (2H) -thione
Reacting 3-methyl-9-nitro-3, 4-dihydrobenzo [ f][1,4]Oxazan-5 (2H) -one (222mg, 1.00mmol) was placed in a 25mL single vial, toluene (15mL), Lawson's reagent (606mg, 1.5mmol) was added, and the reaction was allowed to proceed overnight at 110 ℃. After completion of the reaction, it was washed with water (50mL), extracted with ethyl acetate (3X 50mL), combined organic phases dried and concentrated to give the crude product which was slurried with dichloromethane to afford the title compound. LC-MS M/z [ M + H ]]+=239.
Step 6: preparation of (Z) -5-hydrazono-3-methyl-9-nitro-2, 3,4, 5-tetrahydro-benzo [ f ] [1,4] oxazepine
3-methyl-9-nitro-3, 4-dihydrobenzo [ f ] prepared in the step 5][1,4]Oxazan-5 (2H) -thione (150mg, 0.630mmol) was dissolved in a 25mL one-necked flask with 5mL ethanol, hydrazine hydrate (94.0mg, 1.89mmol) was added, and the mixture was stirred at 40 ℃. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, the resulting residue was washed with water (30mL), extracted with dichloromethane (2X 50mL), and the combined organic phases were dried and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=237.
And 7: preparation of 5-methyl-8-nitro-5, 6-dihydrobenzo [ f ] [1,2,4] triazolo [4,3-d ] [1,4] oxazepine
Subjecting the (Z) -5-hydrazono-3-methyl-9-nitro-2, 3,4, 5-tetrahydro-benzo [ f ] obtained in step 6][1,4]Oxazane (120mg, 0.500mmol) was dissolved in 5mL triethyl orthoformate in a 25mL single neck flask and stirred at 90 ℃ for 3 h. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the obtained residue was slurried with ethyl acetate to obtain the title compound. LC-MS M/z [ M + H ]]+=247.
And 8: preparation of 5-methyl-5, 6-dihydrobenzo [ f ] [1,2,4] triazolo [4,3-d ] [1,4] oxazepin-8-amine
The 5-methyl-8-nitro-5, 6-dihydrobenzo [ f ] prepared in the step 7][1,2,4]Triazolo [4,3-d][1,4]Oxazane (80.0mg, 0.325mmol) was dissolved in 5mL of methanol in a 25mL single-necked flask, 10% Pd/C (10mg) was added, and the mixture was stirred at room temperature. After the reaction was complete, filtration was carried out, and the filtrate was concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=217.
And step 9: preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methyl-N- (5-methyl-5, 6-dihydrobenzo [ f ] [1,2,4] triazolo [4,3-d ] [1,4] oxazepin-8-yl) benzamide
5- (4-cyclopropyl-1H-imidazol-1-yl) -2-fluoro-4-methylbenzoic acid (86.0mg, 0.330mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (115mg, 0.600mmol) were placed in a 25mL single vial, 3mL pyridine was added, stirred in an ice-water bath for 15min, then 5-methyl-5, 6-dihydrobenzo [ f][1,2,4]Triazolo [4,3-d][1,4]A solution of oxazel-8-amine (65.0mg, 0.300mmol) in pyridine (5mL) was reacted at room temperature. After the reaction was completed, the reaction solution was diluted with water (50mL), extracted with dichloromethane: ethanol ═ 5:1(3 × 30mL), and the combined organic phases were dried and concentrated to obtain a crude product, which was then subjected to preparative high performance liquid chromatography to obtain the title compound.1HNMR(400MHz,DMSO-d6)δ9.85(d,1H),8.79(s,1H),8.24-8.27(m,1H),8.11-8.15(m,1H),7.67-7.77(m,2H),7.49(d,1H),7.18-7.25(m,2H),4.52-4.58(m,1H),4.34-4.44(m,2H),2.24(s,3H),1.82-1.89(m,1H),1.51(d,3H),0.780-0.850(m,2H),0.680-0.730(m,2H).LC-MS m/z:[M+H]+=459.
Example 32: 5- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
Step 1 preparation of 5-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
5-Bromoisoindolin-1-one (600mg, 2.84mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (718mg, 2.70mmol) were dissolvedTo 10mL of 1, 4-dioxane were added cuprous iodide (54.0mg, 0.284mmol), N, N' -dimethylethylenediamine (25.0mg, 0.284mmol), potassium phosphate (903mg, 4.26mmol), and the mixture was reacted at 120 ℃ for 3 hours under nitrogen atmosphere. After the reaction is completed, the reaction product is filtered, the filtrate is decompressed, the solvent is removed by evaporation, and the title compound is obtained by column chromatography separation. LC-MS M/z [ M + H ]]+=398.
Step 2: preparation of 5- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
5-bromo-2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one (400mg, 1.01mmol), 4-cyclopropyl-1H-imidazole (131mg, 1.21mmol) were dissolved in 10mL of 1, 4-dioxane, cuprous iodide (19.2mg, 0.101mmol), 8-hydroxyquinoline (14.6mg, 0.101mmol), cesium carbonate (493mg, 1.51mmol) were added, nitrogen was purged, and the reaction was carried out at 130 ℃ for 5H by microwave. After the reaction is completed, the reaction solution is filtered by suction, and the filtrate is subjected to reduced pressure evaporation to remove the solvent, and then the title compound is obtained by preparative separation.1H NMR(400MHz,DMSO-d6)δ8.96(s,1H),8.64(d,1H),8.30(s,1H),8.09(t,1H),8.04(s,1H),7.87-7.97(m,2H),7.84(d,1H),7.67(s,1H),5.51-5.58(m,1H),5.20(s,2H),1.84-1.91(m,1H),1.59-1.61(m,6H),0.82-0.86(m,2H),0.71-0.74(m,2H).LC-MS m/z:[M+H]+=426.
Example 33: 6- (4-cyclopropyl-1H-imidazol-1-yl) -3, 3-dimethyl-2- (2- (4- (1,1, 1-trifluoropropan-2-yl) -4H-1,2, 4-triazol-3-yl) thiazol-4-yl) isoindolin-1-one
Step 1 preparation of 4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazole-2-carboxamide
In a 100mL single-neck flask, 4-bromo-1, 3-thiazole-2-carboxylic acid (2.06g, 10.0mmol) was added, dichloromethane (30mL) was added for dissolution, thionyl chloride (1.19g, 10.0mmol) was added under ice water bath, LC-MS detection of the completion of the reaction was performed, and the reaction solution was concentrated under reduced pressure. The above residue was dissolved in methylene chloride, and 2-amino-1, 1, 1-trifluoropropane hydrochloride (1.50g, 10.0mmol) and triethylamine (1ml) were added thereto, followed by stirring at room temperature. After the reaction was complete, it was washed with water (50mL), extracted with ethyl acetate (3X 50mL), and the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the title compound. LC-MS M/z [ M + H ]]+=303.
Step 2 preparation of 4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazole-2-thiocarboxamide
The 4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazole-2-carboxamide (2.00g, 6.62mmol) prepared in step 1 was dissolved in a 100mL single-necked flask containing 30mL of toluene, and Lawson's reagent (4.00g, 4.96mmol) was added thereto, followed by reflux reaction overnight. After completion of the reaction, the reaction mixture was washed with water (50mL), extracted with ethyl acetate (3X 50mL), the combined organic phases were dried and concentrated, and the crude product was subjected to column chromatography (petroleum ether) to afford the title compound. LC-MS M/z [ M + H ]]+=319.
Step 3 preparation of 4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazol-2-aminomethanamidine
The 4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazole-2-thiocarboxamide prepared in step 2 (2.00g, 6.29mmol) was dissolved in a 100mL single-necked flask with 30mL hydrazine hydrate and refluxed for 2 h. The reaction is completeAfter that, water (50mL) was washed, extracted with ethyl acetate (3X 50mL), the organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the resulting crude product was subjected to column chromatography (ethyl acetate) to give the title compound. LC-MS M/z [ M + H ]]+=317.
Step 4 preparation of 4-bromo-2- (4- (1,1, 1-trifluoropropan-2-yl) -4H-1,2, 4-triazol-3-yl) thiazole
4-bromo-N- (1,1, 1-trifluoroprop-2-yl) thiazol-2-aminomethanamidine (1.45g, 4.59mmol) obtained in step 3 was dissolved in a 50mL single-neck flask containing 20mL triethyl orthoformate and stirred at 90 ℃ for 3 h. After the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was subjected to column chromatography (petroleum ether: ethyl acetate 1:1) to give the title compound. LC-MS M/z [ M + H ]]+=327.
Step 5 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (4-methoxybenzyl) -3, 3-dimethylisoindolin-1-one
6-bromo-2- (4-methoxybenzyl) -3, 3-dimethylisoindolin-1-one (1.20g, 3.34mmol) was dissolved in a 100mL single-necked flask containing N-methylpyrrolidone (20mL), and potassium carbonate (922mg, 6.69mmol), 2-acetylcyclohexylketone (47mg, 0.334mmol), and 4-cyclopropyl-1H-imidazole (397mg, 3.67mmol) were added and stirred at 150 ℃ overnight. After the reaction, the reaction mixture was washed with water (50mL), extracted with ethyl acetate (3X 50mL), the combined organic phases were dried and concentrated, and the resulting crude product was subjected to column chromatography (petroleum ether: ethyl acetate 1:2) to give the title compound. LC-MS M/z [ M + H ]]+=388.
Step 6: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -3, 3-dimethylisoindolin-1-one
6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (4-methoxybenzyl) -3, 3-dimethylisoindolin-1-one (600mg, 1.55mmol) obtained in step 5 was placed in a closed-cell flask, and trifluoroacetic acid (5mL) was added to react at 160 ℃ for 6H. After the reaction was completed, the reaction mixture was concentrated under reduced pressure, the pH was adjusted to neutral, extracted with dichloromethane (3X 50mL), and the combined organic phases were dried and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=268.
And 7: preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -3, 3-dimethyl-2- (2- (4- (1,1, 1-trifluoropropan-2-yl) -4H-1,2, 4-triazol-3-yl) thiazol-4-yl) isoindolin-1-one
6- (4-cyclopropyl-1H-imidazol-1-yl) -3, 3-dimethylisoindolin-1-one (100mg, 0.375mmol) obtained in step 6 was placed in a microwave tube, and 4-bromo-2- (4- (1,1, 1-trifluoroprop-2-yl) -4H-1,2, 4-triazol-3-yl) thiazole (146mg, 0.450mmol) obtained in step 4, cuprous iodide (14.0mg, 0.0750mmol), N-dimethylethylenediamine (6.60mg, 0.0750mmol), potassium carbonate (103mg, 0.750mmol), dioxane (8mL), nitrogen gas purged 3 times, and reacted at 110 ℃ for 80 min. After the reaction was complete, the reaction was filtered, the filtrate was washed with water (30mL), extracted with dichloromethane (3X 30mL), the combined organic phases were dried and concentrated to give the crude product, which was then prepared by preparative high performance liquid chromatography to give the title compound.1H NMR(400MHz,DMSO-d6)δ9.32(s,1H),8.32(s,1H),8.27(s,1H),7.95-8.04(m,3H),7.68(s,1H),6.31-6.42(m,1H),1.89(d,3H),1.86(s,3H),1.76(s,3H),1.20-1.27(m,1H),0.790-0.870(m,2H),0.690-0.770(m,2H).LC-MS m/z:[M+H]+=514.
Example 34: 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5-morpholinoisoindolin-1-one
Step 1 preparation of methyl 4-fluoro-2-methylbenzoate
4-fluoro-2-methylbenzoic acid (25.0g, 162mmol) was dissolved in 300mL of methanol, thionyl chloride (23.1g, 194mmol) was slowly added thereto at 0 ℃ and after the addition, the reaction was transferred to an oil bath at 60 ℃ and heated for 2 hours. After the reaction was complete, the reaction solution was washed with water (300mL), extracted with ethyl acetate (150 mL. times.3), and the combined organic phases were dried and concentrated to give the title compound.
Step 2 preparation of methyl 2- (bromomethyl) -4-fluorobenzoate
Methyl 4-fluoro-2-methylbenzoate (25.0g, 148mmol), N-bromosuccinimide (31.6g, 178mmol) were dissolved in 300mL of carbon tetrachloride and benzoyl peroxide (3.58g, 14.8mmol) was added and the reaction was heated in an oil bath at 80 ℃ for 16 hours. After the reaction was completed, the reaction solution was filtered, and the crude product obtained by spin-drying the filtrate was separated by column (petroleum ether: ethyl acetate 10:1) to obtain the title compound. LC-MS M/z [ M + H ]]+=247.
Step 3 preparation of 5-fluoroisoindolin-1-one
Methyl 2- (bromomethyl) -4-fluorobenzoate (31.6g, 128mmol) was dissolved in 150mL of methanol, and 50mL of aqueous ammonia was added thereto, and the reaction was stirred at room temperature for 2 hours. Inverse directionAnd after the reaction is completed, removing methanol from the reaction solution to separate out a white solid, filtering, and drying a filter cake to obtain the title compound. LC-MS M/z [ M + H ]]+=152.
Step 4 preparation of 5-fluoro-6-nitroisoindol-1-one
The 5-fluoroisoindolin-1-one (16.0g, 105mmol) prepared in step 3 was dissolved in 150mL of concentrated sulfuric acid, fuming nitric acid (5.14mL, 126mmol) was slowly added at 0 ℃ and the reaction was carried out at that temperature for 2 hours. After the reaction is completed, the reaction solution is slowly poured into ice water (300mL) and stirred, light yellow solid is separated out and filtered, and a filter cake is dried to obtain the title compound. LC-MS M/z [ M + H ]]+=197.
Step 5 preparation of 5-morpholino-6-nitroisoindol-1-one
5-fluoro-6-nitroisoindol-1-one (1.00g, 5.07mmol), morpholine (661mg,7.61mmol), and potassium carbonate (1.40g, 10.1mmol) were dissolved in 15mL of N-dimethylformamide and reacted with stirring at room temperature for 1 hour. After completion of the reaction, the reaction mixture was slowly poured into ice water (100mL), extracted with ethyl acetate (50 mL. times.3), and the combined organic phases were dried and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=264.
Step 6 preparation of 5-morpholino-6-nitro-1-oxoisoindoline-2-carboxylic acid tert-butyl ester
5-Morpholino-6-nitroisoindol-1-one (1.20g, 4.54mmol), di-tert-butyl dicarbonate (1.18g,5.45mmol) were dissolved in 15mL of dichloromethane, N-dimethyl-4-pyridylamine (55mg,0.454mmol) was stirred at room temperature for 1 hour. After the reaction is completed, the reaction solution is washed by adding water, separated, dried and concentrated by an organic phase to obtain a title compound. LC-MS M/z [ M + H ]]+=364.
Step 7 preparation of tert-butyl 6-amino-5-morpholino-1-oxoisoindoline-2-carboxylate
5-Morpholino-6-nitro-1-oxoisoindoline-2-carboxylic acid tert-butyl ester (1.40g, 3.84mmol) and Pd/C (140mg) were dissolved in 20mL of methanol, and the reaction was stirred at room temperature for 1 hour under a hydrogen atmosphere. After the reaction was complete, the reaction was filtered and the filtrate was dried and concentrated to give the title compound. LC-MS M/z [ M + H ]]+=334.
Step 8 preparation of tert-butyl 6- ((2-cyclopropyl-2-oxoethyl) amino) -5-morpholino-1-oxoisoindoline-2-carboxylate
Tert-butyl 6-amino-5-morpholino-1-oxoisoindoline-2-carboxylate (1.21g, 3.62mmol), potassium carbonate (750mg, 5.43mmol), and potassium iodide (720mg, 4.34mmol) obtained in step 7 were dissolved in 15ml of N-dimethylformamide and stirred at room temperature under nitrogen for 10 min. 2-Bromocyclopropyl-1-one (703mg,4.34mmol) was added and reacted at 60 ℃ for 4 hours. After completion of the reaction, the reaction mixture was cooled and poured into ice water (100mL), extracted with ethyl acetate (50mL × 3), and the combined organic phases were dried and concentrated to obtain a crude product, which was then separated by column (petroleum ether: ethyl acetate ═ 1:1) to obtain the title compound. LC-MS M/z [ M + H ]]+=416.
Step 9 preparation of tert-butyl 6- (N- (2-cyclopropyl-2-oxoethyl) carboxamido) -5-morpholino-1-oxoisoindoline-2-carboxylate
Tert-butyl 6- ((2-cyclopropyl-2-oxoethyl) amino) -5-morpholino-1-oxoisoindoline-2-carboxylate (0.920g, 2.21mmol) from step 8 was dissolved in a mixture of 5mL acetic anhydride and 10mL formic acid at 0 ℃ and reacted at this temperature for 30 min. After the reaction is completed, the reaction solution is spun dry to obtain a crude product, the crude product is dissolved by adding water (100mL), the solution is neutralized by sodium bicarbonate until the pH value is 8, ethyl acetate (50mL multiplied by 3) is used for extraction, and the combined organic phases are dried and concentrated to obtain the crude product which is directly used in the next step. LC-MS M/z [ M + H ]]+=444.
Step 10 preparation of tert-butyl 6- (4-cyclopropyl-1H-imidazol-1-yl) -5-morpholino-1-oxoisoindoline-2-carboxylate
6- (N- (2-cyclopropyl-2-oxoethyl) carboxamido) -5-morpholino-1-oxoisoindoline-2-carboxylic acid tert-butyl ester (1.00g, crude) was dissolved in 15ml of AcOH and NH was added4OAc (867mg,11.2mmol), reacted at 110 ℃ for 4 hours. After the reaction is completed, the reaction solution is spin-dried to obtain a crude product, and the crude product is dissolved by adding water (100mL), and NaHCO is added3The solution was neutralized to pH 8, extracted with ethyl acetate (50mL × 3), the combined organic phases were dried and concentrated to give the crude product, which was isolated by column (petroleum ether: ethyl acetate 1:2) to give the title compound. LC-MS M/z [ M + H ]]+=425.
Step 11 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -5-morpholinoisoindolin-1-one
Tert-butyl 6- (4-cyclopropyl-1H-imidazol-1-yl) -5-morpholino-1-oxoisoindoline-2-carboxylate (490mg, 1.15mmol) obtained in step 10 was dissolved in 10mL of EDCM, and TFA (3mL) was added and reacted at room temperature for 1 hour. After the reaction is completed, the reaction solution is spin-dried to obtain crude productDissolving in water (100mL), NaHCO3The solution was neutralized to pH 8, extracted with ethyl acetate (50mL × 3), the combined organic phases were dried and concentrated to give the crude product, which was isolated via column (ethyl acetate) to give the title compound. LC-MS M/z [ M + H ]]+=325
Step 12 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5-morpholinoisoindolin-1-one
The 6- (4-cyclopropyl-1H-imidazol-1-yl) -5-morpholinoisoindolin-1-one (230mg, 0.707mmol), 2-bromo-6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridine (188mg, 0.707mmol), Cs prepared in step 11 was added2CO3(460mg, 1.41mmol) was dissolved in dioxane (8mL), and CuI (24.0mg, 0.126mmol) and N, N-dimethylethylenediamine (11.1mg, 0.126mmol) were added, and the mixture was reacted with nitrogen by microwave at 130 ℃ for 90 min. After completion of the reaction, the reaction solution was filtered, the filtrate was diluted with water (50mL), extracted with dichloromethane (50mL × 3), and the combined organic phases were dried and concentrated to give the crude product, which was purified by prep-TLC (DCM: methanol ═ 15:1) to afford the title compound.1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.62(d,1H),8.05-8.09(m,1H),7.90-7.95(m,2H),7.65(s,1H),7.50(s,1H),7.35(s,1H),5.49-5.59(m,1H),5.14(s,2H),3.62(d,4H),2.73(d,4H),1.86-1.92(m,1H),1.59(d,6H),0.80-0.85(m,2H),0.68-0.72(m,2H).LC-MS m/z:[M+H]+=511.
Example 27: 5- ((1S,4S) -2-oxa-5-azabicyclo [2.2.1] heptan-5-yl) -6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) isoindolin-1-one
Preparation methodIn analogy to the preparation of example 34, the starting material morpholine was replaced by (1S,4S) -2-oxa-5-azabicyclo [2.2.1]Heptane hydrochloride to yield the title compound.1H NMR(400MHz,DMSO-d6)δ8.93(s,1H),8.62(d,1H),8.03-8.07(m,1H),7.88(d,1H),7.68(s,1H),7.49(s,1H),7.23(s,1H),7.12(s,1H),5.48-5.55(m,1H),5.10(d,2H),4.50(s,1H),4.15(s,1H),3.84(d,1H),3.71(d,1H),2.70(d,2H),1.80-1.85(m,3H),1.59(d,6H),0.79-0.82(m,2H),0.67-0.70(m,2H).LC-MS m/z:[M+H]+=523.
Example 35: 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (1-isopropyl-1H-tetrazol-5-yl) pyridin-2-yl) -3, 3-dimethylisoindolin-1-one
Step 1 preparation of 6- (4-cyclopropyl-1H-imidazol-1-yl) -2- (6- (1-isopropyl-1H-tetrazol-5-yl) pyridin-2-yl) -3, 3-dimethylisoindolin-1-one
6- (4-cyclopropyl-1H-imidazol-1-yl) -3, 3-dimethylisoindolin-1-one (100mg,0.374mmol), 2-bromo-6- (1-isopropyl-1H-tetrazol-5-yl) pyridine (100mg,0.373mmol) were added sequentially to the microwave tube; adding 5mL of 1, 4-dioxane, stirring and dissolving; cesium carbonate (243mg,0.746mmol), cuprous iodide (14.2mg,0.0746mmol), N' -dimethylethylenediamine (7.00mg,0.0746mmol) were then added in that order; the reaction was carried out at 130 ℃ for 75 min. Preparative isolation by prep-TLC (DCM: methanol ═ 30:1) gave the title compound.1H NMR(400MHz,DMSO-d6)δ8.24(dd,3H),8.12–8.00(m,3H),7.94(d,1H),7.68(s,1H),5.66(dq,1H),1.87(td,1H),1.84–1.71(m,6H),1.60(t,6H),0.86–0.79(m,2H),0.76–0.69(m,2H).LC-MS m/z:[M+H]+=455.
Experimental example 1 inhibition of apoptosis Signal-regulated kinase 1(ASK1) in vitro
1. Experimental Material
1.1 kinase: ASK1, purchased from Carna Bioscience;
1.2 reagent: poly (Glu, Tyr) sodium salt, available from sigma;
staurosporine, purchased from bioanstralis;
ADP-Glo Kinase Assay, available from Promega;
1.3 test compounds are compounds of the invention prepared in the above examples.
2. Experimental methods
2.1 dilution of the Compound
1) Diluting the test compound to 1.11mM or 1mM using DMSO;
2) compounds were diluted to 11 concentrations using a TECAN EVO200 system, 3-fold gradient;
3) echo550 was used to transfer 20nL of compound to 384-well plates. The highest concentration of test compound used for the kinase ASK1 assay was 1.11 μ M or 1 μ M.
2.2 enzyme reaction experiments
1) Configuring a 1.3 x enzyme reaction system as shown in table 2, wherein the system comprises kinase, a fluorescence labeling substrate and factors required by the reaction;
2) to the reaction well, 15. mu.L of 1.3 Xenzyme reaction system was added and incubated at room temperature for 30 minutes.
TABLE 2.1.3 Xenzyme reaction System
3) The enzyme reaction was initiated by adding 5. mu.L of 4 × ATP solution to each well as described in Table 3.
TABLE 3.4 XATP solution
4) After the reaction system was left at 25 ℃ for 90 minutes, 20. mu.L of LADP-Glo Reagent was added and incubated for 40 min.
5) 40 μ LKINAse Detection Reagent was added and incubated for 35min, and fluorescence was read by EnSpire.
3. Experimental data processing
1) The residual enzyme activity (% RemainingActivity) was calculated using the following formula:
high Control (HC): DMSO; low Control (LC): 1 mu M of staurosporine;
2) computing IC Using XLfit50The value is obtained.
4 results of the experiment (see Table 4)
TABLE 4
"-" indicates not measured
The experimental results show that the compound has good inhibitory activity on ASK 1.
Experimental example 2 evaluation of liver distribution in mice
1. Experimental Material
1.1 animals
Male BALB/c mice, SPF grade, purchased from changzhou calvens laboratory animals ltd; 17-21g, license number: SCXK (threo) 2016-; an adaptation period of 2-3 days was given before the experiment.
1.2 Primary reagents
Methanol and acetonitrile were purchased from Merck; absolute ethyl alcohol, PEG200 and physiological saline were purchased from south kyo kaki biotechnology development ltd; nordiphenhydramine is available from biotechnology limited, shanghai.
1.3 instruments
API model 4000 triple quadrupole LC MS and Analyst QS A01.01 chromatography workstation were purchased from AB SCIEX, USA; Milli-Q ultrapure water was purchased from Millipore corporation; CF16R XII bench-top high speed refrigerated centrifuge from Hitachi; the Qilinbeier Vortex-5 oscillator was purchased from IKA, Germany; the electric heating constant temperature water bath kettle is purchased from China electric appliance, Inc. of Changzhou; electric pipettors were purchased from Thermo corporation, usa; microanalysis balances were purchased from Shanghai Meltrier, Inc.
2. Experimental methods
2.1 preparation of test drugs
Weighing 1.5mg (calculated by free base) of a test compound, adding the test compound into 7.5mL ethanol-PEG 200-normal saline (5:20:75), vortexing for 2min, and carrying out ultrasonic treatment for 3min to prepare a test solution with the concentration of 0.2mg/mL for oral administration; taking 100 mu L of the test solution, diluting the solution to a constant volume of 10ng/mL by using methanol, preparing a reference substance with equal concentration, performing HPLC (high performance liquid chromatography) sample loading detection on the concentrations of the test solution and the reference solution, and calculating the accuracy of the test solution.
2.2 sample Collection
BALB/c mice were given 2mg/kg of test compound orally in a single dose of 10mL/kg, and the mice were bled and sacrificed at 1,4, and 6h after administration, and blood and total liver were collected and placed on ice.
2.3 liver sample processing and analysis
Weighing all livers, cutting into pieces, adding 4 times volume of methanol-water (50:50) homogenate, then homogenizing, centrifuging the homogenate (centrifugation condition: 8000rpm/min, 5min, 4 ℃), transferring 40. mu.L of supernatant, adding 400. mu.L of methanol-acetonitrile (50:50) precipitant (containing 5ng/mL of o-tolylhydramine), shaking for 3min, centrifuging (4500rpm, 5min, 4 ℃), taking supernatant, adding mobile phase for dilution, and analyzing the content of compounds in supernatant samples by LC-MS/MS.
2.4 plasma sample processing and analysis
The whole blood was anticoagulated with heparin sodium and centrifuged (centrifugation conditions: 8000rpm/min, 5min, 4 ℃), 40. mu.L of the supernatant plasma was transferred, 400. mu.L of methanol-acetonitrile (50:50) precipitant (containing 5ng/mL of o-tolylhydramine) was added, shaking was carried out for 3min, centrifugation (4500rpm, 5min, 4 ℃) was carried out, the supernatant was diluted with a mobile phase, the supernatant was taken, and the content of the compound in the supernatant sample was analyzed by LC-MS/MS, and the results are shown in Table 5.
3 results of the experiment
TABLE 5
The higher the concentration of the compound in the liver, the higher the potency for treating liver diseases, the better the therapeutic effect at the same dose, and the higher the liver/plasma ratio, indicating that the stronger the target organ selectivity of the test compound, the better the safety of the compound may be. From the results, the compounds of the invention have higher distribution in the liver, and the liver selectivity and targeting are good. Therefore, the compound is expected to become a more effective and safer medicament for treating metabolic liver diseases such as fatty liver, non-alcoholic fatty liver disease (NASH) and the like.
Experimental example 3 evaluation of efficacy of HFD-CCL 4-induced NASH and hepatic fibrosis
The method comprises the steps of firstly inducing animal liver steatosis by High Fat Diet (HFD), and then inducing liver inflammation, necrosis and fibrosis by carbon tetrachloride (CCL4), wherein the model is similar to the occurrence process and pathological phenomena of human NASH diseases. The purpose of this experiment was to evaluate the efficacy of the compounds of the invention in the NASH model of HFD-CCL 4-induced C57BL/6 mice, with GS-4997 as a control compound. HFD-CCL4 was induced for 10 weeks, drug intervention was performed for 4 weeks, and the effect of the drug on NASH and hepatic fibrosis was observed.
1. Experimental Material
1.1 instruments
A dehydrator Leica HistoCore PEARL; a paraffin embedding machine Leica HistoCore Arcadia C & H; paraffin slicer Leica RM 2235; automatic stain Leica ST 5020; scanner HAMAMATSU NANO Zoomer S210; SR staining analysis software Visiopharm VIS 6.6.0.2516.
1.2 control Compounds
As a positive control, a compound represented by the following formula (a) (i.e., GS-4997) disclosed in WO2013/112741(PCT/US2013/022997) was used,
the compound was prepared and identified by hydrogen spectroscopy and mass spectrometry with reference to the method described in WO 2013/112741.
1.3 animals
C57BL/6 mice (male, 18-20g) were purchased from Wei Tony Hua, Inc., Beijing. Experimental animal feeding all experimental procedures were approved by the KCI animal use and welfare committee (IACUC). The mouse feeding conditions were as follows: the temperature is 20-25 ℃, the humidity is 40-70%, and the light and shade alternation time is 12 hours/12 hours. Bedding was changed 2 times per week.
2. Experimental methods
2.1 preparation of Compounds
The test compound and GS-4997 of the present invention were diluted to 0.3mg/mL, 1mg/mL, 3mg/mL with 0.045M hydrochloric acid-propylene glycol (50:50) solution for use in the present formulation.
2.2 animal Molding
HFD-CCL4 induced the C57BL/6 mouse NASH model: after the animals are adaptively raised in a central SPF barrier of a KCI experimental animal for 3-7 days, the animals are replaced with HFD feed for raising, and the raising period is 10 weeks. At the end of week 6 of HFD feeding, HFD groups were randomized into groups of 10 animals per group and were given CCl4 orally (three times a week, 9-10 am) for 4 weeks. Detailed modelling methods a HFD-CCl 4-induced male C57BL/6 mouse NASH model was established according to established methods for KCI, with the modelling agent being Olive Oil + CCl4 solution (formulated from KCI). The randomly grouped 6 animals were given normal maintenance diet with concomitant feeding as a normal control group.
Animals were divided into a normal control group, a model control group of HFD-CCL4 (model group, given 0.045M hydrochloric acid-propylene glycol (50:50)), and a compound group (test compound group of the present invention, GS-4997 group).
2.3 dosing regimens for Compounds
After the end of week 6 of HFD feeding, gavage of test compound of the invention and GS-4997 was started, once daily for 4 weeks, and the dosing was ended at week 10. Wherein the dose of the test compound group of the present invention is 3mg/kg/d, and the dose of the GS-4997 group is 30mg/kg/d, i.e., the test compound group of the present invention is administered at a dose one tenth of the dose of the GS-4997 group.
2.4 Experimental sample Collection
The next day after the end of the last dose, i.e., 48 hours after the last dose of CCl4, animals in each group were fasted for six hours and were euthanized according to KCI standard operating procedures. Dissecting animals according to KCI animal dissection experiment operating procedures, collecting livers after the animals are perfused into the whole body by low-temperature PBS, and rapidly freezing part of animal livers (the same liver lobe on the left side of each animal is fixedly selected) by liquid nitrogen and storing at the low temperature of-80 ℃. And fixing the rest animal liver with formalin (the volume ratio of the liver to the fixing solution is 1:10), and performing related pathological correlation detection.
2.5 hematoxylin-eosin staining
Liver left leaf was fixed with 10% formalin, embedded with paraffin, and prepared into 5 μm sections for hematoxylin-eosin (H & E) staining. Hematoxylin-eosin staining reflects the degree of tissue inflammation, fat deposition, vacuolar degeneration and tissue fibrosis, and the semi-quantitative analysis criteria for the degree of lesions are shown in table 6.
TABLE 6 pathological scoring criteria for hepatic fibrosis
2.6 Tianlang scarlet staining
The liver tissue was sliced to 5 μm, dried for 2h, rehydrated, stained with Tianlang scarlet (Beijing Haidechun, cat # 26357) for 30min at room temperature, and then dehydrated and mounted for image analysis. The pathological sections were scanned 200 × fold with Aperio ScanScope CS2 (lycra), and the scanned pictures were opened in the Aperio ImageScope program to remove blood vessels, leaving the target image for analysis with Color resolution v9 algorithm. The portion of fibrosis stained red was identified as a positive signal using the software and the percentage of fibrosis calculated.
3. Statistical analysis
Data are expressed as mean ± sem. The significance analysis used students t-test, one wayANOVA or twowayANOVA and post-hoc Dunnett's test.
4. Results of the experiment
4.1 hepatocyte necrosis
Compared with a normal control group, the degree of the hepatocyte necrosis of the model group is obviously increased; after treatment with each compound group, the degree of hepatocyte necrosis was significantly reduced compared to the model group. The reduction of hepatocyte necrosis was significantly higher or similar to that of GS-4997 group (30mg/kg/d) in the compound groups of example 5 and example 29 (3 mg/kg/d). The results are shown in Table 7.
TABLE 7 hepatocyte necrosis
**P<0.01vs. normal control group;#P<0.05vs. model set;##P<0.01vs. model set;###P<0.001vs. model set
4.2 hepatic fibrosis score
After CCL4 and high-fat diet induction, compared with a normal control group, the liver fibrosis area of the model group is obviously increased; the compound groups (3mg/kg/d) of example 5 and example 29 significantly reduced the area of liver fibrosis compared to the model group, and the degree of improvement on fibrosis was significantly better than that of the GS-4997 group (30 mg/kg/d). The results are shown in Table 8.
TABLE 8 hepatic fibrosis score
**P<0.01vs. normal control group;#P<0.05vs. model set;##P<0.01vs. model set;###P<0.001vs. model set
Therefore, the compound has a certain therapeutic effect on a mouse NASH model induced by HFD-CCL 4; compared with a model group, the liver fibrosis model can effectively reduce liver steatosis and liver cell injury and alleviate the liver fibrosis degree in histopathology.
Although the present invention has been described in detail above, those skilled in the art will appreciate that various modifications and changes can be made to the present invention without departing from the spirit and scope of the invention. The scope of the invention is not to be limited by the above detailed description but is only limited by the claims.

Claims (10)

1. A compound shown in a general formula I or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof,
wherein:
ring A is selected from C3-12Cycloalkyl radical, C6-18Aryl and 5-20 membered heteroaryl, wherein said cycloalkyl, aryl and heteroaryl are optionally substituted with one or more halogen, hydroxy, halogen,C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
ring B is selected from the group consisting of 10-18 membered bicyclic aryl, 9-or 10-membered bicyclic heteroaryl, 9-or 10-membered bicyclic heterocyclyl, wherein said bicyclic aryl, bicyclic heteroaryl and bicyclic heterocyclyl are optionally substituted with one or more halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo;
ring C is a 5-12 membered monocyclic heteroaryl optionally substituted with one or more halogens, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl or 5-12 membered heteroaryl;
Z1is acyl, imino C1-6Alkyl acyl radicalOr is absent;
Z2is-CONH-, -NHCO-or is absent; and
R1selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl and 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl;
provided that Z is1When absent, ring a is heteroaryl and is optionally substituted with one or more halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, or oxo groups.
2. The compound of claim 1, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein formula I has the structure of formula Ia below,
wherein,
y is selected from alkylene, alkenylene and cycloalkylene optionally substituted with one or more alkyl, haloalkyl, hydroxy, hydroxyalkyl, halogen, oxo, alkoxy, carboxy, cyano, amino, monoalkylamino or dialkylamino groups, or when Y is alkylene and substituted with two alkyl groups, the two alkyl groups may together with the C atom to which they are attached form a cycloalkyl group;
w is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
m is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; and
R1as defined according to formula I in claim 1 or 2.
3. The compound of claim 2, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
structure of the productIs selected from
4. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, wherein the general formula I has the structure of the following general formula Ib,
wherein:
w is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
ring B, ring C, Z1、Z2、R1As defined according to formula I in claim 1 or 2.
5. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, wherein the formula I has the structure of the following formula Ic,
wherein:
w is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
y is selected from alkylene, alkenylene and cycloalkylene, wherein the alkylene, alkenylene and cycloalkylene are optionally substituted with one or more alkyl, haloalkyl, hydroxy, hydroxyalkyl, halogen, oxo, alkoxy, carboxy, cyano, amino, monoalkylamino or dialkylamino groups, or when Y is alkylene and is substituted with two alkyl groups, the two alkyl groups may together with the C atom to which they are attached form a cycloalkyl group;
X1、X2are each independently selected from C (R)2) And N, wherein R2Selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkylacyl, aminoacyl, alkaneAlkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl; and
ring C and R1As defined according to formula I in claim 1.
6. The compound according to claim 2 or 5, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
y is selected from sub-C1-6Alkyl radical, C2-10Alkenylene and C3-10Cycloalkyl, wherein said alkylene is1-6Alkyl radical, C2-10Alkenylene and C3-10Cycloalkylene radicals may be substituted by one or more C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, halogen, oxo, C1-6Alkoxy, carboxyl, cyano, amino, mono C1-6Alkylamino or di-C1-6Alkylamino, or when Y is alkylene and is substituted by two alkyl groups, said two alkyl groups may form C together with the C atom to which they are attached3-8A cycloalkyl group; further preferably, Y is selected from the group consisting of sub-C1-3Alkyl radical, C2-6Alkenylene and C3-6Cycloalkylene, wherein said alkylene is C1-3Alkyl radical, C2-6Alkenylene and C3-6Cycloalkylene radicals may be substituted by one or more C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl, halogen, oxo, C1-3Alkoxy, carboxyl, cyano, amino, mono C1-3Alkylamino or di-C1-3Alkylamino, or when Y is alkylene and is substituted by two alkyl groups, said two alkyl groups may form C together with the C atom to which they are attached3-6A cycloalkyl group; even more preferably, Y is selected from the group consisting of methylene, ethylene, propylene, vinylene, propenylene and cyclopropylene, wherein said methylene, ethylene, propylene, ethenylene, propenylene and cyclopropylene are optionally substituted with one or more halogen, methyl, ethyl, propyl, isopropyl, oxo groups, or when Y is alkylene and is substituted with two alkyl groups, said two groups areAn alkyl group may form C together with the C atom to which they are attached3-C6A cycloalkyl group;
w is selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl; further preferably, W is selected from hydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl; even more preferably, W is selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-8 membered aryl or 5-8 membered heteroaryl; and
R1selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, haloC1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-12Cycloalkyl and 3-12 membered heterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl; further preferably, R1Selected from hydrogen, halogen, hydroxy, C1-3Alkyl, halo C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl; even more preferably, R1Selected from hydrogen, halogen, hydroxy, methyl, ethyl, propyl, isopropyl, halogenated C1-3Alkyl, hydroxy C1-3Alkyl radical, C1-3Alkoxy, halo C1-3Alkoxy, hydroxy C1-3Alkoxy, nitro, carboxyl, cyano, amino, mono C1-3Alkylamino radical, C1-3Alkylacylamino group, C1-3Alkyl acyl, amino acyl, C1-3Alkylaminoacyl, di-C1-3Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl; and
m is selected from hydrogen, halogen, hydroxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, nitro, carboxyl, cyano, amino, mono C1-6Alkylamino radical, C1-6Alkylacylamino group, C1-6Alkyl acyl, amino acyl, C1-6Alkylaminoacyl, di-C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl, 3-12 membered heterocyclyl, 6-12 membered aryl, 5-12 membered heteroaryl, or oxo.
7. A compound of formula Id or its isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug,
wherein,
ring B is selected from monocyclic aryl optionally substituted with one or more groups selected from halo, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkanoylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamido, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo;
R2one or more groups selected from the group consisting of halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, hydroxyhaloalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylamino, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, and oxo;
w is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, nitro, carboxy, cyano, amino, monoalkylamino, alkylacylamino, alkanoyl, aminoacyl, alkylaminoacyl, dialkylaminoacyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl; and
Z2is-CONH-, -NHCO-or is absent.
8. A compound or isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the compound is a compound selected from the group consisting of:
9. a pharmaceutical composition comprising a compound of any one of claims 1 to 8, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
10. Use of a compound of any one of claims 1 to 8, or an isomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition of claim 9 for the manufacture of a medicament for the treatment and/or prevention of an apoptosis signal-regulating kinase 1-associated disease.
CN201810780971.0A 2017-07-18 2018-07-17 Heterocyclic compounds as ASK inhibitors and uses thereof Active CN109265443B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201710588014 2017-07-18
CN2017105880143 2017-07-18
CN201710799138 2017-09-06
CN2017107991386 2017-09-06

Publications (2)

Publication Number Publication Date
CN109265443A true CN109265443A (en) 2019-01-25
CN109265443B CN109265443B (en) 2022-11-29

Family

ID=65014972

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810780971.0A Active CN109265443B (en) 2017-07-18 2018-07-17 Heterocyclic compounds as ASK inhibitors and uses thereof

Country Status (3)

Country Link
CN (1) CN109265443B (en)
TW (1) TW201908306A (en)
WO (1) WO2019015559A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109071448A (en) * 2017-01-22 2018-12-21 福建广生堂药业股份有限公司 Ask1 inhibitor and its preparation method and application
CN110294742A (en) * 2018-03-21 2019-10-01 山东轩竹医药科技有限公司 And ring class ASK1 inhibitor and its application
CN111423422A (en) * 2019-01-09 2020-07-17 苏州泽璟生物制药股份有限公司 Aryl amide inhibitor and preparation method and application thereof
CN112312905A (en) * 2018-05-02 2021-02-02 英安塔制药有限公司 Tetrazole-containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202012414A (en) 2018-04-12 2020-04-01 美商拓臻股份有限公司 Tricyclic ask1 inhibitors
KR102288434B1 (en) * 2018-10-18 2021-08-12 에이치케이이노엔 주식회사 Novel (isopropyl-triazolyl)pyridinyl-substituted benzooxazinone or benzothiazinone derivatives and use thereof
US11142525B2 (en) 2018-11-15 2021-10-12 Pfizer Inc. Azalactam compounds as HPK1 inhibitors
JP7303948B2 (en) * 2020-05-01 2023-07-05 ファイザー・インク Azalactam compounds as HPK1 inhibitors
WO2021224818A1 (en) * 2020-05-08 2021-11-11 Pfizer Inc. Isoindolone compounds as hpk1 inhibitors
UY39309A (en) 2020-07-02 2021-12-31 Pi Industries Ltd ISOXAZOLINE COMPOUNDS AND THEIR USE AS PEST CONTROL AGENTS
WO2023030478A1 (en) * 2021-09-03 2023-03-09 浙江海正药业股份有限公司 Pyridolactam derivative, and preparation method therefor and use thereof
AR126996A1 (en) 2021-09-08 2023-12-06 Pi Industries Ltd ISOXAZOLINE COMPOUNDS AND THEIR USE AS PEST CONTROL AGENTS
WO2023057883A1 (en) * 2021-10-05 2023-04-13 Pfizer Inc. Crystalline form of azalactam compound

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102482257A (en) * 2009-07-13 2012-05-30 吉利德科学股份有限公司 Apoptosis signal-regulating kinase inhibitors
CN104080771A (en) * 2012-01-27 2014-10-01 吉利德科学公司 Apoptosis signal-regulating kinase inhibitor
WO2018133865A1 (en) * 2017-01-22 2018-07-26 福建广生堂药业股份有限公司 Ask1 inhibitor and preparation method and use thereof
WO2018149284A1 (en) * 2017-02-16 2018-08-23 四川科伦博泰生物医药股份有限公司 Kinase inhibitor and preparation method therefor and use thereof
WO2018157857A1 (en) * 2017-03-03 2018-09-07 江苏豪森药业集团有限公司 Apoptotic signal regulation kinase inhibitor, preparation method and application thereof
CN110325526A (en) * 2017-02-28 2019-10-11 伊莱利利公司 Isoquinolin and naphthyridine compounds

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9586933B2 (en) * 2014-12-23 2017-03-07 Gilead Sciences, Inc. Processes for preparing ASK1 inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102482257A (en) * 2009-07-13 2012-05-30 吉利德科学股份有限公司 Apoptosis signal-regulating kinase inhibitors
CN104080771A (en) * 2012-01-27 2014-10-01 吉利德科学公司 Apoptosis signal-regulating kinase inhibitor
WO2018133865A1 (en) * 2017-01-22 2018-07-26 福建广生堂药业股份有限公司 Ask1 inhibitor and preparation method and use thereof
WO2018149284A1 (en) * 2017-02-16 2018-08-23 四川科伦博泰生物医药股份有限公司 Kinase inhibitor and preparation method therefor and use thereof
CN110325526A (en) * 2017-02-28 2019-10-11 伊莱利利公司 Isoquinolin and naphthyridine compounds
WO2018157857A1 (en) * 2017-03-03 2018-09-07 江苏豪森药业集团有限公司 Apoptotic signal regulation kinase inhibitor, preparation method and application thereof

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109071448A (en) * 2017-01-22 2018-12-21 福建广生堂药业股份有限公司 Ask1 inhibitor and its preparation method and application
CN109071448B (en) * 2017-01-22 2020-03-17 福建广生堂药业股份有限公司 ASK1 inhibitor and preparation method and application thereof
US10787435B2 (en) 2017-01-22 2020-09-29 Fuijan Cosunter Pharmaceutical Co. Ltd. ASK1 inhibitor and preparation method and use thereof
CN110294742A (en) * 2018-03-21 2019-10-01 山东轩竹医药科技有限公司 And ring class ASK1 inhibitor and its application
CN110294742B (en) * 2018-03-21 2023-01-31 山东轩竹医药科技有限公司 Fused ring ASK1 inhibitor and application thereof
CN112312905A (en) * 2018-05-02 2021-02-02 英安塔制药有限公司 Tetrazole-containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
CN112312905B (en) * 2018-05-02 2024-03-15 英安塔制药有限公司 Tetrazole-containing inhibitors of apoptosis signal-regulating kinase 1 and methods of use thereof
CN111423422A (en) * 2019-01-09 2020-07-17 苏州泽璟生物制药股份有限公司 Aryl amide inhibitor and preparation method and application thereof

Also Published As

Publication number Publication date
WO2019015559A1 (en) 2019-01-24
TW201908306A (en) 2019-03-01
CN109265443B (en) 2022-11-29

Similar Documents

Publication Publication Date Title
CN109265443B (en) Heterocyclic compounds as ASK inhibitors and uses thereof
CN109456308B (en) Heterocyclic compounds as ASK inhibitors and their use
CN110016025B (en) Immunomodulator
JP6921114B2 (en) Degradation and use of CDK8 by conjugation of cyclin-dependent kinase 8 (CDK8) inhibitor with E3 ligase ligand
JP6622824B2 (en) Kynurenin-3-monooxygenase inhibitor, pharmaceutical composition thereof, and methods of use thereof
CN113544128A (en) KRAS-G12C inhibitors
JP2019514882A (en) Degradation and use of CDK9 by conjugation of cyclin dependent kinase 9 (CDK 9) inhibitors to E3 ligase ligands
JP2019514883A (en) Degradation and use of CDK4 / 6 by conjugation of cyclin dependent kinase 4/6 (CDK4 / 6) inhibitors with E3 ligase ligands
CN111499617B (en) Immunomodulator
CA2911856A1 (en) Tri-substituted amino hydroxamate compounds and pharmaceutical compositions thereof for use as histone deacetylase inhibitors
CN110036005A (en) Amide derivatives and its application in drug
CN110128415A (en) Indoline-like compound as immunomodulator and preparation method thereof
CN112920183A (en) Compounds as KRAS-G12C inhibitors and uses thereof
CN110573501A (en) Rho-related protein kinase inhibitor, pharmaceutical composition containing same, and preparation method and application thereof
CN110582491A (en) Rho-related protein kinase inhibitor, pharmaceutical composition containing same, and preparation method and application thereof
CN113754653A (en) KRAS G12C inhibitor compound and application thereof
CN107151250B (en) Pyrimidine seven-membered ring compound, preparation method thereof, medicinal composition and application thereof
CN109310679A (en) Combination comprising histone deacetylase inhibitor
WO2023116835A1 (en) Multi-protein degradation agent having imide skeleton
TW201348213A (en) Quinazolinedione derivative
EP2814820A1 (en) Anti -malarial agents
CN112778302A (en) KRAS G12C inhibitor compound and application thereof
CN114667289A (en) Heteroaryl plasma kallikrein inhibitors
CN112513041B (en) Tricyclic compounds
CN111100130A (en) 4-amino pyrrolopyrimidine derivatives, and preparation method and application thereof

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