CN111808079A - Indole ASK1 small molecule inhibitor and preparation method and application thereof - Google Patents

Indole ASK1 small molecule inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN111808079A
CN111808079A CN202010777152.8A CN202010777152A CN111808079A CN 111808079 A CN111808079 A CN 111808079A CN 202010777152 A CN202010777152 A CN 202010777152A CN 111808079 A CN111808079 A CN 111808079A
Authority
CN
China
Prior art keywords
indole
isopropyl
triazol
carboxamide
pyridin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010777152.8A
Other languages
Chinese (zh)
Inventor
陈亚东
侯少华
佟宇
王雨晨
张艳敏
杨玥婧
万勃亨
陈泉威
魏然
陆涛
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical University
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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202010777152.8A priority Critical patent/CN111808079A/en
Publication of CN111808079A publication Critical patent/CN111808079A/en
Pending legal-status Critical Current

Links

Images

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Abstract

The invention discloses an indole ASK1 small molecule inhibitor and a preparation method and application thereof, wherein the inhibitor comprises a compound shown as a general formula (I) and a stereoisomer or pharmaceutically acceptable salt thereof. The experimental data prove that the inhibitor is used for non-alcoholic fatHas certain treatment effect on the sexual hepatitis and the ulcerative colitis and good application prospect.
Figure DDA0002618861600000011

Description

Indole ASK1 small molecule inhibitor and preparation method and application thereof
Technical Field
The invention mainly relates to a compound and a preparation method and application thereof, and particularly relates to an indole ASK1 small-molecule inhibitor and a preparation method and application thereof.
Background
Apoptosis signal-regulated kinase 1(ASK1) belongs to the family of cell mitogen-activated protein kinases (MAPK), and the MAPK signal pathway is one of the important signal transduction systems in organisms and participates in the physiological processes of cell proliferation, differentiation, apoptosis, inflammatory reaction and the like. The MAPK family includes extracellular signal-regulated kinase (ERK), P38MAPK and JNK three major subfamilies. The MAPK family contains tertiary kinases, the first of which is the activation of the cellular mitogen-activated protein kinase (MAP3K) by extracellular and intracellular stimuli, with activated MAP3K phosphorylation activating MAP2K, which in turn activates MAPK. ASK1 belongs to MAP3K5, at P38Upstream of the MAPK and JNK signaling pathways.
ASK1 is capable of being activated by a range of stress conditions, such as oxidative stress, endoplasmic reticulum stress, and calcium influx. Activated ASK1 activates MKK3/6 and MKK4/7 through phosphorylation to activate P38MAPK and JNK pathways. ASK1 plays an important role in regulating cell physiological processes such as apoptosis, cytokine response, cell differentiation, and innate immune response. The research shows that ASK1 has obvious effect on a plurality of diseasesAnd (4) correlation.
Relevant literature reports show that ASK1 small molecule inhibitors show therapeutic potential for various diseases. For example, GS-444217 has obvious effect in various animal experimental models of renal fibrosis and inflammation, and K811 and K812 have obvious effect in SOD1 of ALS (amyotrophic lateral sclerosis)G93AThe survival period of the mouse is prolonged in the transgenic mouse model, K811 inhibits the growth of gastric cancer cells in vitro and in vivo tests, and GS-4997 carries out clinical tests on pulmonary hypertension, diabetic nephropathy and the like. However, the structure type of the ASK1 small-molecule inhibitor is single at present, and further research is needed.
Disclosure of Invention
The purpose of the invention is as follows: the invention aims to provide an indole ASK1 small-molecule inhibitor which influences apoptosis signal regulation kinase 1 and thus shows therapeutic potential for various diseases. The invention also aims to disclose a preparation method of the inhibitor and application of the inhibitor in medicines for treating nonalcoholic steatohepatitis and ulcerative colitis.
The technical scheme is as follows: the indole ASK1 small molecule inhibitor comprises a compound shown as a general formula (I) and a stereoisomer or pharmaceutically acceptable salt thereof:
Figure BDA0002618861580000011
M1is selected from N or CH;
R1is the same or different and is independently selected from hydrogen atom, alkoxy, halogen, heterocyclic radical, aryl, heteroaryl, - (CH)2)nOR4、-(CH2)nNR5R6、-(CH2)nNR5C(O)R4N is 0, 1,2,3, 4 or 5; wherein the heteroaryl is unsubstituted or substituted by one or more substituents selected from alkyl, alkoxy, and heterocyclyl;
R2the same or different, and each is independently selected from hydrogen atom, alkyl, halogen, aryl, heteroaryl;
R3selected from alkyl, hydroxyalkyl or heterocyclyl;
R4selected from alkyl groups;
R5and R6The same or different, and each is independently selected from hydrogen atom, heterocyclic radical;
x is selected from N (R)2);
m is 0, 1,2,3 or 4;
p is 1 or 2.
The indole ASK1 small molecule inhibitor, M1Is N:
Figure BDA0002618861580000021
the indole ASK1 micromolecule inhibitor has the structure that X is imino and R is3Is isopropyl, and is specifically shown as a general formula (III):
Figure BDA0002618861580000022
the indole ASK1 small molecule inhibitor R1Selected from hydrogen atoms, C1-6Alkoxy, halogen, 6-membered heterocyclyl, 6-membered aryl, 5-6-membered heteroaryl, substituted 5-6-membered heteroaryl and- (CH)2)nNR5C(O)R4Preferably a hydrogen atom, C1-3Alkoxy, halogen, 6-membered heterocyclyl, 6-membered aryl, 5-6-membered heteroaryl, substituted 5-membered heteroaryl and- (CH)2)nNHC(O)R4More preferably a hydrogen atom, a methoxy group, a chlorine atom, a fluorine atom, a 3, 6-dihydro-2H-pyran-4-yl group, a 1,2,3, 6-tetrahydropyridin-4-yl group, a phenyl group, a pyrazolyl group, a thienyl group, a furyl group, a pyridyl group, a 1- (1-ethoxyethyl) -1H-pyrazol-4-yl group, a 1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl group, a 1-methyl-1H-pyrazol-4-yl group, an acetamido group; r2Selected from hydrogen atoms, C1-6Alkyl, 5-6 membered heteroaryl, halogen, preferably hydrogen atom, C1-3Alkyl, 5-membered heteroaryl and halogen, more preferably hydrogen atom, methyl, furyl; r3Is selected from C1-6Alkyl radical, C1-6Hydroxyalkyl, preferably C1-3Alkyl radical、C1-3Hydroxyalkyl, more preferably 1-hydroxyprop-2-yl, isopropyl; r4Is C1-6An alkyl group; r5Is a hydrogen atom; r6Is tetrahydro-2H-pyran-4-yl.
The indole ASK1 small molecule inhibitor is selected from the following compounds: n- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-1), 7-chloro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-2), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6-methoxy-1H-indole-2-carboxamide (I-3), 6-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-4), 5-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-5), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-2-carboxamide (I-6), (R) -N- (6- (4- (4- (1-hydroxypropan-2-yl) -4H-1,2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-7), N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-8), 6-fluoro-N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-9), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3-methyl-1H-indole-2-carboxamide (I-10), 3- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-11), 7-acetamido-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-12), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-13), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-14), 6- (1- (1-ethoxyethyl) -1H-pyrazol-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-15), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-16), N- (3- (5- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) -1H-indazol-3-yl) phenyl) -4- (trifluoromethyl) benzamide (I-17), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-18), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -4- (1-methyl-1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-19), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (thiophen-3-yl) -1H-indole-2-carboxamide (I-20), 7- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-21), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7-phenyl-1H-indole-2-carboxamide (I-22), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (pyridin-4-yl) -1H-indole-2-carboxamide (I-23), 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-24), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1,2,3, 6-tetrahydropyridin-4-yl) -1H-indole-2-carboxamide (I-25), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxamide (I-26).
The structural formula of the specific compound is shown in the following table:
Figure BDA0002618861580000041
Figure BDA0002618861580000051
the indole ASK1 small molecule inhibitor, the pharmaceutically acceptable salt is a salt formed by the inhibitor and acid or base, and the acid is hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenesulfonic acid, citric acid, tartaric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, succinic acid, fumaric acid, salicylic acid, phenylacetic acid or mandelic acid; the base is an inorganic base containing a basic metal cation, an alkaline earth metal cation, or an ammonium cation salt.
The preparation method of the indole ASK1 small molecule inhibitor comprises the following steps:
(1) methyl 3-aminobenzoate or methyl 6-aminopyridine-2-carboxylate A is firstly converted into a hydrazide compound B;
(2) a compound C is obtained through ring closing reaction, and the compound C reacts with a carboxylic acid compound D to obtain a compound (I);
Figure BDA0002618861580000052
and (3) adding a corresponding acid or alkali solution into the solution of the compound (I) prepared by the method, and removing the solvent under reduced pressure after salt formation is completed to obtain the pharmaceutically acceptable salt of the ASK1 inhibitor.
A pharmaceutical composition comprising the indole ASK1 small molecule inhibitor of claim 1 and one or more pharmaceutically acceptable carriers, diluents, or excipients.
The indole ASK1 small molecule inhibitor or the pharmaceutical composition is applied to preparation of ASK1 inhibitor drugs.
The indole ASK1 small molecule inhibitor or the pharmaceutical composition is applied to the preparation of drugs for treating inflammatory diseases and ASK1 related diseases; the inflammatory diseases are non-alcoholic steatohepatitis and ulcerative colitis.
Has the advantages that: compared with the prior art, the invention has the following remarkable characteristics: the indole ASK1 small molecule inhibitor shows good ASK1 kinase inhibition activity, is superior to AP1-HEK293 cell activity of GS-4996, the compound I-1 shows human liver microsome stability superior to GS-4997, and the compounds I-1 and I-4 show good in vivo pharmacokinetic properties. Experimental data prove that the compound I-4 has certain treatment effect on non-alcoholic steatohepatitis and ulcerative colitis.
Drawings
FIG. 1 is a graph showing the effect of compounds I-4 on colon length in mice with DSS-induced colitis;
FIG. 2 shows the effect of I-4 on colon histopathology in mice with DSS-induced colitis.
Detailed Description
The structure of the compound of the invention is hydrogen spectrum by nuclear magnetic resonance (1H-NMR) and Mass Spectrometry (MS). Purity of the compound is determined by High Performance Liquid Chromatography (HPLC)And (4) measuring.1The H-NMR was measured using Bruker Advance 300 and Bruker Advance400 NMR spectrometers in deuterated dimethyl sulfoxide (DMSO-d)6) And deuterated chloroform (CDCl)3) Internal standard is Tetramethylsilane (TMS).
Mass Spectrometry MS was performed using an Advion Mass expression CMS Mass spectrometer, and HPLC was performed using an Agilent1260Infinity liquid chromatography system.
Thin Layer Chromatography (TLC) adopts a thin layer chromatography silica gel plate (Nicotiana Yangtze friend silica gel development Co., Ltd.), a self-made silica gel thin layer plate adopts GF254 silica gel (Qingdao ocean chemical plant), and silica gel column chromatography generally adopts 200-plus 300-mesh silica gel (Qingdao ocean chemical plant).
The starting materials in the examples of the present invention are known and commercially available, or may be synthesized using or according to methods known in the art.
All reactions of the present invention are carried out under continuous magnetic stirring, in dry nitrogen atmosphere, in dry solvent, at reaction temperature in degrees centigrade, unless otherwise specified.
Example 1
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-1)
Figure BDA0002618861580000071
First step preparation of 6-aminopyridine carbohydrazide
Figure BDA0002618861580000072
Methyl 6-aminopyridinecarboxylate (5.0g, 32.9mmol) was dissolved in MeOH and N was added2H4·H2O (3.22g, 65.8mmol), heating and refluxing the reaction liquid for 3 h; after cooling to room temperature, a solid precipitates out, the filter cake is washed with EA after suction filtration, and the target product (4.5g, 90%) is obtained after vacuum drying.
ESI-MS m/z:153.1[M+H]+.
Second step preparation of 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine
Figure BDA0002618861580000073
6-Aminopyridoformylhydrazine (4.5g, 29.61mmol) was dissolved with anhydrous Dioxane and triethyl orthoformate (5.92mL, 35.53mmol), N, was added2Reacting for 1h at 75 ℃ under protection. Addition of CH by syringe3COOH (5.08mL, 88.83mmol), cyclopropylamine (2.05mL, 29.61mmol), reacted at 110 ℃ for 4 h; TLC detection reaction is finished, reaction liquid is diluted by water, pH is adjusted to 10, extraction is carried out by EA, drying and spin-drying are carried out by EA, and silica gel column chromatography is carried out to obtain a target product (4.81g, 80%).
ESI-MS m/z:204.0[M+H]+.
Third step preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide
Figure BDA0002618861580000074
Dissolving 1H-indole-2-carboxylic acid (300mg, 1.86mmol) in 10mL DCM, adding 2 drops of DMF, adding oxalyl chloride (355mg, 2.79mmol) under ice bath condition, reacting for 2H after 10min by RT, and removing DCM to obtain acyl chloride for later use; dissolving 6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-amine (343mg, 1.68mmol) in DCM, adding DIEA (434mg, 3.36mmol), adding acyl chloride under ice bath condition, reacting for 4H after 10min by RT; TLC detected the reaction was complete, DCM removed and diluted with water, with DCM/MeOH 10: 1, extracting, drying organic phase, spin-drying, carrying out silica gel column chromatography, and purifying by a self-made silica gel large plate to obtain a target product (180mg, 31%).
1H NMR(300MHz,DMSO)11.89(s,1H),10.67(s,1H),8.90(s,1H),8.23(d,J=8.2Hz,1H),8.04(t,J=8.0Hz,1H),7.86(d,J=7.5Hz,1H),7.71(d,J=8.0Hz,1H),7.58(d,J=1.4Hz,1H),7.50(d,J=8.2Hz,1H),7.26(t,J=7.2Hz,1H),7.10(t,J=7.4Hz,1H),5.68-5.73(m,1H),1.48(d,J=6.7Hz,6H).
ESI-MS m/z:347.2[M+H]+.
Example 2
Preparation of 7-chloro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-2)
Figure BDA0002618861580000081
Preparation of 7-chloro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):12.06(s,1H),10.85(s,1H),8.90(s,1H),8.26(dd,J=6.3,0.3Hz,1H),8.06(t,J=8.7,1H),7.84(dd,J=7.6,0.6Hz,1H),7.68(d,J=6.0Hz,1H),7.55(d,J=2.1Hz,1H),7.37(dd,J=5.7,0.6Hz,1H),7.12(t,J=7.8Hz,1H),5.54-5.65(m,1H),1.48(d,J=5.1Hz,6H).
ESI-MS m/z:381.1.[M+H]+.
Example 3
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6-methoxy-1H-indole-2-carboxamide (I-3)
Figure BDA0002618861580000082
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6-methoxy-1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO)11.70(s,1H),10.56(s,1H),8.88(s,1H),8.23(d,J=8.3Hz,1H),8.02(t,J=7.9Hz,1H),7.84(d,J=7.5Hz,1H),7.63–7.47(m,2H),6.93(s,1H),6.75(d,J=8.7Hz,1H),5.66-6.45(m,1H),3.80(s,3H),1.47(d,J=6.5Hz,6H).
ESI-MS m/z:377.2[M+H]+.
Example 4
Preparation of 6-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-4)
Figure BDA0002618861580000091
Preparation of 6-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.95(s,1H),10.72(s,1H),8.88(s,1H),8.21(d,J=8.3Hz,1H),8.03(t,J=7.9Hz,1H),7.85(d,J=7.6Hz,1H),7.74(m,1H),7.59(s,1H),7.20(d,J=9.9Hz,1H),6.97(t,J=9.3Hz,1H),5.65-6.45(m,1H),1.47(d,J=6.6Hz,6H).
ESI-MS m/z:365.1[M+H]+.
Example 5
Preparation of 5-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-5)
Figure BDA0002618861580000092
Preparation of 5-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1.
ESI-MS m/z:365.1[M+H]+.
Example 6
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-2-carboxamide (I-6)
Figure BDA0002618861580000093
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-2-carboxamide reference example 1.
1H NMR(400MHz,DMSO-d6):10.80(s,1H),8.88(s,1H),8.18(d,J=8.2Hz,1H),8.03(t,J=7.9Hz,1H),7.88(d,J=7.5Hz,1H),7.73(d,J=7.9Hz,1H),7.61(d,J=8.4Hz,1H),7.42(s,1H),7.35(t,J=7.6Hz,1H),7.16(t,J=7.4Hz,1H),5.84–5.68(m,1H),4.04(s,3H),1.45(d,J=6.6Hz,6H).
ESI-MS m/z:361.2[M+H]+
Example 7
Preparation of (R) -N- (6- (4- (4- (1-hydroxypropan-2-yl) -4H-1,2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-7)
Figure BDA0002618861580000101
Preparation of (R) -N- (6- (4- (4- (1-hydroxypropan-2-yl) -4H-1,2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.89(s,1H),10.65(s,1H),8.80(s,1H),8.21(dd,J=8.3,0.6Hz,1H),8.03(t,J=8.0Hz,1H),7.84(dd,J=7.6,0.3Hz,1H),7.71(d,J=7.9Hz,1H),7.55(d,J=1.5Hz,1H),7.49(d,J=8.1Hz,1H),7.26(m,1H),7.09(m,1H),5.61(m,1H),4.99(t,J=5.4Hz,1H),3.66(m,2H),1.48(d,J=6.9Hz,3H).
ESI-MS m/z:363.1[M+H]+.
Example 8
Preparation of N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-8)
Figure BDA0002618861580000102
Preparation of N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.81(s,1H),10.43(s,1H),8.89(s,1H),8.14(s,1H),7.98(d,J=8.2Hz,1H),7.70(d,J=8.0Hz,1H),7.57(t,J=7.9Hz,1H),7.48(d,J=7.7Hz,2H),7.35(d,J=7.7Hz,1H),7.24(t,J=7.6Hz,1H),7.08(t,J=7.5Hz,1H),4.43-4.58(m,1H),1.46(d,J=6.7Hz,6H).
ESI-MS m/z:346.2[M+H]+.
Example 9
Preparation of 6-fluoro-N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-9)
Figure BDA0002618861580000103
Preparation of 6-fluoro-N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.90(s,1H),10.45(s,1H),8.89(s,1H),8.13(t,J=1.6Hz,1H),7.99–7.93(m,1H),7.74(dd,J=8.8,5.5Hz,1H),7.57(t,J=7.9Hz,1H),7.49(d,J=1.5Hz,1H),7.35(d,J=7.7Hz,1H),7.19(dd,J=9.9,2.2Hz,1H),7.01–6.92(m,1H),4.57–4.43(m,1H),1.46(d,J=6.7Hz,6H).
ESI-MS m/z:364.1[M+H]+
Example 10
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3-methyl-1H-indole-2-carboxamide (I-10)
Figure BDA0002618861580000111
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3-methyl-1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.64(s,1H),10.22(s,1H),8.89(s,1H),8.25(dd,J=8.4,2.4Hz,1H),8.03(m,1H),7.84(dd,J=7.5,2.4Hz,1H),7.68(d,J=8.1Hz,1H),7.45(d,J=8.4Hz,1H),7.30(m,1H),7.07(t,J=7.8Hz,1H),5.55-5.74(m,1H),2.55(s,3H),1.47(d,J=6.7Hz,6H)
ESI-MS m/z:361.2[M+H]+.
Example 11
Preparation of 3- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-11)
Figure BDA0002618861580000112
First step preparation of 3-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide
Figure BDA0002618861580000113
N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (200mg, 0.58mmol) was dissolved in THF and reacted with NBS at RT for 3H; TLC detection reaction was completed, THF was removed by rotation, diluted with water, extracted with DCM, dried and dried, and column-chromatographed on silica gel to give the desired product (199mg, 81%).
ESI-MS m/z:425.1[M+H]+.
Second step preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3-methyl-1H-indole-2-carboxamide
Figure BDA0002618861580000121
3-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (100mg, 0.24mmol), Pd (dppf) Cl2(18mg,0.024mmol)、K2CO3(66mg,0.48mmol) and furan-3-boronic acid pinacol ester (70mg,0.36mmol) with dioxane/H2Dissolving O, and reacting for 6 hours at 100 ℃; TLC detection reaction is finished, the reaction solution is cooled to room temperature, diluted by water and extracted by EA, organic phase is dried and dried, and silica gel column chromatography is carried out to obtain a target product (69mg, 69%).
1H NMR(400MHz,DMSO)12.10(s,1H),9.56(s,1H),8.88(s,1H),8.31(d,J=8.0Hz,1H),8.18(s,1H),8.04(t,J=8.0Hz,1H),7.94(t,J=1.6Hz,1H),7.90(d,J=7.5Hz,1H),7.58(d,J=8.1Hz,1H),7.53(d,J=8.3Hz,1H),7.32(t,J=7.3Hz,1H),7.14(t,J=7.5Hz,1H),6.86(d,J=0.9Hz,1H),5.32-5.44(m,1H),1.43(d,J=6.7Hz,6H).
ESI-MS m/z:413.2[M+H]+.
Example 12
Preparation of 7-acetamido-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-12)
Figure BDA0002618861580000122
First step preparation of ethyl (E) -2- (2- (2- (2-nitrophenyl) hydrazino) propionate
Figure BDA0002618861580000123
Dissolving o-nitrophenylhydrazine hydrochloride (1g, 6.53mmol) with EtOH, adding ethyl pyruvate (731mg,6.3mmol), and reacting for 12h by RT; the reaction was concentrated to give the product as a yellow solid (1.2g, 73%).
ESI-MS m/z:252.1[M+H]+.
Second step preparation of 7-nitro-1H-indole-2-carboxylic acid ethyl ester
Figure BDA0002618861580000131
Dissolving (E) -ethyl 2- (2- (2- (2-nitrophenyl) hydrazino) propionate (1.2g,4.78mmol) in polyphosphoric acid, reacting at 80 ℃ for 3h, detecting by TLC that the reaction is finished, cooling to RT, diluting the reaction solution with water, adjusting the pH to 8 with saturated NaOH, extracting the water phase with EA, washing the EA with water and saturated common salt, drying and spin-drying the EA, and carrying out silica gel column chromatography to obtain a target product (800mg, 71%).
ESI-MS m/z:235.1[M+H]+.
Third step preparation of 7-amino-1H-indole-2-carboxylic acid ethyl ester
Figure BDA0002618861580000132
Taking ethyl 7-nitro-1H-indole-2-carboxylate (800mg, 3.42mmol) and using EtOH/H2Dissolving O (6:1), adding Fe (957mg,17.0mmol) and NH4Cl (1.10g, 20.0mmol), reaction at 80 ℃ for 4 h; TLC detection reaction is finished, the solution is filtered while the solution is hot, the filtrate is extracted by DCM, dried and dried by DCM, and separated by silica gel column chromatography to obtain the target product (400mg, 57%).
ESI-MS m/z:205.1[M+H]+.
Fourth step preparation of ethyl 7-acetylamino-1H-indole-2-carboxylate
Figure BDA0002618861580000133
Dissolving 7-amino-1H-indole-2-carboxylic acid ethyl ester (200mg,0.98mmol) in DCM, adding DIEA (253mg,1.96mmol), adding acetyl chloride (115mg,1.47mmol), and reacting at RT for 3H; TLC detection reaction is finished, reaction liquid is concentrated and then diluted by adding water, EA is extracted, the EA layer is washed by water and saturated salt solution, EA is dried and dried, and the target product (150mg, 62%) is obtained by silica gel column chromatography.
ESI-MS m/z:247.1[M+H]+.
Fifth step preparation of 7-acetamido-1H-indole-2-carboxylic acid
Figure BDA0002618861580000134
Taking ethyl 7-acetylamino-1H-indole-2-carboxylate (150mg,0.61mmol) and reacting with THF/H2Dissolving O5:1, adding NaOH (253mg,1.96mmol), and reacting for 3h at RT; TLC detection reaction is finished, the reaction solution is extracted by EA, the pH of the water phase is adjusted to 2 by 2M HCl and then extracted by EA, EA is dried and dried, and the target product (100mg, 75%) is obtained by silica gel column chromatography.
ESI-MS m/z:219.1[M+H]+.
Sixth step preparation of 7-acetylamino-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide
Figure BDA0002618861580000141
Preparation of 7-acetylamino-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1.
1H NMR(300MHz,DMSO-d6):11.82(s,1H),10.80(s,1H),9.94(s,1H),8.89(s,1H),8.22(d,J=8.2Hz,1H),8.05(t,J=7.9Hz,1H),7.96(d,J=7.5Hz,1H),7.86(d,J=7.4Hz,1H),7.62(s,1H),7.44(d,J=7.8Hz,1H),7.05(t,J=7.8Hz,1H),5.63–5.80(m,1H),2.18(s,3H),1.47(d,J=6.5Hz,6H).
ESI-MS m/z:404.2[M+H]+.
Example 13
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-13)
Figure BDA0002618861580000142
First step preparation of 7-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide
Figure BDA0002618861580000143
Preparation of 7-acetylamino-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 1 (third step).
ESI-MS m/z:425.1[M+H]+.
Second step preparation of 7-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide
Figure BDA0002618861580000151
Taking 7-bromo-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (100mg, 0.24mmol), Pd (dppf) Cl2(18mg,0.024mmol)、K3PO4(102mg,0.48mmol), 1-Boc-pyrazole-4-boronic acid pinacol ester (106mg,0.36mmol) with Dioxane/H2Dissolving O5:1, and reacting for 3h at 110 ℃; after the TLC detection reaction, the reaction solution was cooled to room temperature, diluted with water and extracted with EA, which was dried and then separated by silica gel column chromatography to obtain the desired product (50mg, 51%).
1H NMR(300MHz,DMSO-d6):13.14(s,1H),11.01(d,J=47.6Hz,2H),8.89(s,1H),8.45–7.97(m,4H),7.82(d,J=7.1Hz,1H),7.68–7.33(m,3H),7.15(s,1H),5.59(s,1H),1.47(d,J=5.2Hz,6H).
ESI-MS m/z:413.2[M+H]+.
Example 14
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-14)
Figure BDA0002618861580000152
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide reference was made to example 13.
1H NMR(300MHz,DMSO-d6):12.89(s,1H),12.06(s,1H),10.83(s,1H),8.89(s,1H),8.22(d,J=8.0Hz,1H),8.04(t,J=7.9Hz,3H),7.90(s,1H),7.85(d,J=7.3Hz,1H),7.50(q,J=8.3Hz,3H),5.81–5.66(m,1H),1.47(d,J=6.7Hz,6H).
ESI-MS m/z:413.2[M+H]+.
Example 15
Preparation of 6- (1- (1-ethoxyethyl) -1H-pyrazol-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-15)
Figure BDA0002618861580000161
Preparation of 6- (1- (1-ethoxyethyl) -1H-pyrazol-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) reference example 13.
1H NMR(400MHz,DMSO)12.01(s,1H),10.74(s,1H),8.90(s,1H),8.39(s,1H),8.23(d,J=8.0Hz,1H),8.04(t,J=8.0Hz,1H),7.94(s,1H),7.85(d,J=7.5Hz,1H),7.70(d,J=8.4Hz,1H),7.64(s,1H),7.54(s,1H),7.39(dd,J=8.3,1.4Hz,1H),5.73(dt,J=13.3,6.6Hz,1H),5.58(q,J=5.9Hz,1H),3.52–3.44(m,1H),3.29–3.23(m,1H),1.66(d,J=6.0Hz,3H),1.48(d,J=6.7Hz,6H),1.06(t,J=7.0Hz,3H).
ESI-MS m/z:485.2[M+H]+.
Example 16
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-16)
Figure BDA0002618861580000162
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(300MHz,DMSO-d6):12.92(s,1H),11.88(s,1H),10.66(s,1H),8.89(s,1H),8.23(d,J=8.2Hz,1H),8.04(t,J=8.0Hz,2H),7.85(d,J=7.6Hz,1H),7.68(d,J=8.3Hz,1H),7.62(s,1H),7.54(s,1H),7.38(d,J=8.3Hz,1H),5.62-5.73(m,1H),1.47(d,J=6.6Hz,6H).
ESI-MS m/z:413.2[M+H]+.
Example 17
Preparation of N- (3- (5- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) -1H-indazol-3-yl) phenyl) -4- (trifluoromethyl) benzamide (I-17)
Figure BDA0002618861580000163
Preparation of N- (3- (5- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) -1H-indazol-3-yl) phenyl) -4- (trifluoromethyl) benzamide reference was made to example 13.
1H NMR(300MHz,DMSO-d6):12.07(s,1H),10.74(s,1H),8.93(s,1H),8.89(s,1H),8.58(d,J=1.8Hz,1H),8.24(d,J=8.1Hz,1H),8.13(d,J=8.0Hz,1H),8.05(t,J=7.9Hz,1H),7.86(d,J=1.8Hz,1H),7.84(d,J=2.7Hz,1H),7.76(s,1H),7.63(d,J=1.8Hz,1H),7.54(m,1H),7.46(d,J=8.3Hz,1.2Hz,1H),5.77–5.66(m,1H),1.48(d,J=6.7Hz,6H).
ESI-MS m/z:424.2[M+H]+.
Example 18
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-18)
Figure BDA0002618861580000171
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(400MHz,DMSO)11.87(s,1H),10.64(s,1H),8.90(s,1H),8.27(s,1H),8.23(d,J=8.1Hz,1H),8.04(t,J=8.0Hz,1H),7.89(s,1H),7.85(d,J=7.6Hz,1H),7.69(d,J=8.4Hz,1H),7.60(s,1H),7.55(d,J=1.4Hz,1H),7.36(dd,J=8.4,1.3Hz,1H),5.65-5.75(m,1H),4.44(t,J=7.6Hz,1H),3.99(dd,J=8.3,2.9Hz,2H),3.49(td,J=11.6,5.6Hz,2H),2.02(dt,J=12.7,6.2Hz,4H),1.47(d,J=6.7Hz,6H).
ESI-MS m/z:497.2[M+H]+.
Example 19
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -4- (1-methyl-1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-19)
Figure BDA0002618861580000172
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -4- (1-methyl-1H-pyrazol-4-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(400MHz,DMSO-d6):12.00(s,1H),10.78(d,J=7.1Hz,1H),8.89(s,1H),8.27(s,1H),8.22(d,J=8.3Hz,1H),8.10–8.00(m,2H),7.96(s,1H),7.80(d,J=7.6Hz,1H),7.39–7.33(m,1H),7.29–7.20(m,2H),5.46-5.65(m,1H),3.95(s,3H),1.46(d,J=6.7Hz,6H).
ESI-MS m/z:427.2[M+H]+.
Example 20
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (thien-3-yl) -1H-indole-2-carboxamide (I-20)
Figure BDA0002618861580000181
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (thiophen-3-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(300MHz,DMSO-d6):11.13(s,1H),10.90(s,1H),8.89(s,1H),8.26(d,J=7.8Hz,1H),8.05(t,J=8.0Hz,1H),7.98(dd,J=2.8,1.3Hz,1H),7.82(d,J=7.0Hz,1H),7.76(dd,J=5.0,2.9Hz,1H),7.70(d,J=7.8Hz,1H),7.59–7.52(m,2H),7.41(dd,J=7.2,0.9Hz,1H),7.24–7.15(m,1H),5.65–5.49(m,1H),1.46(d,J=6.7Hz,6H).
ESI-MS m/z:429.1[M+H]+.
Example 21
Preparation of 7- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-21)
Figure BDA0002618861580000182
Preparation of 7- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(400MHz,DMSO)11.02(s,1H),10.90(s,1H),8.89(s,1H),8.41(s,1H),8.27(d,J=8.3Hz,1H),8.05(t,J=8.0Hz,1H),7.87(s,1H),7.82(d,J=7.6Hz,1H),7.68(d,J=7.9Hz,1H),7.53(d,J=1.5Hz,1H),7.43(d,J=7.1Hz,1H),7.18(t,J=7.6Hz,1H),7.04(s,1H),5.59(m,5.52-5.65,1H),1.46(d,J=6.7Hz,7H).
ESI-MS m/z:413.2[M+H]+.
Example 22
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7-phenyl-1H-indole-2-carboxamide (I-22)
Figure BDA0002618861580000191
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (thiophen-3-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(400MHz,DMSO)11.28(s,1H),10.87(s,1H),8.89(s,1H),8.25(d,J=8.3Hz,1H),8.04(t,J=8.0Hz,1H),7.83–7.78(m,1H),7.72(t,J=7.8Hz,3H),7.59–7.52(m,3H),7.47(t,J=7.4Hz,1H),7.29(dd,J=7.2,1.0Hz,1H),7.22(t,J=7.5Hz,1H),5.47-5.57(m,1H),1.46(d,J=6.7Hz,6H).
ESI-MS m/z:423.2[M+H]+.
Example 23
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (pyridin-4-yl) -1H-indole-2-carboxamide (I-23)
Figure BDA0002618861580000192
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (pyridin-4-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(300MHz,DMSO)11.59(s,1H),10.84(s,1H),8.90(s,1H),8.74(s,2H),8.25(d,J=8.3Hz,1H),8.05(t,J=8.0Hz,1H),7.83(dd,J=7.6,2.4Hz,2H),7.75(s,2H),7.60(d,J=1.8Hz,1H),7.39(d,J=6.3Hz,1H),7.32–7.20(m,1H),5.52-5.62(m,1H),1.45(t,J=8.1Hz,6H).
ESI-MS m/z:424.2[M+H]+.
Example 24
Preparation of 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-24)
Figure BDA0002618861580000193
Preparation of 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(400MHz,DMSO)11.31(s,1H),10.79(s,1H),8.89(s,1H),8.25(t,J=11.4Hz,1H),8.05(t,J=7.1Hz,1H),7.83(d,J=6.9Hz,1H),7.56(dt,J=26.5,8.9Hz,3H),7.22–7.01(m,1H),6.17(s,1H),5.81–5.47(m,1H),4.27(d,J=31.8Hz,2H),3.97(d,J=37.7Hz,2H),2.13(d,J=35.7Hz,2H),1.44(t,J=18.2Hz,6H).
ESI-MS m/z:429.2[M+H]+.
Example 25
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1,2,3, 6-tetrahydropyridin-4-yl) -1H-indole-2-carboxamide (I-25)
Figure BDA0002618861580000201
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1,2,3, 6-tetrahydropyridin-4-yl) -1H-indole-2-carboxamide reference example 13.
1H NMR(300MHz,DMSO)11.38(s,1H),10.86–10.63(m,1H),8.88(d,J=26.9Hz,1H),8.26(t,J=12.3Hz,1H),8.13–7.95(m,1H),7.86(d,J=7.5Hz,1H),7.63(t,J=9.9Hz,1H),7.50(d,J=19.8Hz,1H),7.22–7.04(m,3H),6.08(s,1H),5.68(dd,J=13.0,6.5Hz,1H),4.10(s,3H),3.66(s,3H),1.51(s,6H).
ESI-MS m/z:428.2[M+H]+.
Example 26
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxamide (I-26)
Figure BDA0002618861580000202
First step preparation of ethyl 6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxylate
Figure BDA0002618861580000203
Ethyl 6-amino-1H-indole-2-carboxylate (500mg, 2.45mmol) was dissolved in DCM, DIEA (632mL, 4.9mmol) and tetrahydropyranone (370mg, 3.70mmol) were added, stirring was carried out at RT for 2H, sodium borohydride acetate (2.6g, 12.3mmol) was added, and reaction was carried out at RT for 2H. TLC detection reaction is finished, DCM is removed, residue is diluted by water and extracted by EA, EA is dried and dried, and silica gel column chromatography is carried out to obtain a target product (442mg, 60%).
ESI-MS m/z:289.1[M+H]+.
Second step preparation of 6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxylic acid
Figure BDA0002618861580000211
Ethyl 6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxylate (442mg, 1.47mmol) was taken in MeOH/H2O5:1, NaOH (118mg,2.94mmol) was added and the reaction was carried out at RT for 4 h. And (3) after TLC detection reaction is finished, adding a small amount of water into the reaction liquid, extracting with EA, adjusting the pH of the water phase to 2, extracting the water phase with EA, drying and spin-drying the EA to obtain the target product (300mg, 78%).
ESI-MS m/z:261.1[M+H]+.
Third step preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxamide
Figure BDA0002618861580000212
Preparation of N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxamide reference is made to example 1.
1H NMR(400MHz,DMSO)11.85(s,1H),8.89(s,1H),8.29(d,J=8.1Hz,1H),8.07(q,J=8.2Hz,1H),7.91(dd,J=7.7,0.8Hz,1H),7.85–7.78(m,2H),7.65(t,J=8.9Hz,1H),7.51(t,J=7.9Hz,1H),5.55-5.65(m,J,1H),4.87(s,1H),4.09–3.96(m,2H),3.57(t,J=11.1Hz,2H),3.17(d,J=5.3Hz,1H),2.71(dd,J=15.4,7.8Hz,2H),1.69(d,J=9.8Hz,2H),1.48(t,J=7.0Hz,6H).
ESI-MS m/z:446.2[M+H]+.
Biological activity assay
1. ASK1 protein kinase Activity assay
The test uses a homogeneous time-resolved fluorescence (HTRF) assay to test compounds for inhibitory activity against ASK1 kinase, in contrast to the positive compound GS-4997.
(1) Diluted compounds were added to 384-well plates, and AssayBuffer diluted DMSO was added to positive and negative control wells, respectively.
(2) The compound and positive control wells were loaded with STK-Substrate 3-biotin/ATP and ASK1(MAP3K5), and the negative control wells were loaded with STK-Substrate 3-biotin/ATP and Assay Buffer.
(3) After incubation for 2.0h at room temperature, STK S3 Antibody-Eu was added.
(4) Incubating at room temperature for 1.0h, measuring the fluorescence signal value of each plate hole by an enzyme-labeling instrument, and calculating the inhibition rate and IC according to the fluorescence signal value50Values were calculated by prism (graphpad software) fitting curves.
The ASK1 protein kinase activity of some of the compounds of the examples of the invention is shown in Table 1
TABLE 1 ASK1 protein kinase Activity of some of the compounds of the examples of the invention
Compound numbering ASK1 IC50 Compound numbering ASK1 IC50
I-1 42nM I-14 31.49nM
GS-4997 14nM I-16 37.22nM
I-4 32.65nM I-19 18.46nM
I-13 38.86nM I-23 23.3nM
The compounds of the above examples have strong ASK1 kinase inhibitory activity. As a powerful ASK1 small-molecule inhibitor, the compounds have great potential clinical application value aiming at the treatment of ASK1 related diseases.
2. AP1-HEK293 cell inhibitory Activity assay
The method utilizes AP1 Reporter-HEK293 recombinant cells to evaluate the influence of compounds on a downstream signal path of ASK1 at a cellular level.
(1) HEK293-AP1 cells were seeded into 96-well plates and 100uL Thaw Medium 1 was added.
(2) Exposing the cells to CO2Incubate overnight in the incubator.
(3) The medium was slowly poured out of the well plate, Assay medium was diluted with Assay medium and added to the wells, Assay medium dilutions of the same concentration of DMSO were added to the wells as controls, and Assay medium dilutions of the same concentration of DMSO were added to the wells without cells to remove background fluorescence signals.
(4) Incubate for 1 h.
(5) Background fluorescence signals were removed by adding dilutions of PMA Assay medium to the wells, 0.1% DMSO Assay medium to the wells as a control, and 0.1% DMSO Assay medium to the wells without cells.
(6) Using ONE-Step according to the operating specificationTMLuciferase assay system for luciferase assay: adding ONE-StepTMLuciferase reagent was added to the wells, shaken at room temperature for 15min, and the fluorescent signal was measured using a luminometer.
(7) And (3) data analysis: background fluorescence values were subtracted. ComputingInhibition per compound and IC was fitted from inhibition50The value is obtained.
The cell activity of the compound AP1-HEK293 of some examples of the invention is shown in Table 2
TABLE 2 AP1-HEK293 cell Activity of some of the compounds of the examples of the invention
Figure BDA0002618861580000221
Figure BDA0002618861580000231
The compound of some examples shows cell activity superior to that of the positive medicine GS-4997, and the compound has excellent physical and chemical properties and drug forming property.
3. Human liver microsome stability
The method preliminarily evaluates the in vitro metabolic stability of the compound through a human liver microsome stability experiment
The experimental steps are as follows:
(1) buffer A1.0L 0.1M Potassium dihydrogen phosphate buffer containing 1.0mM EDTA
Buffer B1.0L 0.1M dipotassium phosphate buffer containing 1.0mM EDTA
Buffer C0.1M potassium phosphate buffer, 1.0mM EDTA, pH7.4, 700mL of buffer B was titrated with buffer A while monitoring with a pH meter.
(2) Reference compound (Ketanserin) and test compound spiking solutions:
500 μ M spiking solution: mu.L of 10mM DMSO stock solution was added to 190. mu.L ACN.
Add 1.5. mu.M spiking solution (0.75mg/mL) to microsomes: mu.L of 500. mu.M spiking solution and 18.75. mu.L of 20mg/mL liver microsomes were added to 479.75. mu.L of buffer C on ice.
(3) An NADPH stock solution (6mM) was prepared by dissolving NADPH in buffer C.
(4) 30 μ L of 1.5 μ Mspiking solution containing 0.75mg/mL microsome solution was dispensed onto assay plates designated for different time points (0, 5, 15, 30, 45 minutes) on ice.
(5) For 0min, 135. mu.L of IS-containing ACN were added to the wells of the 0min plate, followed by 15. mu.L of NADPH stock solution (6 mM).
(6) All other plates were preincubated at 37 ℃ for 5 minutes.
(7) To the plate, 15. mu.L of NADPH stock solution (6mM) was added to start the reaction and timing.
(8) At 5min, 15min, 30 min and 45 min, 135. mu.L of IS-containing ACN was added to the wells of the respective plates to stop the reaction.
(9) After quenching, plates were shaken on a shaker (IKA, MTS 2/4) for 10 minutes (600 rpm) and then centrifuged at 5594g for 15 minutes (Thermo Multifuge. times.3R).
(10) From each well, 50 μ L of supernatant was transferred to a 96-well sample plate containing 50 μ L of ultrapure water (Millipore, ZMDS 50F01) for LC/MS analysis.
(11) And fitting a curve according to the peak areas of the compounds at different time points, and calculating the half-life and the clearance rate.
TABLE 3 hepatic microsome stability data for some of the compounds of the examples of the invention
Compound numbering T1/2(minute) Clint(mL/min/kg)
GS-4997 212 8.21
I-1 821.4 2.12
The I-1 compound shows in vitro human liver microsome metabolic stability superior to the positive drug GS-4997.
4. Rat in vivo PK
Experimental procedures
(1)6 male SD Rat,198-225g (JH Laboratory Animal Co. LTD), three intravenous injection groups, and 3 oral administration groups. The intravenous injection group is normally fed with food and water; oral groups were fasted overnight in advance and fed 4h after dosing. (2) Intravenous injection is carried out on the dorsum of foot at the concentration of 1mg/kg in an intravenous injection group, and oral gavage is carried out at the concentration of 10mg/kg in an oral group. (3) Animals were manually restrained at the indicated time points and approximately 150 μ L of blood samples were collected via the tail vein for serial infiltration into EDTA-K2 tubes. Blood samples were first stored in wet ice and centrifuged within 15 minutes after sampling to obtain plasma (2000g, 4 ℃, 5 minutes). The groups were given 0.083, 0.25, 0.5, 1,2,4, 8, 24h,8 time points for the group given intravenous injection, and 0.25, 0.5, 1,2,4, 8, and 24h, 7 time points for the group given oral administration. (4) Blood samples were stored at around-70 ℃ until analysis. (5) According to the blood concentration at each time point, the oral group calculates CL and Vss、AUClast、AUCINF、T1/2、MRTINF(ii) a Calculation of T by intravenous injection groupmax、Cmax、AUClast、AUCINF、T1/2、F。
TABLE 4 in vivo PK data in rats for I-1 Compounds
Figure BDA0002618861580000241
NA:Not available
TABLE 5 in vivo PK data in rats for I-4 Compounds
Figure BDA0002618861580000242
The I-1 and I-4 compounds show good in vivo pharmacokinetic properties, have conditions for carrying out in vivo pharmacodynamic experiments, and have excellent druggability.
5. CDAA (CDAA-induced nucleic acid sequence analysis) induced NASH (NASH) mouse pharmacodynamic model research
Purpose of the experiment: establishing a NASH (non-alcoholic steatohepatitis) in-vivo pharmacodynamic evaluation model, and carrying out pharmacodynamic study on the tested medicament in the NASH model.
Experimental materials: CDAA feed (Packo: Research Diets: A06071302), 10% neutral formaldehyde fixing solution (formalin, Chengdu Li Bio-technology Co., Ltd.), methylcellulose (MC, Shanghai Merlian Bio Co., Ltd.), Tween 80 (Shanghai Merlian Bio Co., Ltd.), C57BL/6J mouse (Beijing Wintoli laboratory animal technology Co., Ltd.), animal weighing scale (Shanghai Hua Chao electric appliances Co., Ltd.), microscope (MIC01964, Chuiss optical instrument), pipette (Eppendorf)
And (3) experimental operation:
(1) c57BL/6J mice were housed in SPF cell houses for at least one week of acclimation.
(2) The group was divided into a normal Control group (Control), a model group (Vehicle), and a compound of example 5 50mpk group by body weight and food intake. The other animals were fed CDAA feed except the normal control group. The molding time of CDAA was 12 weeks, and the administration was from week 5 to week 12. The administration route is intragastric administration, and the administration frequency is once a day. Performing cage-side observation every day, and measuring food intake and body weight once per week before administration; after administration, food intake and body weight were measured twice a week and once at 3-4 d.
(3) Fasting for more than 6 hours before dissection, anesthetizing by using 3% sodium pentobarbital, collecting blood by heart blood collection, centrifuging, taking supernatant, and sending the supernatant to a sample to detect blood biochemical indexes: ALP, TC, HDL, LDL.
(4) Experimental data for each group of animals are described as mean ± standard error (X ± SEM). The comparison between groups with uniform normal and variance adopts One-Way ANOVA (One-Way ANOVA), and the comparison between groups adopts LSD (least squares difference) method for inspection; p <0.05 is statistically significant for differences. All statistical analyses were done using Graphpad Prism 8.0 software.
The experimental results are as follows:
TABLE 6 Effect of I-4 Compounds in a mouse model of NASH
Liver weight (g) ALP(U/L) HDL(mmol/L) LDL/TC
Control group 1.097 116.8 2.553 0.06895
Vehicle group 1.534 196.9 1.276 0.1302
EXAMPLE 4 group 1.382 158.0 1.404 0.1225
Compared with Vehicle, the I-4 compound down-regulates the liver weight, down-regulates ALP, up-regulates HDL and down-regulates LDL/TC of NASH mice, and shows certain NASH treatment potential.
6. Research on drug effect model of DSS-induced acute colitis mouse
The experimental method comprises the following steps: (1) ICR mice, weight 18-22g, 6-8 weeks old, adaptive feeding 5 days later, randomly divided into 4 groups: control, DSS, GS-4997, example 4, 6 pieces per group. (2) Acute colitis was induced by 3% DSS, and the 3% DSS-containing aqueous solution was administered to the DSS, GS-4997, example 4 groups and allowed to drink freely. GS-4997 was administered at a dose of 25mg/kg, example 5 at a dose of 25mg/kg, and the Control (Control) was given normal drinking water for 7 consecutive days, with replacement every two days. During the molding period, the medicine is administered by means of intragastric administration, and the Control group and the DSS group are administered with the corresponding solvent CMC-Na once a day for 7 days continuously. (3) After 7 days, the mice were sacrificed by cervical dislocation, colon tissues were taken, the length of the colon was measured, 3 of each group were fixed in formalin solution for HE staining, and the remaining tissues were stored at-80 ℃.
The experimental results are as follows: the effect of the I-4 compounds on colon length in DSS-induced colitis mice is shown in FIG. 1.
The colon length was significantly shortened in DSS mice compared to Control mice: (###P<0.001). The colon length of the I-4 mice was significantly increased compared to the DSS group (. about.P)<0.05), while the colon length of the GS-4997 group mice is increased to a certain extent without significant difference. The effect of the I-4 compound on colon histopathology in DSS-induced colitis mice is shown in FIG. 2.
The colon tissue of the mice in the Control group is intact, and inflammatory cell infiltration phenomenon is not seen. DSS mice have severe damage to colon tissue and a large number of inflammatory cells infiltrate. The colon tissue of the mice in the I-4 compound administration group is relatively complete, and the inflammation is obviously reduced; the GS-4997 group mice had some reduction in tissue damage.
The results of two experiments of the I-4 compound in a mouse model of DSS-induced colitis show that the I-4 compound has a certain treatment effect on ulcerative colitis and is superior to the positive drug GS-4997.

Claims (10)

1. An indole ASK1 small molecule inhibitor, which is characterized by comprising a compound shown as a general formula (I) and a stereoisomer or pharmaceutically acceptable salt thereof:
Figure FDA0002618861570000011
M1is selected from N or CH;
R1is the same or different and is independently selected from hydrogen atom, alkoxy, halogen, heterocyclic radical, aryl, heteroaryl, - (CH)2)nOR4、-(CH2)nNR5R6、-(CH2)nNR5C(O)R4N is 0, 1,2,3, 4 or 5; wherein the heteroaryl is unsubstituted or substituted by one or more substituents selected from alkyl, alkoxy, and heterocyclyl;
R2the same or different, and each is independently selected from hydrogen atom, alkyl, halogen, aryl, heteroaryl;
R3selected from alkyl, hydroxyalkyl or heterocyclyl;
R4selected from alkyl groups;
R5and R6The same or different, and each is independently selected from hydrogen atom, heterocyclic radical;
x is selected from N (R)2);
m is 0, 1,2,3 or 4;
p is 1 or 2.
2. The indole ASK1 small-molecule inhibitor according to claim 1, wherein M is1Is N:
Figure FDA0002618861570000012
3. the indole ASK1 small-molecule inhibitor according to claim 2, wherein X is imino and R is R3Is isopropyl, and is specifically shown as a general formula (III):
Figure FDA0002618861570000021
4. the indole ASK1 small molecule inhibitor according to any one of claims 1-2, wherein R is1Selected from the group consisting of a hydrogen atom, a methoxy group, a chlorine atom, a fluorine atom, a 3, 6-dihydro-2H-pyran-4-yl group, a 1,2,3, 6-tetrahydropyridin-4-yl group, a phenyl group, a pyrazolyl group, a thienyl group, a furyl group, a pyridyl group, a 1- (1-ethoxyethyl) -1H-pyrazol-4-yl group, a 1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl group, a 1-methyl-1H-pyrazol-4-yl group, and an acetamido group; r2Selected from hydrogen atom, methyl, furyl; r3Selected from 1-hydroxypropan-2-yl, isopropyl; r4Is C1-6An alkyl group; r5Is a hydrogen atom; r6Is tetrahydro-2H-pyran-4-yl.
5. The indole ASK1 small molecule inhibitor according to claim 1, selected from the group consisting of: n- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-1), 7-chloro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-2), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6-methoxy-1H-indole-2-carboxamide (I-3), 6-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-4), 5-fluoro-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-5), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1-methyl-1H-indole-2-carboxamide (I-6), (R) -N- (6- (4- (4- (1-hydroxypropan-2-yl) -4H-1,2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-7), N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-8), 6-fluoro-N- (3- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-indole-2-carboxamide (I-9), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -3-methyl-1H-indole-2-carboxamide (I-10), 3- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-11), 7-acetamido-N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-12), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-13), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -5- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-14), 6- (1- (1-ethoxyethyl) -1H-pyrazol-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-15), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-16), N- (3- (5- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) -1H-indazol-3-yl) phenyl) -4- (trifluoromethyl) benzamide (I-17), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- (1- (tetrahydro-2H-pyran-4-yl) -1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-18), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -4- (1-methyl-1H-pyrazol-4-yl) -1H-indole-2-carboxamide (I-19), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (thiophen-3-yl) -1H-indole-2-carboxamide (I-20), 7- (furan-3-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-21), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7-phenyl-1H-indole-2-carboxamide (I-22), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (pyridin-4-yl) -1H-indole-2-carboxamide (I-23), 7- (3, 6-dihydro-2H-pyran-4-yl) -N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -1H-indole-2-carboxamide (I-24), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -7- (1,2,3, 6-tetrahydropyridin-4-yl) -1H-indole-2-carboxamide (I-25), N- (6- (4-isopropyl-4H-1, 2, 4-triazol-3-yl) pyridin-2-yl) -6- ((tetrahydro-2H-pyran-4-yl) amino) -1H-indole-2-carboxamide (I-26).
6. The indole ASK1 small molecule inhibitor according to claim 1, wherein the pharmaceutically acceptable salt is a salt of the inhibitor with an acid or a base, the acid being hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, naphthalenesulfonic acid, citric acid, tartaric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, succinic acid, fumaric acid, salicylic acid, phenylacetic acid, or mandelic acid; the base is an inorganic base containing a basic metal cation, an alkaline earth metal cation, or an ammonium cation salt.
7. The preparation method of the indole ASK1 small-molecule inhibitor as claimed in claim 1, comprising the following steps:
(1) methyl 3-aminobenzoate or methyl 6-aminopyridine-2-carboxylate A is firstly converted into a hydrazide compound B;
(2) a compound C is obtained through ring closing reaction, and the compound C reacts with a carboxylic acid compound D to obtain a compound (I);
Figure FDA0002618861570000031
and (3) adding a corresponding acid or alkali solution into the solution of the compound (I) prepared by the method, and removing the solvent under reduced pressure after salt formation is completed to obtain the pharmaceutically acceptable salt of the ASK1 inhibitor.
8. A pharmaceutical composition comprising the indole ASK1 small molecule inhibitor of claim 1 and one or more pharmaceutically acceptable carriers, diluents, or excipients.
9. The use of the indole ASK1 small molecule inhibitor of claim 1 or the pharmaceutical composition of claim 8 for the preparation of an ASK1 inhibitor medicament.
10. Use of the indole ASK1 small molecule inhibitor of claim 1 or the pharmaceutical composition of claim 8 for the preparation of a medicament for the treatment of inflammatory diseases and ASK 1-related diseases; the inflammatory diseases are non-alcoholic steatohepatitis and ulcerative colitis.
CN202010777152.8A 2020-08-05 2020-08-05 Indole ASK1 small molecule inhibitor and preparation method and application thereof Pending CN111808079A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010777152.8A CN111808079A (en) 2020-08-05 2020-08-05 Indole ASK1 small molecule inhibitor and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010777152.8A CN111808079A (en) 2020-08-05 2020-08-05 Indole ASK1 small molecule inhibitor and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN111808079A true CN111808079A (en) 2020-10-23

Family

ID=72863886

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010777152.8A Pending CN111808079A (en) 2020-08-05 2020-08-05 Indole ASK1 small molecule inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111808079A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112300134A (en) * 2020-10-30 2021-02-02 中国药科大学 ASK1 inhibitor and derivative thereof, preparation method, pharmaceutical composition and application

Citations (7)

* 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
CN102834393A (en) * 2010-02-03 2012-12-19 武田药品工业株式会社 Apoptosis signal-regulating kinase 1 inhibitors
CN104080771A (en) * 2012-01-27 2014-10-01 吉利德科学公司 Apoptosis signal-regulating kinase inhibitor
CN105198789A (en) * 2015-10-26 2015-12-30 山东大学 Substituted3-indole Bcl-2 protein inhibitor and preparation method and application thereof
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
WO2018233553A1 (en) * 2017-06-19 2018-12-27 广东东阳光药业有限公司 Fused bicyclic compound and use thereof in medicine

Patent Citations (8)

* 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
CN102834393A (en) * 2010-02-03 2012-12-19 武田药品工业株式会社 Apoptosis signal-regulating kinase 1 inhibitors
CN104080771A (en) * 2012-01-27 2014-10-01 吉利德科学公司 Apoptosis signal-regulating kinase inhibitor
CN105198789A (en) * 2015-10-26 2015-12-30 山东大学 Substituted3-indole Bcl-2 protein inhibitor and preparation method and application thereof
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
CN109071498A (en) * 2017-02-16 2018-12-21 四川科伦博泰生物医药股份有限公司 Kinase inhibitor and its preparation method and application
WO2018233553A1 (en) * 2017-06-19 2018-12-27 广东东阳光药业有限公司 Fused bicyclic compound and use thereof in medicine

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112300134A (en) * 2020-10-30 2021-02-02 中国药科大学 ASK1 inhibitor and derivative thereof, preparation method, pharmaceutical composition and application

Similar Documents

Publication Publication Date Title
CN106928219B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition and application
KR20070089201A (en) Pyrrolopyrazines and pyrazolopyrazines useful as inhibitors of protein kinases
AU2006210626A1 (en) Raf inhibitor compounds and methods
AU2006284751A1 (en) Raf inhibitor compounds and methods of use thereof
JPH10512896A (en) Aryl and heteroaryl purine compounds
JP2003513091A (en) Pyrazolones substituted with heterocycles
WO2014040549A1 (en) Alkynyl heteroaromatic ring compound and application thereof
WO2000023444A1 (en) 5,7-disubstituted-4-aminopyrido[2,3-d]pyrimidine compounds
AU2018248581B2 (en) Heteroaromatic compounds useful in therapy
CN109311891A (en) The crystallization of Pyrrolopyrimidine compounds as JAK inhibitor
CN111542522B (en) Substituted pyrazolopyrimidines useful as kinase inhibitors
KR20180066985A (en) Thienopyrimidine derivative and use thereof
CN116406271A (en) Bicyclic compounds
CA2508319A1 (en) Jnk inhibitors
JP2001520655A (en) 5,7-Disubstituted 4-aminopyrido [2,3-D] pyrimidine compounds and their use as adenosine kinase inhibitors
CA2602303A1 (en) Tricyclic azole derivatives, their manufacture and use as pharmaceutical agents
WO2020125513A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CN111808079A (en) Indole ASK1 small molecule inhibitor and preparation method and application thereof
CN112513027A (en) Indazole amine derivative, preparation method and medical application thereof
CN113416181B (en) Quinazoline derivative and application thereof
CN116782903A (en) Substituted pyridone compounds and uses thereof
KR101546743B1 (en) Indole derivatives, Abl kinase inhibiting composition and pharmaceutical compositions for prevention and treatment of abnormal cell growth diseases comprising the same
CN112654604B (en) 7-substituted indazole derivative, preparation method and medical application thereof
CN114315837A (en) Crystalline form of ERK inhibitor and preparation method thereof
CN108752336B (en) Imidazoquinoline derivatives 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
RJ01 Rejection of invention patent application after publication

Application publication date: 20201023

RJ01 Rejection of invention patent application after publication