CN112174945A - Indazole compound with anticancer effect and preparation method and application thereof - Google Patents

Indazole compound with anticancer effect and preparation method and application thereof Download PDF

Info

Publication number
CN112174945A
CN112174945A CN201910589317.6A CN201910589317A CN112174945A CN 112174945 A CN112174945 A CN 112174945A CN 201910589317 A CN201910589317 A CN 201910589317A CN 112174945 A CN112174945 A CN 112174945A
Authority
CN
China
Prior art keywords
alkyl
compound
halogen
unsubstituted
reaction
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
CN201910589317.6A
Other languages
Chinese (zh)
Other versions
CN112174945B (en
Inventor
余洛汀
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.)
Sichuan University
Original Assignee
Sichuan 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 Sichuan University filed Critical Sichuan University
Priority to CN201910589317.6A priority Critical patent/CN112174945B/en
Publication of CN112174945A publication Critical patent/CN112174945A/en
Application granted granted Critical
Publication of CN112174945B publication Critical patent/CN112174945B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to an indazole compound with an anticancer effect, a preparation method and application thereof, belonging to the field of medicines. The invention provides a compound shown as a formula I or a pharmaceutically acceptable salt thereof. Biological experiments show that the compound of the invention has obvious inhibition effect on BRD4 protein and is a potential cancer treatment drug targeting BRD 4. And, partIC for inhibiting proliferation of acute myeloid leukemia cells by differential compounds50The value can reach below 2 mu M, and the cell can be blocked at G0/G1And effectively inhibit the transcription and expression of downstream genes. The application of the invention can provide a new drug selection for the clinical treatment of cancer.
Figure DDA0002115456680000011

Description

Indazole compound with anticancer effect and preparation method and application thereof
Technical Field
The invention relates to an indazole compound with an anticancer effect, a preparation method and application thereof, belonging to the field of medicines.
Background
Cancer is one of the major public health problems recognized in the world, and the traditional means for treating cancer include surgical resection, radiotherapy, chemotherapy and the like. Epigenetic targeted therapy has achieved significant efficacy in the treatment of certain types of cancer starting in the late 90 s, as effective as chemotherapy, but with much reduced side effects compared to chemotherapy, and is currently being investigated for fire fever and a very promising area. Epigenetic refers to the phenomenon that changes in gene expression do not involve changes in DNA sequence, but can be stably inherited as cells divide and proliferate. When affected by external environment, the normal epigenetic state of the cell may be disrupted, resulting in abnormal activation of the oncogene or inactivation of the tumor suppressor gene, promoting tumor formation. The posttranslational modification of histone is an important component of epigenetic regulation, wherein, lysine acetylation modification on histone is an important mechanism for regulating chromatin structure and activating transcription, and is one of important action mechanisms for survival and growth of tumor cells. The acetylation level of histone is commonly regulated by multiple enzyme families and proteins, and the whole regulation process is completed through a ' write ', read and erase ' mechanism. The key to this process is the recognition of specific histone post-translational modifications by the reader bromodomain protein. Abnormal acetylation levels are closely associated with the occurrence of various diseases, such as: inflammation, immune and metabolic diseases, various malignant tumors, and the like. Thus, the bromodomain and terminal domain (BET) subfamilies have become a popular anti-tumor target as proteins that play an important role in epigenetic regulation of gene transcription. Bromodomain-containing protein 4(BRD4) is one of the important members of the BET family and has been considered as the most attractive candidate target for targeted gene transcription therapy in several cancers.
Disclosure of Invention
The invention aims to provide an indazole compound with an anticancer effect, and a preparation method and application thereof.
The present invention provides a compound of formula i or a pharmaceutically acceptable salt thereof:
Figure BDA0002115456660000011
R1、R2、R3、R4among which only one is selected from
Figure BDA0002115456660000021
The remaining three are independently selected from-H, halogen, alkyl, alkoxy, -NH2、-NO2or-CN, wherein R10、R11、R12、R13、R14Independently selected from-H or alkyl;
R5is selected from-H or-X-R7Wherein X is-CH2-or is absent, R7Selected from aryl, cycloalkyl, acyl or alkyl;
R6is selected from-H or-NH-R8Wherein R is8Is an aryl group.
Further, R1、R2、R3、R4Among which only one is selected from
Figure BDA0002115456660000022
Figure BDA0002115456660000023
The other three are independently selected from-H, halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen substituted C1-C6 alkyl, -NH2、-NO2or-CN, wherein R10、R11、R12、R13、R14Independently selected from-H or unsubstituted C1-C6 alkyl.
Preferably, R1、R2、R3、R4Among which only one is selected from
Figure BDA0002115456660000024
The other three are independently selected from-H, halogen, -OCH3or-CH3Wherein R is10、R11、R12、R13、R14Independently selected from-H, -CH3or-CH2CH3
Preferably, R1、R2、R3、R4Middle R2Or R3Is composed of
Figure BDA0002115456660000025
Or R3Is composed of
Figure BDA0002115456660000026
The other three are independently selected from-H or-OCH3Wherein R is10、R11、R14Are all-CH3,R12is-H, R13is-CH3or-CH2CH3
Most preferably, R3Is composed of
Figure BDA0002115456660000031
R1、R2、R4Independently selected from-H or-OCH3
Further, R5Is selected from-H or-X-R7Wherein X is-CH2-or is absent, R7Selected from 5-to 14-membered aryl, 3-to 6-membered cycloalkyl,
Figure BDA0002115456660000032
Or C1-C6 alkyl, wherein R9Is C1-C6 alkoxy.
Preferably, R7Selected from 6-membered aryl, 3-to 6-membered cycloalkyl,
Figure BDA0002115456660000033
Or C1-C6 alkyl, wherein R9Is C1-C6 alkoxy.
Preferably, R7Selected from substituted or unsubstituted phenyl, 3-6 membered cycloalkyl or
Figure BDA0002115456660000034
Wherein R is9is-OCH2CH3
Preferably, the substituted phenyl group containsAt least one substituent selected from the group consisting of: halogen, alkyl, alkoxy, -NO2
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen substituted C1-C6 alkyl, -NO2
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: -F, -CH3、-CF3、-OCH3、-NO2
Preferably, the 3-6 membered cycloalkyl group is selected from
Figure BDA0002115456660000035
Most preferably, R5is-X-R7Wherein X is-CH2-,R7Is selected from
Figure BDA0002115456660000036
Figure BDA0002115456660000041
Further, R6Is selected from-H or-NH-R8Wherein R is8Is a 5-to 14-membered aryl group.
Preferably, R8Is 6-membered aryl.
Preferably, R8Is a substituted or unsubstituted phenyl group.
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, alkyl, alkoxy or-CN.
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen substituted C1-C6 alkoxy or-CN.
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: -F, -Cl, -OCH3、-CF3
Figure BDA0002115456660000042
or-CN.
Most preferably, R6is-H.
Further, the compound is selected from:
Figure BDA0002115456660000043
Figure BDA0002115456660000051
the invention provides a preparation method of the compound or the pharmaceutically acceptable salt thereof, which comprises the following steps:
when R is1、R2、R3、R4Among them and only one is
Figure BDA0002115456660000052
In time, the method one is adopted: carrying out coupling reaction on the raw materials SM-1 and SM-2 to obtain a target product:
Figure BDA0002115456660000053
wherein R is1a、R2a、R3a、R4aOne and only one is halogen, and the other three are R at the same position of benzene ring in the product1、R2、R3、R4The groups are the same; a is
Figure BDA0002115456660000054
R15、R16Independently selected from-H or alkyl, or joined to form cycloalkyl;
or, when R is1、R2、R3、R4Among them and only one is
Figure BDA0002115456660000055
Then, the method II is adopted: carrying out coupling reaction on the raw materials SM-3 and SM-4 to obtain a target product:
Figure BDA0002115456660000061
wherein R is1b、R2b、R3b、R4bOne and only one is
Figure BDA0002115456660000062
The other three R are at the same position of benzene ring in the product1、R2、R3、R4The radicals R are the same17、R18Independently selected from-H or alkyl, or joined to form a cycloalkyl group, Y is halogen.
Preferably, R15、R16Independently selected from-H or unsubstituted C1-C6 alkyl, or joined to form a 5-membered cycloalkyl group.
Most preferably, the halogen is-Br and A is
Figure BDA0002115456660000063
Preferably, R17、R18Independently selected from-H or unsubstituted C1-C6 alkyl, or joined to form a 5-membered cycloalkyl group.
Most preferably, R1b、R2b、R3b、R4bOne and only one is
Figure BDA0002115456660000064
Y is-Br.
Further, the preparation method meets at least one of the following conditions:
in the first method and the second method, a palladium catalyst and alkali are added into a reaction system;
preferably, the palladium catalyst is Pd (dppf) Cl2
Preferably, the base is sodium carbonate;
preferably, SM-2: SM-1: pd (C)dppf)Cl2: the molar ratio of sodium carbonate is 1: 1.5: 0.05: 3;
preferably, SM-3: SM-4: pd (dppf) Cl2: the molar ratio of sodium carbonate is 1: 1.5: 0.05: 3;
the reaction solvent is a mixed solvent of dioxane and water;
preferably, the ratio of dioxane: the volume ratio of water is 2: 1;
carrying out reaction under a protective atmosphere;
the reaction temperature is 90 ℃;
the invention provides application of the compound or the pharmaceutically acceptable salt thereof in preparing BRD4 inhibitor medicines.
The invention provides application of the compound or the pharmaceutically acceptable salt thereof in preparing a medicament for treating and/or preventing cancer.
Further, the medicament is a medicament for treating and/or preventing leukemia.
Preferably, the leukemia is acute myeloid leukemia.
The invention provides a pharmaceutical composition for preventing and treating cancer, which is a preparation prepared by taking the compound or pharmaceutically acceptable salt thereof as an active ingredient and adding acceptable auxiliary materials or auxiliary ingredients.
Further, the agent is an agent for treating and/or preventing leukemia.
Preferably, the leukemia is acute myeloid leukemia.
Definition of terms:
the compounds and derivatives provided by the present invention may be named according to the IUPAC (international union of pure and applied chemistry) or CAS (chemical abstracts service, Columbus, OH) naming system.
The term "alkyl" is a radical of a straight or branched chain saturated hydrocarbon group. C1~C6Examples of alkyl groups include, but are not limited to, methyl (C)1) Ethyl (C)2) N-propyl (C)3) Isopropyl (C)3) N-butyl (C)4) Tert-butyl (C)4) Sec-butyl (C)4) Isobutyl (C)4) N-pentyl group (C)5) 3-pentyl radical (C)5) Pentyl group (C)5) Neopentyl (C)5) 3-methyl-2-butyl (C)5) Tert-amyl (C)5) And n-hexyl (C)6). Unless otherwise indicated, each instance of alkyl is independently optionally substituted, i.e., unsubstituted or substituted with one or more substituents. "substituted" means that a hydrogen atom in a molecule is replaced by a different atom or molecule. In some embodiments, said C1~C6Alkyl is C substituted by halogen (fluorine, chlorine, bromine, iodine)1~C6An alkyl group. At C1~C6In the case where an alkyl group is substituted with a substituent, the number of carbon atoms of the substituent is not counted in.
The term "alkoxy" refers to the group-OR, wherein R is alkyl as defined above. C1~C6Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1, 2-dimethylbutoxy. Said C is1~C6Alkoxy in the case where R is substituted with a substituent, the number of carbon atoms of the substituent is not counted in.
The term "aryl" refers to a radical of a 4n +2 aromatic ring system with or without heteroatoms in the aromatic ring system, wherein the heteroatoms are selected from nitrogen, oxygen and/or sulfur, among others. Unless otherwise indicated, each instance of an aryl group is independently optionally substituted, i.e., unsubstituted or substituted with one or more substituents.
The term "cycloalkyl" refers to a saturated cyclic hydrocarbon group, with or without heteroatoms, which may be a single ring structure or two or more rings, wherein the heteroatoms are selected from boron, phosphorus, sulfur, oxygen and/or nitrogen, and the like. Unless otherwise indicated, each instance of a cycloalkyl group is independently optionally substituted, i.e., unsubstituted or substituted with one or more substituents.
The term "acyl" refers to R '-c (o) -, where R' is an optional group.
The term "pharmaceutically acceptable" means that the carrier, cargo, diluent, adjuvant, and/or salt formed is generally chemically or physically compatible with the other ingredients comprising a pharmaceutical dosage form and physiologically compatible with the recipient.
The term "pharmaceutically acceptable salts" refers to acid and/or base salts of the compounds of the present invention with inorganic and/or organic acids and bases, and also includes zwitterionic salts (inner salts), and also includes quaternary ammonium salts, such as alkylammonium salts. These salts can be obtained directly in the final isolation and purification of the compounds. The compound may be obtained by appropriately (e.g., equivalent) mixing the above compound with a certain amount of an acid or a base. These salts may form precipitates in the solution which are collected by filtration, or they may be recovered after evaporation of the solvent, or they may be prepared by reaction in an aqueous medium followed by lyophilization. The salt in the invention can be hydrochloride, sulfate, citrate, benzene sulfonate, hydrobromide, hydrofluoride, phosphate, acetate, propionate, succinate, oxalate, malate, succinate, fumarate, maleate, tartrate or trifluoroacetate of the compound.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration.
The pharmaceutically acceptable auxiliary material of the invention refers to a substance contained in a dosage form except for an active ingredient.
The pharmaceutically acceptable auxiliary components have certain physiological activity, but the addition of the components does not change the dominant position of the pharmaceutical composition in the disease treatment process, but only plays auxiliary effects, and the auxiliary effects are only the utilization of the known activity of the components and are auxiliary treatment modes which are commonly used in the field of medicine. If the auxiliary components are used in combination with the pharmaceutical composition of the present invention, the protection scope of the present invention should still be included.
The invention provides an indazole derivative with novel structure. Biological experiments show that the compounds have obvious inhibition effect on BRD4 protein and are potential cancer treatment drugs targeting BRD4. Furthermore, part of the compounds inhibit the IC of acute myeloid leukemia cell proliferation50The value can reach below 2 mu M, and the cell can be blocked at G0/G1And effectively inhibit the transcription and expression of downstream genes. The application of the invention can provide a new drug selection for the clinical treatment of cancer.
Drawings
FIG. 1 is a graph showing the results of the cytotoxicity test in test example 1;
FIG. 2 is a graph showing the results of cell cycle measurements in test example 2;
FIG. 3 is a graph showing the results of Western blot analysis in test example 3.
Detailed Description
The scheme of the invention will be explained with reference to the examples. It will be appreciated by those skilled in the art that the following examples are illustrative of the invention only and should not be taken as limiting the scope of the invention. The examples, where specific techniques or conditions are not indicated, are to be construed according to the techniques or conditions described in the literature in the art or according to the product specifications. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
Example 1 compound 5 a: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -N- (4-fluorophenyl) -1H-indazol-3-amine
Figure BDA0002115456660000091
Dissolving compound 1(1.0eq) and potassium hydroxide solid (2.0eq) in DMF, stirring at room temperature, and slowly adding elementary iodine I after complete dissolution2(1.0eq), and the reaction is continued for 1-2 hours to obtain an intermediate 2. The yield was 85%.
Figure BDA0002115456660000092
Weighing the intermediate 2(1.0eq) and cesium carbonate (2.0eq) in a dry reaction flask, using dry DMF as a reaction solvent, and adding para-fluoroaniline liquid (1.2eq) into the reaction flask by using a liquid transfer gun. N is a radical of2Replacement protection is carried out by pumping out water and O in the reaction flask2And the anhydrous and oxygen-free environment is kept. Adding catalyst and ligand Pd (AcO)2(0.1eq), Xantphos (0.1 eq). Then nitrogen replacement is carried out for many times. Transferring to an oil bath at 60 ℃ for reaction, detecting the reaction progress by TLC after 6-7 hours, and supplementing Pd (AcO) if the reaction is not completely carried out2The reaction was continued with Xantphos. After the reaction is completed, filtering with diatomite, removing solvent by rotary evaporation, and using EA/H2Extracting with O/saturated saline solution, drying with anhydrous magnesium sulfate, and performing column chromatography purification on the crude product obtained after rotary evaporation. The purified product is put into the next reaction.
Figure BDA0002115456660000093
The product obtained above (1.0eq) and 3, 5-dimethylisoxazoleboronic acid (1.5eq) were dissolved in a mixture of dioxane and water (dioxane: water 2:1) and stirred at room temperature. Sodium carbonate solids (3.0eq) were then added and stirred well. Finally weighing Pd (dppf) Cl2(0.05eq) is added into the mixed solution, nitrogen is replaced and protected, and the mixed solution is transferred into an oil bath at the temperature of 90 ℃ for heating reaction for 6 to 7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. And (3) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 5 a. The yield was 50%.
1HNMR(400MHz,Chloroform-d)9.55(s,1H),7.60(dd,J=8.3,0.8Hz,1H),7.41–7.35(m,2H),7.23(t,J=1.1Hz,1H),7.04–6.99(m,2H),6.97(dd,J=8.3,1.3Hz,1H),6.33(s,1H),2.43(s,3H),2.30(s,3H).
13CNMR(101MHz,Chloroform-d)165.56,158.77,156.40,145.81,141.85,138.46,138.44,129.85,121.13,119.90,118.36,118.28,116.79,115.81,115.58,114.44,110.38,11.61,10.84.
ESI-HRMS:323.1311,[M+1]+.
Example 2 compound 5 b: preparation of 6- (3,5-dimethylisoxazol-4-yl) -N- (4-fluoro-3-methoxy phenyl) -1- (tetrahydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000101
1.0eq of intermediate 2 was dissolved in dichloromethane solvent and stirred at room temperature. 3, 4-dihydropyran (2.0eq) and p-toluenesulfonic acid (0.03eq) were then added slowly and left at room temperature overnight. After TLC detection reaction is completed, extracting with water/ethyl acetate/saturated saline; dried over anhydrous magnesium sulfate. And carrying out reduced pressure distillation to obtain a crude product, and carrying out column chromatography purification to obtain the target intermediate 3 a. The yield was 60%.
Figure BDA0002115456660000102
Putting the intermediate 3a (1.0eq) and cesium carbonate (2.0eq) in a dry reaction flask, using dry DMF as a reaction solvent, and adding m-methoxy-p-fluoroaniline (1.2eq) into the reaction flask by using a liquid-transferring gun. N is a radical of2Replacement protection is carried out by pumping out water and O in the reaction flask2And the anhydrous and oxygen-free environment is kept. Adding catalyst and ligand Pd (AcO)2(0.1eq), Xantphos (0.1 eq). Then nitrogen replacement is carried out for many times. Transferring to an oil bath at 60 ℃ for reaction, detecting the reaction progress by TLC after 6-7 hours, and supplementing Pd (AcO) if the reaction is not completely carried out2The reaction was continued with Xantphos. After the reaction is completed, filtering with diatomite, removing solvent by rotary evaporation, and using EA/H2Extracting with O/saturated saline solution, drying with anhydrous magnesium sulfate, and performing column chromatography purification on the crude product obtained after rotary evaporation.
Figure BDA0002115456660000111
The resulting purified product and 3, 5-dimethylisoxazoleboronic acid (1.5eq) were dissolved in a mixture of dioxane and water (dioxane: water: 2:1) and stirred at room temperature. Sodium carbonate solids (3.0eq) were then added and stirred well. Finally weighing Pd (dppf) Cl2(0.05eq) is added into the mixed solution, nitrogen is replaced and protected, and the mixed solution is transferred into an oil bath at the temperature of 90 ℃ for heating reaction for 6 to 7 hours. TLC detection reaction is completeThen, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline; dried over anhydrous magnesium sulfate. And (3) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 5 b. The yield was 50%.
1HNMR(400MHz,Chloroform-d)8.15(dd,J=8.7,6.0Hz,1H),7.66(d,J=8.2Hz,1H),7.33(s,1H),6.98(dd,J=8.2,1.3Hz,1H),6.82(s,1H),6.73–6.64(m,2H),5.61(dd,J=9.3,2.7Hz,1H),4.05(d,J=11.4Hz,1H),3.95(s,3H),3.73(t,J=9.5Hz,1H),2.62(q,J=9.2,8.5Hz,1H),2.44(s,3H),2.30(s,3H),2.16(dd,J=28.9,12.3Hz,2H),1.81–1.62(m,3H).
13CNMR(101MHz,Chloroform-d)165.39,165.27,158.61,158.51,135.94,133.55,130.21,129.46,128.81,127.71,123.71,122.28,121.69,116.82,116.08,109.62,67.37,56.00,52.86,29.38,25.21,22.72,11.55,11.53,10.77.
ESI-HRMS:437.1980,[M+1]+.
Example 3 compound 5 c: preparation of 6- (3,5-dimethylisoxazol-4-yl) -N- (4-fluorophenyl) -1- (tetrahydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000112
Compound 5c was prepared according to a similar procedure as example 2.
1HNMR(400MHz,Chloroform-d)7.54(dd,J=8.3,0.8Hz,1H),7.41–7.36(m,2H),7.34(d,J=1.1Hz,1H),7.04–6.98(m,2H),6.96(dd,J=8.3,1.3Hz,1H),6.21(s,1H),5.61(dd,J=9.5,2.6Hz,1H),4.06(d,J=11.8Hz,1H),3.77–3.72(m,1H),2.58(d,J=12.0Hz,1H),2.44(s,3H),2.30(s,3H),2.17(s,1H),2.11(d,J=14.3Hz,1H),1.76(d,J=7.9Hz,1H),1.67(s,1H).
13CNMR(101MHz,Chloroform-d)165.50,158.76,144.51,140.91,138.34,133.76,131.02,129.69,121.41,119.90,118.14,118.06,116.89,115.72,115.50,110.52,67.52,29.43,25.18,22.74,11.64,10.86.
ESI-HRMS:407.1883,[M+1]+.
Example 4 compound 5 d: preparation of 6- (3,5-dimethylisoxazol-4-yl) -N- (4-fluoro-2-methoxy phenyl) -1- (tetra-hydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000121
Compound 5d was prepared according to a similar procedure as example 2.
1HNMR(400MHz,Chloroform-d)8.15(dd,J=8.7,6.0Hz,1H),7.66(dd,J=8.3,0.8Hz,1H),7.33(t,J=1.1Hz,1H),6.98(dd,J=8.3,1.3Hz,1H),6.82(s,1H),6.73–6.65(m,2H),5.61(dd,J=9.3,2.7Hz,1H),4.05(d,J=11.3Hz,1H),3.95(s,3H),3.76–3.68(m,1H),2.62(q,J=9.4,8.5Hz,1H),2.44(s,3H),2.30(s,3H),2.22–2.15(m,1H),2.12(d,J=13.0Hz,1H),1.82–1.70(m,2H),1.67(s,1H).
13CNMR(101MHz,Chloroform-d)165.45,144.41,140.64,129.65,121.22,119.52,116.96,116.14,115.91,115.82,110.42,106.57,106.36,98.67,98.40,85.06,67.37,56.00,31.59,29.38,25.21,22.72,22.66,14.12,11.64.
ESI-HRMS:437.1979,[M+1]+.
Example 5 compound 5 e: preparation of N- (4-chlorophenylyl) -6- (3,5-dimethylisoxazol-4-yl) -1- (tetrahydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000122
Compound 5e was prepared in 40% yield according to a similar procedure as example 2.
1HNMR(400MHz,Chloroform-d)7.57(d,J=8.3Hz,1H),7.37(d,J=2.1Hz,1H),7.35(q,J=1.7,1.2Hz,2H),7.24(d,J=2.1Hz,1H),6.98(dd,J=8.3,1.3Hz,1H),6.32(s,1H),5.62(dd,J=9.4,2.7Hz,1H),4.09–4.02(m,1H),3.78–3.70(m,1H),2.58(dtd,J=11.4,9.3,5.7Hz,1H),2.44(s,3H),2.30(s,3H),2.20–2.07(m,2H),1.82–1.71(m,3H).
13CNMR(101MHz,Chloroform-d)165.52,158.75,143.89,140.87,129.75,129.01,125.21,121.56,119.81,117.69,116.86,116.00,110.57,85.08,67.50,29.41,25.17,22.69,11.64,10.86.
ESI-HRMS:423.1591,[M+1]+.
Example 6 compound 5 f: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -N- (4-fluorophenyl) -1- (4-methyllbenzyl) -1H-indazol-3-amine
Figure BDA0002115456660000131
Sodium hydride solid (1.5eq) was weighed into a parallel reaction tube, replaced with nitrogen, and water vapor and oxygen were withdrawn. 1.0eq of intermediate 2 was dissolved in DMSO solvent, added to the reaction tube and stirred for 15 minutes. P-methylbenzyl bromide (1.2eq) was then added and nitrogen substitution was again carried out a number of times, maintaining an anhydrous and oxygen-free environment. The reaction was carried out at room temperature overnight. And detecting the reaction degree by TLC, adding water for quenching after the reaction is completed, and separating out a solid. Filtering with a baffle funnel, and washing with water for multiple times. Drying to obtain intermediate 4 a-2. The yield was 85%.
Figure BDA0002115456660000132
Taking the intermediate 4a-2(1.0eq) and cesium carbonate (2.0eq) in a dry reaction flask, taking dry DMF as a solvent, and taking para-fluoroaniline liquid (1.2eq) by a liquid transfer gun and adding the para-fluoroaniline liquid into the reaction flask. N is a radical of2Replacement protection, and maintenance of anhydrous and oxygen-free environment. Adding catalyst Pd (AcO)2(0.1eq) and ligand Xantphos (0.1 eq). Carrying out oil bath reaction at 60 ℃, detecting the reaction progress by TLC after 6-7 hours, and supplementing Pd (AcO) if the reaction is not completely carried out2The reaction was continued with Xantphos. After the reaction is completed, filtering with diatomite, removing solvent by rotary evaporation, and using EA/H2Extracting with O/saturated saline solution, drying with anhydrous magnesium sulfate, and performing column chromatography purification on the crude product obtained after rotary evaporation. The purified product is put into the next reaction.
Figure BDA0002115456660000133
The product obtained above (1.0eq) and 3, 5-dimethylisoxazoleboronic acid (1.5eq) were dissolved in a mixture of dioxane and water (dioxane: water 2:1) and stirred at room temperature. Sodium carbonate solid (3.0eq) and Pd (dppf) Cl were then added2(0.05eq), protected by nitrogen, and transferring to a 90 ℃ oil bath for heating reaction for 6-7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. And (3) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 5 f. The yield was 50%.
Example 7 compound 5 g: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -1- (4-methylbesyl) -N- (4- (trifluoromethylphenyl) -1H-indazol-3-amine
Figure BDA0002115456660000141
Compound 5g was prepared in 50% yield according to a similar method to example 6.
1HNMR(400MHz,Chloroform-d)7.53(d,J=8.4Hz,1H),7.45(d,J=8.5Hz,2H),7.34(d,J=8.5Hz,2H),7.11(d,J=8.0Hz,2H),7.05(d,J=8.1Hz,2H),6.99(d,J=1.1Hz,1H),6.88(dd,J=8.3,1.3Hz,1H),6.47(s,1H),5.40(s,2H),2.28(s,3H),2.24(s,3H),2.14(s,3H).
13CNMR(101MHz,Chloroform-d)165.50,158.64,145.65,142.44,140.59,137.73,133.59,129.48,129.46,127.43,126.56,126.52,121.02,119.94,116.70,115.71,115.29,110.03,53.03,21.09,11.55,10.76.
ESI-HRMS:477.1895,[M+1]+.
Example 8 compound 5 h: preparation of 4- ((6- (3, 5-dimethyllisozol-4-yl) -1- (4-methylbesyl) -1H-indazol-3-yl) amino) nitrile
Figure BDA0002115456660000142
Compound 5h was prepared in 50% yield according to a similar procedure as example 6.
1HNMR(400MHz,Chloroform-d)7.55(d,J=8.3Hz,1H),7.51–7.46(m,2H),7.34(dd,J=8.9,2.2Hz,2H),7.12–7.04(m,4H),7.00(s,1H),6.90(dd,J=8.3,1.3Hz,1H),5.41(s,2H),2.28(s,3H),2.24(s,3H),2.14(s,3H).
13CNMR(101MHz,Chloroform-d)165.53,158.60,146.53,141.72,140.51,137.82,133.61,133.43,129.64,129.49,127.41,121.29,119.86,119.74,116.63,115.71,115.59,110.10,102.26,53.10,21.10,11.56,10.76.
ESI-HRMS:434.1972,[M+1]+.
Example 9 compound 5 i: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -N- (4-methoxyphenyl) -1- (4-methyllbenzyl) -1H-indazol-3-amine
Figure BDA0002115456660000151
Compound 5i was prepared in 50% yield according to a similar method to example 6.
1HNMR(400MHz,Chloroform-d)7.50(d,J=8.3Hz,1H),7.34(d,J=8.5Hz,2H),7.17(d,J=7.9Hz,2H),7.11(d,J=7.9Hz,2H),6.98(s,1H),6.91–6.83(m,3H),6.12(s,1H),5.42(s,2H),3.80(s,3H),2.34(s,3H),2.31(s,3H),2.20(s,3H).
13CNMR(101MHz,Chloroform-d)165.38,158.69,154.33,140.82,137.47,136.06,133.98,129.37,129.11,127.38,120.33,120.21,118.78,116.83,115.21,114.56,109.77,55.67,52.79,21.09,11.54,10.76.
ESI-HRMS:439.2123,[M+1]+.
Example 10 compound 5 j: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -N- (4-fluorophenyl) -1- (4-methoxybenzyl) -1H-indazol-3-amine
Figure BDA0002115456660000152
Sodium hydride solid (1.5eq) was weighed into a parallel reaction tube, purged with nitrogen, and water and oxygen were removed. Dissolving the intermediate 2(1.0eq) in DMSO solvent, adding p-methoxybenzyl bromide (1.2eq), and performing nitrogen replacement for multiple times while maintaining anhydrous and oxygen-free environment. The reaction was carried out at room temperature overnight. And detecting the reaction degree by TLC, adding water for quenching after the reaction is completed, and separating out a solid. Filtering with a baffle funnel, and washing with water for multiple times. Drying to obtain intermediate 4 a-3. The yield was 85%.
Figure BDA0002115456660000161
The intermediate 4a-3(1.0eq) and cesium carbonate (2.0eq), para-fluoroaniline liquid (1.2eq) were dissolved in dry DMF solvent. N is a radical of2Replacement protection, and maintenance of anhydrous and oxygen-free environment. Adding catalyst Pd (AcO)2(0.1eq) and ligand Xantphos (0.1 eq). Carrying out oil bath reaction at 60 ℃, and detecting the reaction progress by TLC after 6-7 hours. After the reaction is completed, filtering by diatomite, removing solvent by rotary evaporation, and using EA/H2Extracting with O/saturated saline solution, drying with anhydrous magnesium sulfate, and performing column chromatography purification on the crude product obtained after rotary evaporation. The purified product is put into the next reaction.
Figure BDA0002115456660000162
The product obtained above (1.0eq) and 3, 5-dimethylisoxazoleboronic acid (1.5eq) were dissolved in a mixture of dioxane and water (dioxane: water 2:1) and stirred at room temperature. Sodium carbonate solid (3.0eq) and Pd (dppf) Cl were then added2(0.05eq), protected by nitrogen, and transferring to a 90 ℃ oil bath for heating reaction for 6-7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. And (4) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 5 j. The yield was 50%.
1HNMR(400MHz,Chloroform-d)7.55(dd,J=8.3,0.8Hz,1H),7.38–7.32(m,2H),7.24–7.20(m,2H),7.04–6.97(m,3H),6.90(dd,J=8.3,1.3Hz,1H),6.86–6.82(m,2H),5.41(s,2H),3.77(s,3H),2.36(s,3H),2.22(s,3H).
13CNMR(101MHz,Chloroform-d)165.43,159.26,158.66,143.94,140.64,138.73,129.30,128.95,128.75,120.51,120.05,117.81,116.79,115.78,115.56,115.32,114.12,109.84,55.30,52.57,11.58,10.80.
ESI-HRMS:443.1875,[M+1]+.
Example 11 compound 5 k: preparation of 6- (3, 5-dimethyllisoxazol-4-yl) -N- (3-isopropylphenyl) -1- (4-methoxybenzyl) -1H-indazol-3-amine
Figure BDA0002115456660000171
Compound 5k was prepared in 50% yield according to a similar method to example 10.
1HNMR(400MHz,Chloroform-d)7.59(dd,J=8.3,0.8Hz,1H),7.26(s,1H),7.25–7.19(m,4H),7.04(t,J=1.1Hz,1H),6.89(dd,J=8.3,1.3Hz,1H),6.87–6.79(m,3H),6.24(s,1H),5.42(s,2H),3.77(s,3H),2.89(p,J=6.9Hz,1H),2.36(s,3H),2.23(s,3H),1.27(s,3H),1.25(s,3H).
13CNMR(101MHz,Chloroform-d)165.40,159.24,158.69,150.07,143.84,142.60,140.62,129.13,129.07,129.02,128.85,120.39,120.32,118.78,116.84,115.67,114.57,114.09,113.89,109.76,55.29,52.54,34.21,23.97,11.59,10.81.
ESI-HRMS:467.2435,[M+1]+.
Example 12 compound 5 l: preparation of 5- (3,5-dimethylisoxazol-4-yl) -N- (4-fluorophenyl) -1- (tetrahydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000172
Compound 5l was prepared in 50% yield according to a similar method to example 2.
1HNMR(400MHz,Chloroform-d)7.55(dd,J=8.6,0.8Hz,1H),7.42–7.36(m,2H),7.32(t,J=1.1Hz,1H),7.26(d,J=1.8Hz,1H),7.24(d,J=1.5Hz,1H),7.03–6.95(m,2H),6.31(s,1H),5.63(dd,J=9.6,2.6Hz,1H),4.12–4.02(m,1H),3.81–3.74(m,1H),2.67–2.53(m,1H),2.36(s,3H),2.22(s,3H),2.10(dq,J=13.4,3.3Hz,1H),1.84–1.73(m,2H),1.70–1.55(m,2H).
13CNMR(101MHz,Chloroform-d)165.21,158.95,144.68,140.02,128.78,122.06,120.27,118.36,116.90,116.76,115.73,115.43,111.18,110.42,102.46,85.12,67.66,29.46,25.16,22.83,11.47,10.74.
ESI-HRMS:407.1871,[M+1]+.
Example 13 compound 5 m: preparation of 5- (3,5-dimethylisoxazol-4-yl) -N- (4-fluoro-2-methoxyphenyl) -1- (tetra-hydro-2H-pyran-2-yl) -1H-indazol-3-amine
Figure BDA0002115456660000181
Compound 5m was prepared in 50% yield according to a similar method to example 2.
1HNMR(400MHz,Chloroform-d)8.14–8.08(m,1H),7.55(dd,J=8.7,0.8Hz,1H),7.42(dd,J=1.5,0.8Hz,1H),7.27(d,J=1.7Hz,1H),7.24(d,J=1.6Hz,1H),6.77(s,1H),6.72–6.63(m,2H),5.64(dd,J=9.4,2.7Hz,1H),4.08(d,J=11.0Hz,1H),3.94(s,3H),3.74(dd,J=11.3,2.8Hz,1H),2.69–2.57(m,1H),2.39(s,3H),2.25(s,3H),2.21(dd,J=8.5,4.1Hz,1H),2.15–2.07(m,1H),1.84–1.72(m,2H),1.70–1.61(m,1H).
13CNMR(101MHz,Chloroform-d)165.18,158.98,144.36,139.74,128.82,121.94,119.80,117.19,116.82,115.94,115.86,110.35,106.52,106.31,98.71,98.43,85.09,67.53,56.00,53.42,29.40,25.20,22.81,11.48,10.75.
ESI-HRMS:437.1977,[M+1]+.
Example 14 compound 5 n: preparation of 5- (3, 5-dimethyllisozol-4-yl) -N- (4-fluoro-2-methoxyphenyl) -1- (4-methoxybenzyl) -1H-indazol-3-amine
Figure BDA0002115456660000182
Compound 5n was prepared in 50% yield according to a similar method to example 10.
1HNMR(400MHz,Chloroform-d)8.02(dd,J=9.6,5.9Hz,1H),7.42(dd,J=1.5,0.8Hz,1H),7.29(dd,J=8.7,0.8Hz,1H),7.24(d,J=2.1Hz,1H),7.18(dd,J=8.6,1.5Hz,1H),6.89–6.82(m,2H),6.75–6.62(m,3H),5.43(s,2H),3.94(s,3H),3.78(s,3H),2.38(s,3H),2.24(s,3H).
13CNMR(101MHz,Chloroform-d)165.14,159.20,143.92,129.14,128.73,128.67,127.93,121.15,120.06,116.76,116.35,115.82,115.73,114.08,109.65,106.55,106.34,98.85,98.58,56.00,55.27,52.33,11.52,10.78.
ESI-HRMS:473.1974,[M+1]+.
Example 15 compound 5 o: preparation of 4- (1- (4-fluorobenzyl) -5-methoxy-1H-indazol-6-yl) -3, 5-dimethylisoxazole
Figure BDA0002115456660000191
The 6-bromo-5 methoxy-1H-indazole starting material was purchased, 1.0eq and sodium hydride (1.5eq) were dissolved in DMSO solvent, added to a reaction tube and then replaced with nitrogen, appropriately heated with a hair dryer, and water and O in the reaction tube were removed2And stirred for 15 minutes. P-fluorobenzyl bromide (1.2eq) was then added and nitrogen substitution was again carried out several times, maintaining an anhydrous and oxygen-free environment. At room temperature overnight. And detecting the reaction degree by TLC, adding water for quenching after the reaction is completed, separating out solids, filtering by using a clapboard funnel, and washing by water for multiple times. The crude product obtained is purified by column chromatography and then put into the next reaction.
Figure BDA0002115456660000192
The product (1.0eq) and 3, 5-dimethylisoxazoleboronic acid (1.5eq) were dissolved in a mixture of dioxane and water (dioxane: water: 2:1) and stirred at room temperature. Then adding carbonic acidSodium solid (3.0eq) and Pd (dppf) Cl2(0.05eq), protected by nitrogen, and transferring to a 90 ℃ oil bath for heating reaction for 6-7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. And (4) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 5 o. The yield was 60%.
1HNMR(400MHz,Chloroform-d)7.96(s,1H),7.44(s,1H),7.25–7.19(m,2H),7.08–6.96(m,2H),6.70(s,1H),5.54(s,2H),3.79(s,3H),2.29(s,3H),2.14(s,3H).
13CNMR(101MHz,Chloroform-d)165.90,160.06,157.50,140.43,133.60,132.49,132.45,128.98,128.90,123.79,118.69,115.85,115.64,115.35,113.42,89.63,55.48,52.20,11.52,10.68.
ESI-HRMS:352.1446,[M+1]+.
EXAMPLE 16 preparation of Compound 11a 5- (1- (4-fluorobenzyl) -1H-indazol-6-yl) -1-methyl-3- (methylamino) pyridine-2(1H) -one
Figure BDA0002115456660000193
Dissolving compound 1(1.0eq) and sodium hydride solid (1.5eq) in DMSO solvent, adding into a reaction tube, placing in a parallel reaction tube, replacing with nitrogen, and extracting water vapor and O in the reaction tube2Then, p-fluorobenzyl bromide (1.2eq) was added thereto, and nitrogen substitution was performed again a plurality of times while maintaining an anhydrous and oxygen-free atmosphere. At room temperature overnight. And detecting the reaction degree by TLC, adding water for quenching after the reaction is completed, and separating out a solid. Filtering with a baffle funnel, washing with water for several times, and drying. The obtained product is put into the next reaction.
Figure BDA0002115456660000201
The product obtained above (1.0eq) and intermediate 9a (1.5eq), sodium carbonate solid (3.0eq) and Pd (dppf) Cl2(0.05eq) was dissolved in a mixed solvent of dioxane and water (dioxane: water ═ water)2:1), transferring to a 90 ℃ oil bath for heating and stirring under the protection of nitrogen, and continuing the reaction for 6-7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. The solvent was removed by distillation under reduced pressure, and the obtained crude product was purified by column chromatography to obtain the target compound 11 a. The yield was 70%.
1HNMR(400MHz,Chloroform-d)8.04(d,J=1.0Hz,1H),7.76(dd,J=8.4,0.8Hz,1H),7.32(q,J=1.0Hz,1H),7.27(d,J=1.4Hz,1H),7.25(d,J=1.4Hz,1H),7.22–7.17(m,2H),7.02–6.97(m,2H),6.84(d,J=2.2Hz,1H),6.36(d,J=2.3Hz,1H),5.60(s,2H),5.12(s,1H),3.64(s,3H),2.90(d,J=4.9Hz,3H).
13CNMR(101MHz,Chloroform-d)140.09,139.65,137.18,133.56,130.95,128.97,128.89,123.49,121.88,121.60,121.34,120.31,115.79,115.58,106.14,105.75.
ESI-HRMS:363.1608,[M+1]+.
EXAMPLE 17 preparation of Compound 11b 5- (1- (4-methoxybenzyl) -1H-indazol-6-yl) -1-methyl-3- (methylamino) pyridine-2(1H) -one
Figure BDA0002115456660000202
Compound 11b was prepared in 70% yield according to a similar method to example 16.
1HNMR(400MHz,Chloroform-d)8.03(d,J=0.9Hz,1H),7.75(dd,J=8.4,0.8Hz,1H),7.34(q,J=1.1Hz,1H),7.25(dd,J=8.4,1.5Hz,1H),7.21–7.16(m,2H),6.87–6.82(m,3H),6.39(d,J=2.2Hz,1H),5.57(s,2H),3.76(s,3H),3.65(s,3H),2.90(s,3H).
13CNMR(101MHz,Chloroform-d)159.25,157.40,140.05,139.63,136.93,133.26,128.87,128.66,123.49,121.86,121.49,121.35,120.10,114.16,106.37,105.76,55.27,52.55,37.62,29.87.
ESI-HRMS:375.1808,[M+1]+.
EXAMPLE 18 preparation of the compound 11c 1-methyl-3- (methylamino) -5- (1- (4-methylbenzyl) -1H-indazol-6-yl) pyridine-2(1H) -one
Figure BDA0002115456660000211
Compound 11c was prepared in 70% yield according to a similar method to example 16.
1HNMR(400MHz,Chloroform-d)8.04(d,J=0.9Hz,1H),7.75(dd,J=8.3,0.8Hz,1H),7.35(q,J=1.1Hz,1H),7.24(d,J=1.5Hz,1H),7.12(s,4H),6.85(d,J=2.3Hz,1H),6.39(d,J=2.3Hz,1H),5.60(s,2H),3.64(s,3H),2.90(s,3H),2.30(s,3H).
13CNMR(101MHz,Chloroform-d)157.40,140.14,139.62,137.54,136.94,133.78,133.28,129.43,127.24,123.48,121.85,121.49,121.36,120.10,106.36,105.77,52.81,37.61,29.87,21.09.
ESI-HRMS:359.1861,[M+1]+.
EXAMPLE 19 preparation of Compound 11d 5- (1- (3,5-dimethoxybenzyl) -1H-indazol-6-yl) -1-methyl-3- (methylimino) pyridine-2(1H) -one
Figure BDA0002115456660000212
Compound 11d was prepared in 70% yield according to a similar method to example 16.
1HNMR(400MHz,Chloroform-d)8.04(d,J=1.0Hz,1H),7.75(dd,J=8.4,0.8Hz,1H),7.36(q,J=1.1Hz,1H),7.24(d,J=1.4Hz,1H),6.85(d,J=2.2Hz,1H),6.39(d,J=2.2Hz,1H),6.35(s,3H),5.56(s,2H),3.72(s,6H),3.64(s,3H),2.90(s,3H).
13CNMR(101MHz,Chloroform-d)161.13,157.40,140.26,139.63,139.19,137.05,133.41,123.47,121.82,121.50,121.39,120.21,106.33,105.75,105.35,99.36,55.31,53.01,37.61,29.85.
ESI-HRMS:405.1919,[M+1]+.
EXAMPLE 20 preparation of Compound 11e 5- (1- (3,4-difluorobenzyl) -1H-indazol-6-yl) -1-methyl-3- (methylimino) pyridine-2(1H) -one
Figure BDA0002115456660000221
Compound 11e was prepared in 70% yield according to a similar procedure as example 16.
1HNMR(400MHz,Chloroform-d)8.06(d,J=0.9Hz,1H),7.77(dd,J=8.4,0.8Hz,1H),7.32(q,J=1.1Hz,1H),7.28(dd,J=8.4,1.4Hz,1H),7.10(dt,J=10.1,8.2Hz,1H),6.98(d,J=10.8Hz,2H),6.84(d,J=2.2Hz,1H),6.37(d,J=2.3Hz,1H),5.60–5.55(m,2H),3.64(s,3H),2.90(d,J=5.3Hz,3H).
13CNMR(101MHz,Chloroform-d)157.41,140.14,139.68,137.49,133.90,123.51,123.20,123.16,123.13,123.10,121.69,121.40,120.53,117.68,117.50,116.36,116.18,105.90,105.57,51.74,37.62,29.84.
ESI-HRMS:381.1528,[M+1]+.
EXAMPLE 21 preparation of Compound 11f 5- (1- (4-fluoro-3-nitrobenzyl) -1H-indazol-6-yl) -1-methyl-3- (methyl-amino) pyridine-2(1H) -one
Figure BDA0002115456660000222
Compound 11f was prepared in 70% yield according to a similar procedure as example 16.
1HNMR(400MHz,Chloroform-d)8.10–8.06(m,1H),7.96(dd,J=7.0,2.3Hz,1H),7.78(d,J=8.4Hz,1H),7.45(ddd,J=8.7,4.2,2.4Hz,1H),7.37(s,1H),7.31(dd,J=8.4,1.4Hz,1H),7.23(dd,J=10.4,8.6Hz,1H),6.87(d,J=2.3Hz,1H),6.39(d,J=2.2Hz,1H),5.66(s,2H),3.65(s,3H),2.90(d,J=3.4Hz,3H).
13CNMR(101MHz,CDCl3)157.39,140.14,139.72,137.85,134.41,134.27,134.18,134.01,124.80,123.56,121.85,121.61,121.44,120.80,119.11,118.90,105.60,105.46,51.17,37.65,29.85.
ESI-HRMS:408.1459,[M+1]+.
EXAMPLE 22 preparation of 11g of the compound 1-methyl-3- (methylamino) -5- (1- (4- (trifluoromethyl) benzyl) -1H-indazol-6-yl) pyridin-2(1H) -one
Figure BDA0002115456660000223
Compound 11g was obtained in 70% yield according to a similar manner to example 16.
1HNMR(400MHz,Chloroform-d)8.07(d,J=0.9Hz,1H),7.78(d,J=8.3Hz,1H),7.56(d,J=8.1Hz,2H),7.27(s,4H),6.83(d,J=2.2Hz,1H),6.36(d,J=2.2Hz,1H),5.69(s,2H),3.63(s,3H),2.88(s,3H).
13CNMR(101MHz,Chloroform-d)157.35,140.82,140.23,139.65,137.43,133.90,127.44,127.37,126.80,125.79,125.75,123.51,121.86,121.71,121.36,120.52,105.96,105.74,52.28,37.65,29.81.
ESI-HRMS:413.1579,[M+1]+.
EXAMPLE 23 preparation of Compound 11H 5- (1- (3-fluoro-4-methoxybenzyl) -1H-indazol-6-yl) -1-methyl-3- (methyl-amino) pyridine-2(1H) -one
Figure BDA0002115456660000231
Compound 11h was prepared according to a similar procedure to example 16. The yield was 70%.
1HNMR(400MHz,Chloroform-d)8.04(d,J=1.0Hz,1H),7.75(dd,J=8.4,0.8Hz,1H),7.23–7.14(m,3H),6.95(d,J=1.3Hz,1H),6.93–6.88(m,2H),6.77(s,1H),5.54(s,2H),3.87(s,3H),3.84(s,3H),3.67(s,3H).
13CNMR(101MHz,Chloroform-d)158.94,141.64,140.04,136.53,133.57,127.02,124.25,124.22,123.50,123.10,123.06,121.73,120.27,120.12,119.57,113.57,113.11,106.02,56.31,56.29,38.56,38.25.
ESI-HRMS:393.1727,[M+1]+.
EXAMPLE 24 preparation of the compound 11i 3- (ethyllamino) -5- (1- (4-fluorobenzyl) -1H-indazol-6-yl) -1-methylpyridin-2(1H) -one
Figure BDA0002115456660000232
Dissolving compound 1(1.0eq) and sodium hydride solid (1.5eq) in DMSO solvent, adding into a reaction tube, placing in a parallel reaction tube, replacing with nitrogen, and extracting water vapor and O in the reaction tube2Then, p-fluorobenzyl bromide (1.2eq) was added thereto, and nitrogen substitution was performed again a plurality of times while maintaining an anhydrous and oxygen-free atmosphere. At room temperature overnight. And detecting the reaction degree by TLC, adding water for quenching after the reaction is completed, and separating out a solid. Filtering with a baffle funnel, washing with water for several times, and drying. The obtained product is put into the next reaction.
The product obtained above (1.0eq) and intermediate 9b (1.5eq), sodium carbonate solid (3.0eq) and Pd (dppf) Cl2(0.05eq) is dissolved in a mixed solvent of dioxane and water (dioxane: water: 2:1), the mixture is transferred to a 90 ℃ oil bath for heating and stirring under the protection of nitrogen, and the reaction lasts for 6-7 hours. After TLC detection reaction is completed, filtering by using diatomite, and extracting by using water/ethyl acetate/saturated saline water; dried over anhydrous magnesium sulfate. And (3) distilling under reduced pressure to remove the solvent, and purifying the obtained crude product by column chromatography to obtain the target compound 11 i. The yield was 70%.
1HNMR(400MHz,Chloroform-d)8.04(d,J=1.0Hz,1H),7.75(dd,J=8.4,0.8Hz,1H),7.30(q,J=1.1Hz,1H),7.26–7.23(m,1H),7.22–7.17(m,2H),7.03–6.96(m,2H),6.83(d,J=2.2Hz,1H),6.37(d,J=2.2Hz,1H),5.59(s,2H),3.64(s,3H),3.18(dt,J=7.5,4.7Hz,2H),1.32(t,J=7.2Hz,3H).
13CNMR(101MHz,Chloroform-d)157.37,140.09,138.64,137.24,133.56,129.00,128.91,123.49,121.77,121.59,121.24,120.32,115.81,115.59,106.14,105.93,52.25,37.82,37.70,14.17.
ESI-HRMS:377.1778,[M+1]+.
EXAMPLE 25 preparation of the Compound 11j 5- (1- (3,5-dimethoxybenzyl) -1H-indazol-6-yl) -3- (ethyllamino) -1-methyl-pyridine-2 (1H) -one
Figure BDA0002115456660000241
Compound 11j was obtained in 70% yield according to a similar manner to example 24.
1HNMR(400MHz,Chloroform-d)8.04(d,J=1.0Hz,1H),7.74(dd,J=8.4,0.8Hz,1H),7.34(q,J=1.0Hz,1H),7.24(dd,J=8.4,1.4Hz,1H),6.83(d,J=2.2Hz,1H),6.40(d,J=2.3Hz,1H),6.36–6.35(m,2H),5.55(s,2H),3.71(s,6H),3.64(s,3H),3.17(d,J=5.6Hz,2H),1.32(t,J=7.2Hz,3H).
13CNMR(101MHz,Chloroform-d)161.13,140.25,139.18,138.60,137.08,133.41,123.46,121.81,121.49,121.27,120.21,106.32,106.04,105.39,105.34,99.37,55.31,53.01,37.81,37.68,14.19.
ESI-HRMS:419.2071,[M+1]+.
EXAMPLE 26 preparation of the compound 11k 5- (1- (cyclohexymethyl) -1H-indazol-6-yl) -1-methyl-3- (methylimino) pyri-din-2(1H) -one
Figure BDA0002115456660000242
Compound 11k was prepared in 70% yield according to a similar procedure as example 16.
1HNMR(400MHz,Chloroform-d)7.99(d,J=0.9Hz,1H),7.74(dd,J=8.4,0.8Hz,1H),7.38(q,J=1.1Hz,1H),7.24(dd,J=8.4,1.4Hz,1H),6.89(d,J=2.3Hz,1H),6.45(d,J=2.2Hz,1H),5.14(d,J=5.6Hz,1H),4.23(d,J=7.2Hz,2H),3.67(s,3H),2.93(d,J=5.3Hz,3H),1.75–1.63(m,4H),1.30–0.98(m,10H).
13CNMR(101MHz,Chloroform-d)157.38,140.44,139.64,136.71,132.63,122.94,122.21,121.42,121.33,119.94,106.25,106.02,55.15,38.81,30.98,30.93,29.90,29.69,26.28,25.71,25.66.
ESI-HRMS:351.2177,[M+1]+.
EXAMPLE 27 preparation of 11l of the compound ethyl2- (6- (1-methyl-5- (methylamino) -6-oxo-1,6-dihydropyridin-3-yl) -1H-indazol-1-yl) acetate
Figure BDA0002115456660000251
Compound 11l was prepared in 70% yield according to a similar method to example 16.
1HNMR(400MHz,Chloroform-d)7.99(d,J=0.9Hz,1H),7.74(dd,J=8.4,0.8Hz,1H),7.38(q,J=1.1Hz,1H),7.24(dd,J=8.4,1.4Hz,1H),6.89(d,J=2.3Hz,1H),6.45(d,J=2.2Hz,1H),5.14(d,J=5.6Hz,1H),4.23(d,J=7.2Hz,2H),3.67(s,3H),2.93(d,J=5.3Hz,3H),1.75–1.63(m,4H),1.30–0.98(m,10H).
13CNMR(101MHz,Chloroform-d)157.38,140.44,139.64,136.71,132.63,122.94,122.21,121.42,121.33,119.94,106.25,106.02,55.15,38.81,30.98,30.93,29.90,29.69,26.28,25.71,25.66.
ESI-HRMS:341.1601,[M+1]+.
EXAMPLE 28 preparation of the compound 11m 5- (1- (cyclopropropylethyl) -1H-indazol-6-yl) -1-methyl-3- (methylimino) pyridine-2(1H) -one
Figure BDA0002115456660000252
Compound 11m was prepared in 70% yield according to a similar method to example 16.
1HNMR(400MHz,Chloroform-d)8.00(d,J=1.0Hz,1H),7.75(dd,J=8.4,0.8Hz,1H),7.44(t,J=1.1Hz,1H),7.24(d,J=1.4Hz,1H),6.90(d,J=2.3Hz,1H),6.46(d,J=2.2Hz,1H),4.31(d,J=6.8Hz,2H),3.67(s,3H),2.93(s,3H),1.38(dddd,J=11.2,8.1,4.9,1.2Hz,1H),0.64–0.58(m,2H),0.45(dt,J=6.2,4.8Hz,2H).
13CNMR(101MHz,CDCl3)157.41,139.92,139.67,136.78,132.76,123.23,122.01,121.46,121.32,119.95,106.15,105.77,53.47,37.62,29.91,11.30,4.00.
ESI-HRMS:309.1719,[M+1]+.
The beneficial effects of the invention are demonstrated by biological experiments below.
Test example 1BRD4(1, 2) protein Activity test and cell proliferation inhibition test
1.1BRD4(1, 2) protein Activity assay
The compounds of the present invention were tested by Shanghai Ruizi chemical company for BRD4 protein. The compounds tested inhibited BRD4(1, 2) protein activity using the HTRF method. The test method is as follows:
the compounds were diluted 5-fold from 25. mu.M, 5 concentration gradients were set, and 5. mu.l of the prepared BRD4(1, 2) protein solution, GST-tagged substrate, and 5. mu.l of [ Lys (5,8,12,16) AC were added to each well plate]H4(1-21) -biotin, SA acceptor, anti-GST donor solution, 5. mu.l of blank buffer was added to the control group. Incubate at room temperature for 3 hours under closed and dark conditions. Detecting the fluorescence emission at 665nm and 620nm by using a tester compatible with HTRF, calculating the inhibition rate of the compounds at different concentrations on BRD4(1, 2) protein, fitting by using Excel software to obtain an inhibition value, and fitting and calculating by using GraphPad to obtain IC50The value is obtained. The experimental results are shown in tables 1 and 2.
1.2 cell proliferation inhibition assay
1.2.1 Experimental materials
Human acute myeloid leukemia cell MV4-11 and normal human stem cell LO2, thiazole blue tetrazolium bromide (MTT) were purchased from Sigma; DMEM medium and RPMI1640 medium, fetal bovine serum and trypsin were purchased from GibcoBRL; MV4-11 cell line was cultured in complete medium composed of 10% fetal bovine serum DMEM (FBS; Gibco, Auckland, N.Z.) and 1% antibiotics (100U/mL penicillin and 100U/mL streptomycin) at 37 ℃ in 5% CO2Culturing in an incubator under the conditions.
1.1.2 Experimental principles and methods
The MTT method is also called MTT colorimetric method, and is a method for detecting cell survival and growth. The detection principle is that succinate dehydrogenase in mitochondria of living cells can reduce exogenous MTT into water-insoluble blue-violet crystalline Formazan (Formazan) and deposit the Formazan in the cells, and dead cells do not have the function. Dimethyl sulfoxide (DMSO) can dissolve formazan in cells, and an enzyme linked immunosorbent assay detector is used for measuring the light absorption value of formazan at the wavelength of 540 or 720nm, so that the quantity of living cells can be indirectly reflected. Within a certain range of cell number, MTT crystals are formed in an amount proportional to the cell number. Therefore, the larger the number of living cells, the larger the amount of formazan precipitated, and the larger the absorbance measured by the microplate reader.
The stock solutions of the test compounds were diluted to different concentrations (3 duplicate wells) with a solution volume of 100. mu.L per well and added to 96-well plates. Collecting tumor cells in logarithmic growth phase, and culturing at 2.5-10 × 103The density of individual cells/well was seeded into 96-well plates at a cell suspension volume of 100 μ L per well and cells were treated with 1% DMSO medium and pure medium as negative controls. The well plate was then placed at 37 ℃ and 5% CO2Culturing in a cell culture incubator for 24 h. After formazan formation, the culture was stopped, 150. mu.L of LDMSO (adherent cells) was added to each well, and 50. mu.L of 20% SDS solution was directly added to suspension cells MV4-11, followed by shaking for 15-20 min. Detecting the absorbance (OD) of each well cell under the wavelength of 570nm by using a microplate reader570nm) And taking the average value to record the result. Cell proliferation inhibition rate (control group OD)570nmExperimental group OD570nm) Control group OD570nm100%. Half maximal Inhibitory Concentration (IC) was calculated for each compound by Graphpadprism550). Proliferation experiments were repeated three times for each tumor cell. The experimental results are shown in tables 1 and 2. The normal human stem cell LO2 was also subjected to a cytotoxicity test using the MTT method, and the results are shown in fig. 1.
TABLE 1 inhibitory Rate of the partial Compounds of the invention against BRD4(1, 2) and IC for inhibition of MV4-1150
Figure BDA0002115456660000271
TABLE 2 IC inhibition of BRD4(1, 2) and MV4-11 by a portion of the compounds of the invention50
Figure BDA0002115456660000272
Figure BDA0002115456660000281
Note: for IC50The value: a represents IC50Less than 2 μ M; b represents IC502-10 μ M; c represents IC50Greater than 10. mu.M.
Inhibition for 1 μ M dose: a represents more than 40%; b represents between 20% and 40%; c represents less than 20%.
JQ1 is a positive control drug, a known BRD4 inhibitor, and has the following structural formula:
Figure BDA0002115456660000282
test example 2 cell cycle arrest test
MV4-11 log phase cells were collected and appropriate cell suspension concentrations were made to be about 10X 10 per well5Individual cells (2 mL in volume) were seeded into 6-well plates; then placing the mixture in an incubator for 24 hours, and incubating the mixture at 37 ℃ under 5% CO2(ii) a The supernatant was added to 2mL of the media at different concentrations, and DMSO was used as a control. Then continuously placing the mixture at 37 ℃ and 5% CO2Incubate for 48 hours. Final cell treatment: (1) collecting cells in each hole, numbering the cells according to concentration in sequence, loading the cells into a flow tube, centrifuging the cells at the temperature of 4 ℃ at 2000r/min for 3 minutes, and removing supernatant; (2) resuspending the cells with 3ml PBS, centrifuging in the same manner, and discarding PBS; (3) fixing the cells with 75% ethanol for 30min, centrifuging in the same manner, and removing the supernatant; (4) then 3ml PBS is used for resuspending the cells, the centrifugation is carried out by the same method, and PBS is discarded; (5) adding 1mL of PI dye solution containing TritonX-100, uniformly mixing, and keeping out of the sun for 20 minutes at room temperature; and (4) detecting by using a flow cytometer. Cell cycle was examined using Flow Cytometry (FCM), PI staining. MV4-11 cells were treated with compound 11e for 48h before harvesting, fixed with 75% ethanol and stained with sodium Propyliodide (PI). Data were analyzed using FlowJo software. The results of the experiment are shown in FIG. 2.
As can be seen from fig. 2: in comparison with DMSO control, MV4-11 cell line was treated with a low concentration (0.1. mu.M) of compound 11eThe phases showed significant changes. When the concentration is increased to 0.5. mu.M and 2.5. mu.M, the number of cells in the G0/G1 phase is increased. The above experimental results show that the compound 11e of the present invention can block cells at G0/G1And exhibits concentration dependence.
Test example 3 immunoblotting analysis experiment
MV4-11 cells were treated with compound 11e at various concentrations for 72 hours, and the cells were harvested and lysed with a RicA lysate (China, Beijing, Byunnan) supplemented with cocktail and a phosphatase inhibitor, after 30 minutes on ice, 13000 rpm was applied, centrifuged at 4 ℃ for 15 minutes, and the supernatant was collected. Protein concentration determination was performed using the BCA method. Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) was performed to ensure consistent total protein loading for each sample, and then proteins were transferred to nitrocellulose (NC membrane) at 100V for 2 hours. NC membranes were blocked with 5% skim milk powder in TBS/T for 1.5 hours at room temperature, followed by 4 degrees overnight incubation of primary antibody. After incubation of horseradish peroxidase (HRP) conjugated secondary antibodies, the membrane was reacted with a chemiluminescent substrate and then exposed. The quality of western blot experiments was assessed using ImageJ to combine the results of three independent experiments. The results of the experiment are shown in FIG. 3.
As can be seen in FIG. 3, the expression of c-Myc decreased in a dose-dependent manner with Compound 11e, indicating that Compound 11e of the present invention is able to affect transcription and expression of downstream genes.

Claims (12)

1. A compound of formula I or a pharmaceutically acceptable salt thereof:
Figure FDA0002115456650000011
R1、R2、R3、R4among which only one is selected from
Figure FDA0002115456650000012
The remaining three are independently selected from-H, halogen, alkyl, alkoxy, -NH2、-NO2or-CN, wherein R10、R11、R12、R13、R14Independently selected from-H or alkyl;
R5is selected from-H or-X-R7Wherein X is-CH2-or is absent, R7Selected from aryl, cycloalkyl, acyl or alkyl;
R6is selected from-H or-NH-R8Wherein R is8Is an aryl group.
2. The compound of claim 1, wherein: r1、R2、R3、R4Among which only one is selected from
Figure FDA0002115456650000013
The other three are independently selected from-H, halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen substituted C1-C6 alkyl, -NH2、-NO2or-CN, wherein R10、R11、R12、R13、R14Independently selected from-H or unsubstituted C1-C6 alkyl;
preferably, R1、R2、R3、R4Among which only one is selected from
Figure FDA0002115456650000014
The other three are independently selected from-H, halogen, -OCH3or-CH3Wherein R is10、R11、R12、R13、R14Independently selected from-H, -CH3or-CH2CH3
Preferably, R1、R2、R3、R4Middle R2Or R3Is composed of
Figure FDA0002115456650000021
Or R3Is composed of
Figure FDA0002115456650000022
The other three are independently selected from-H or-OCH3Wherein R is10、R11、R14Are all-CH3,R12is-H, R13is-CH3or-CH2CH3
Most preferably, R3Is composed of
Figure FDA0002115456650000023
R1、R2、R4Independently selected from-H or-OCH3
3. The compound of claim 1, wherein: r5Is selected from-H or-X-R7Wherein X is-CH2-or is absent, R7Selected from 5-to 14-membered aryl, 3-to 6-membered cycloalkyl,
Figure FDA0002115456650000024
Or C1-C6 alkyl, wherein R9Is C1-C6 alkoxy;
preferably, R7Selected from 6-membered aryl, 3-to 6-membered cycloalkyl,
Figure FDA0002115456650000025
Or C1-C6 alkyl, wherein R9Is C1-C6 alkoxy;
preferably, R7Selected from substituted or unsubstituted phenyl, 3-6 membered cycloalkyl or
Figure FDA0002115456650000026
Wherein R is9is-OCH2CH3
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, alkyl, alkoxy, -NO2
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen-substituted C1-C6 alkyl, -NO2
Preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: -F, -CH3、-CF3、-OCH3、-NO2
Preferably, the 3-6 membered cycloalkyl group is selected from
Figure FDA0002115456650000031
Most preferably, R5is-X-R7Wherein X is-CH2-,R7Is selected from
Figure FDA0002115456650000032
Figure FDA0002115456650000033
4. The compound of claim 1, wherein: r6Is selected from-H or-NH-R8Wherein R is8Is a 5-to 14-membered aryl group;
preferably, R8Is a 6-membered aryl group;
preferably, R8Is substituted or unsubstituted phenyl;
preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, alkyl, alkoxy or-CN;
preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: halogen, unsubstituted C1-C6 alkyl, halogen substituted C1-C6 alkyl, unsubstituted C1-C6 alkoxy, halogen substituted C1-C6 alkoxy or-CN;
preferably, the substituted phenyl group contains at least one substituent selected from the group consisting of: -F, -Cl, -OCH3、-CF3
Figure FDA0002115456650000034
or-CN;
most preferably, R6is-H.
5. The compound according to any one of claims 1 to 4, wherein: the compound is selected from:
Figure FDA0002115456650000035
Figure FDA0002115456650000041
6. a process for the preparation of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, characterized in that: the method comprises the following steps:
when R is1、R2、R3、R4Among them and only one is
Figure FDA0002115456650000042
In time, the method one is adopted: carrying out coupling reaction on the raw materials SM-1 and SM-2 to obtain a target product:
Figure FDA0002115456650000043
wherein R is1a、R2a、R3a、R4aOne and only one is halogen, and the other three are R at the same position of benzene ring in the product1、R2、R3、R4The groups are the same; a is
Figure FDA0002115456650000044
R15、R16Independently selected from-H or alkyl, or joined to form cycloalkyl;
preferably, R15、R16Independently selected from-H or unsubstituted C1-C6 alkyl, or joined to form a 5-membered cycloalkyl;
most preferably, the halogen is-Br and A is
Figure FDA0002115456650000051
Or, when R is1、R2、R3、R4Among them and only one is
Figure FDA0002115456650000052
Then, the method II is adopted: carrying out coupling reaction on the raw materials SM-3 and SM-4 to obtain a target product:
Figure FDA0002115456650000053
wherein R is1b、R2b、R3b、R4bOne and only one is
Figure FDA0002115456650000054
The other three R are at the same position of benzene ring in the product1、R2、R3、R4The radicals R are the same17、R18Independently selected from-H or alkyl, or joined to form cycloalkyl, Y is halogen;
preferably, R17、R18Independently selected from-H or unsubstituted C1-C6 alkyl, or joined to form a 5-membered cycloalkyl;
most preferably, R1b、R2b、R3b、R4bOne and only one is
Figure FDA0002115456650000055
Y is-Br.
7. The method of claim 6, wherein: at least one of the following is satisfied:
in the first method and the second method, a palladium catalyst and alkali are added into a reaction system;
preferablyThe palladium catalyst is Pd (dppf) Cl2
Preferably, the base is sodium carbonate;
preferably, SM-2: SM-1: pd (dppf) Cl2: the molar ratio of sodium carbonate is 1: 1.5: 0.05: 3;
preferably, SM-3: SM-4: pd (dppf) Cl2: the molar ratio of sodium carbonate is 1: 1.5: 0.05: 3;
the reaction solvent is a mixed solvent of dioxane and water;
preferably, the ratio of dioxane: the volume ratio of water is 2: 1;
carrying out reaction under a protective atmosphere;
the reaction temperature was 90 ℃.
8. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of BRD 4.
9. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment and/or prophylaxis of cancer.
10. Use according to claim 9, characterized in that: the medicament is a medicament for treating and/or preventing leukemia; preferably, the leukemia is acute myeloid leukemia.
11. The pharmaceutical composition for preventing and treating the cancer is characterized in that: the compound or the pharmaceutically acceptable salt thereof according to any one of claims 1 to 5 is used as an active ingredient, and an acceptable auxiliary material or an auxiliary ingredient is added to prepare the preparation.
12. The pharmaceutical composition of claim 11, wherein: the preparation is an agent for treating and/or preventing leukemia; preferably, the leukemia is acute myeloid leukemia.
CN201910589317.6A 2019-07-02 2019-07-02 Indazole compound with anticancer effect and preparation method and application thereof Active CN112174945B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910589317.6A CN112174945B (en) 2019-07-02 2019-07-02 Indazole compound with anticancer effect and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910589317.6A CN112174945B (en) 2019-07-02 2019-07-02 Indazole compound with anticancer effect and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112174945A true CN112174945A (en) 2021-01-05
CN112174945B CN112174945B (en) 2022-06-07

Family

ID=73915042

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910589317.6A Active CN112174945B (en) 2019-07-02 2019-07-02 Indazole compound with anticancer effect and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112174945B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113402515A (en) * 2021-05-12 2021-09-17 四川大学华西医院 Indole compound and preparation method and application thereof
CN114014842A (en) * 2021-11-04 2022-02-08 中国药科大学 Heterocyclic ketone compound and composition thereof as well as preparation method and application thereof
CN116535389A (en) * 2023-04-28 2023-08-04 四川大学 6-pyridine-3-quinoxaline urea derivative and application thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105073747A (en) * 2013-03-15 2015-11-18 普莱希科公司 Heterocyclic compounds and uses thereof
CN105407888A (en) * 2013-06-21 2016-03-16 齐尼思表观遗传学公司 Novel bicyclic bromodomain inhibitors
US20160075712A1 (en) * 2014-09-15 2016-03-17 Plexxikon Inc. Heterocyclic compounds and uses thereof
CN105492439A (en) * 2013-06-21 2016-04-13 齐尼思表观遗传学公司 Novel substituted bicyclic compounds as bromodomain inhibitors
WO2016097870A1 (en) * 2014-12-17 2016-06-23 Zenith Epigenetics Corp. Substituted bicyclic compounds as bromodomain inhibitors
CN105828820A (en) * 2013-10-18 2016-08-03 赛尔基因昆蒂赛尔研究公司 Bromodomain inhibitors
CN107635558A (en) * 2015-04-15 2018-01-26 赛尔基因昆蒂赛尔研究公司 Bu Luomo domain inhibitor
CN108137539A (en) * 2015-08-12 2018-06-08 尼奥迈德研究所 Substituted benzimidazole, their preparation and their purposes as drug

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105073747A (en) * 2013-03-15 2015-11-18 普莱希科公司 Heterocyclic compounds and uses thereof
CN105407888A (en) * 2013-06-21 2016-03-16 齐尼思表观遗传学公司 Novel bicyclic bromodomain inhibitors
CN105492439A (en) * 2013-06-21 2016-04-13 齐尼思表观遗传学公司 Novel substituted bicyclic compounds as bromodomain inhibitors
CN109939113A (en) * 2013-06-21 2019-06-28 齐尼思表观遗传学有限公司 Bicyclic bromine structural domain inhibitor
CN105828820A (en) * 2013-10-18 2016-08-03 赛尔基因昆蒂赛尔研究公司 Bromodomain inhibitors
US20160075712A1 (en) * 2014-09-15 2016-03-17 Plexxikon Inc. Heterocyclic compounds and uses thereof
WO2016097870A1 (en) * 2014-12-17 2016-06-23 Zenith Epigenetics Corp. Substituted bicyclic compounds as bromodomain inhibitors
CN107635558A (en) * 2015-04-15 2018-01-26 赛尔基因昆蒂赛尔研究公司 Bu Luomo domain inhibitor
CN108137539A (en) * 2015-08-12 2018-06-08 尼奥迈德研究所 Substituted benzimidazole, their preparation and their purposes as drug

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113402515A (en) * 2021-05-12 2021-09-17 四川大学华西医院 Indole compound and preparation method and application thereof
CN113402515B (en) * 2021-05-12 2022-05-27 四川大学华西医院 Indole compound and preparation method and application thereof
CN114014842A (en) * 2021-11-04 2022-02-08 中国药科大学 Heterocyclic ketone compound and composition thereof as well as preparation method and application thereof
CN114014842B (en) * 2021-11-04 2024-03-01 中国药科大学 Heterocyclic ketone compound and composition thereof, and preparation method and application thereof
CN116535389A (en) * 2023-04-28 2023-08-04 四川大学 6-pyridine-3-quinoxaline urea derivative and application thereof
CN116535389B (en) * 2023-04-28 2024-05-03 四川大学 6-Pyridine-3-quinoxaline urea derivative and application thereof

Also Published As

Publication number Publication date
CN112174945B (en) 2022-06-07

Similar Documents

Publication Publication Date Title
CN112174945B (en) Indazole compound with anticancer effect and preparation method and application thereof
JP6884707B2 (en) Isooxazolyl-substituted imidazopyridines
CN105849099A (en) Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
EA005994B1 (en) Glucopyranosyloxybenzylbenzene derivatives and medical compositions containing the same
JPH08504834A (en) Further added pyrimidine derivatives and processes for their preparation
CN106573906A (en) Piperidine-dione derivatives
JP2021176847A (en) Substituted five-membered and six-membered heterocyclic compound, its preparation method, combination with medicine and its usage
CN107531683B (en) USP7 inhibitor compounds and methods of use
CN110036002A (en) Medical compounds
CN108948002A (en) Five yuan and hexa-atomic nitrogen heteroaromatic rings class compound, preparation method, Pharmaceutical composition and its application
CN109476642A (en) Compound and its purposes for reducing uric acid level
CN108003155A (en) 3,4 dihydro -1H [1,8] naphthyridones of the homopiperidinyl substitution of antibacterial
AU2019249560A1 (en) Heteroaryl-substituted pyrazole compound and medicinal use thereof
WO2017189893A1 (en) 1,5-disubstituted 1,2,3-triazoles are inhibitors of rac/cdc42 gtpases
CN110914253A (en) Isoindolone-imide ring-1, 3-diketone-2-alkene compounds, compositions and uses thereof
CN107501279B (en) Furoquinoline cyclohexadione compounds and its medical usage
EP4342466A1 (en) Therapeutic or prophylactic method for diabetes using combination medicine
CN104603133B (en) For treating cancer and immunosuppressant combination treatment
WO2023125928A1 (en) Menin inhibitor and use thereof
CN109641909A (en) The mechanism target and its treatment use of rapamycin signal pathway inhibitor
CN111108083B (en) Use of aminomethylene cyclohexane 1, 3-dione compounds
WO2023276828A1 (en) Pharmaceutical composition for preventing and/or treating kidney injury, and autophagy activator
CN115477608A (en) Tubulin inhibitor and preparation method and application thereof
CN111247137A (en) Pyrimidine compound, preparation method and medical application thereof
CN106573927A (en) Oxepan-2-yl-pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use

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