CN114539223A - Aryl-containing aza-heptacyclic compound and preparation method and application thereof - Google Patents

Aryl-containing aza-heptacyclic compound and preparation method and application thereof Download PDF

Info

Publication number
CN114539223A
CN114539223A CN202210194719.8A CN202210194719A CN114539223A CN 114539223 A CN114539223 A CN 114539223A CN 202210194719 A CN202210194719 A CN 202210194719A CN 114539223 A CN114539223 A CN 114539223A
Authority
CN
China
Prior art keywords
compound
synthesis
reaction
radical
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202210194719.8A
Other languages
Chinese (zh)
Other versions
CN114539223B (en
Inventor
吴筱星
华德翔
陈晓禹
舒成霞
李文强
罗光美
杨可欣
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 CN202210194719.8A priority Critical patent/CN114539223B/en
Publication of CN114539223A publication Critical patent/CN114539223A/en
Application granted granted Critical
Publication of CN114539223B publication Critical patent/CN114539223B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Abstract

The invention belongs to the field of pharmaceutical chemistry, and particularly relates to an aryl-containing aza-heptacyclic compound and a preparation method and application thereof. An aryloazepine heptacyclic compound having the general formula I and pharmaceutically acceptable salts, enantiomers, diastereomers, tautomers, solvates, polymorphs, or prodrugs thereof; the invention prepares a brand new compound of an aryl aza seven-membered ring based on SHP099 as a lead compound to solve the problem of SH at presentThe P2 inhibitor has single structural framework and the like; the invention has the important significance of providing a plurality of modification sites and providing a foundation for later structural modification. Meanwhile, the embodiment of the invention proves that the compound has allosteric inhibition effect on SHP2 phosphatase and provides framework support for the subsequent development of SHP2 phosphatase inhibitors.

Description

Aryl-containing aza-heptacyclic compound and preparation method and application thereof
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to an aryl-containing aza-heptacyclic compound and a preparation method and application thereof.
Background
Src homology 2-containing protein tyrosine phosphatase (SHP 2) belongs to the PTPs protein family and plays a key role in regulating the processes of cell growth, proliferation, differentiation and apoptosis. In human cells, SHP2 acts as an important junction linking multiple oncogenic signaling pathways, and its mutation and aberrant expression can cause a variety of diseases including developmental disorders, leukemia, and solid tumors. In recent years, SHP2 phosphatase has attracted considerable attention because targeting SHP2 is considered an effective anti-cancer strategy. On one hand, the SHP2 is used as an upstream core regulatory factor of a RAS/Raf/MAPK signal pathway, and the inhibition of SHP2 can block the activation of the RAS/Raf/MAPK signal pathway, thereby inhibiting the growth of tumor cells; on the other hand, SHP2 is an important synergistic factor for regulating tumor immunity, and inhibition of SHP2 can activate T cell immune function and specifically kill tumor cells.
In the research of the SHP2 inhibitor, the development of an inhibitor targeting the SHP2 catalytic domain was mainly focused at the beginning, but the inhibitor has the problems of low bioavailability, poor selectivity and the like, and subsequent clinical research is difficult to carry out. In view of this, researchers began to turn their eyes to the development of SHP2 allosteric inhibitors. In 2016, the first SHP2 allosteric inhibitor SHP099 was pioneered by nova corporation, and this finding greatly pushed the development of SHP2 inhibitors. Compared with a SHP2 inhibitor targeting a catalytic domain, the SHP099 has better activity, higher selectivity and better drugability. Currently, ten allosteric SHP2 inhibitors have entered clinical trials. However, no SHP2 small molecule inhibitor is yet available in the market. Therefore, the development of a novel SHP2 small-molecule inhibitor with good anti-tumor activity is of great significance.
Disclosure of Invention
One of the technical problems to be solved by the invention is to provide a series of aryl-containing aza-heptacyclic compounds for treating various related tumor diseases by inhibiting SHP2 protein and regulating KRAS-MEK-ERK signaling pathway.
The scheme for solving the technical problems is as follows:
an aromatic aza heptacyclic compound shown as general formula I, and pharmaceutically acceptable salt, enantiomer, non-isomer, tautomer, solvate, polymorph or prodrug thereof
Figure BDA0003526744790000021
Ring A is
Figure BDA0003526744790000022
X is N or CR4
R1、R2And R4Independently hydrogen, halogen, amino;
l is a bond, C1-C2Carbon chain of (3), O or S;
when n is 1, m is 1;
when n is 2, m is 0;
R3is hydrogen, deuterium, halogen, amino, hydroxyl, nitro, cyano, amido, carboxyl, sulfonyl, -C (O) NR5R6、-C(O)R5、-C(O)OR5、-NR5C(O)R6、-NR5C(O)NR6R7、-NR5C(O)OR6、-NR5R6、-OC(O)R5、-OC(O)NR5R6、-SR5、-S(O)2R5、-S(O)R5、-S(O)2NR5R6、C1-C6Alkyl radical, C1-C6Haloalkyl, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C3-C7Epoxy group, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10Heteroaromatic rings, mono-heterocycles, spirocycles, bridged rings, and fused rings; wherein R is5、R6And R7Independently hydrogen, deuterium, halogen, C1-C7Alkyl radical, C1-C7Haloalkyl, C1-C7Alkoxy radical, C1-C7Haloalkoxy, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10A heteroaromatic ring;
an aromatic aza heptacyclic compound shown as general formula I, and pharmaceutically acceptable salt, enantiomer, non-isomer, tautomer, solvate, polymorph or prodrug thereof
Figure BDA0003526744790000023
Ring A is
Figure BDA0003526744790000024
X is N or CR4
R1、R2And R4Independently hydrogen, halogen, amino;
l is a bond, C1-C2Carbon chain of (3), O or S;
when n is 1, m is 1;
when n is 2, m is 0;
R3is hydrogen, deuterium, halogen, amino, hydroxyl, nitro, cyano, amido, carboxyl, sulfonyl, -C (O) NR5R6、-C(O)R5、-C(O)OR5、-NR5C(O)R6、-NR5R6、-OC(O)R5、-SR5、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10A heteroaromatic ring; wherein R is5And R6Independently hydrogen, deuterium, halogen, C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl;
the aromatic aza seven-membered ring compound is any one of the following structural formulas:
Figure BDA0003526744790000031
the pharmaceutical composition is characterized by comprising the aryl-aza-heptacyclic compound and pharmaceutically acceptable auxiliary materials.
The pharmaceutical composition is characterized in that the pharmaceutical composition is prepared into tablets, capsules, injection or freeze-dried powder.
The aryl aza seven-ring compound and the pharmaceutical composition are applied to the preparation of antitumor drugs, prodrugs of the antitumor drugs or intermediates of the antitumor drugs.
Advantageous effects
The aromatic nitrogen-containing aza-seven-membered ring compound with a brand-new framework is prepared based on SHP099 as a lead compound, has a novel structure, has enzyme activity remarkably superior to that of SHP099, and provides support for subsequent development of antitumor drugs.
Detailed Description
Intermediates
Figure BDA0003526744790000041
(A) The synthesis of (2):
Figure BDA0003526744790000042
the method comprises the following steps: synthesis of 2-fluoro-3-chloro-4-iodopyridine (A-2):
n-butyllithium (76mL,1.25eq.) was slowly added dropwise to a solution of A-1(20.00g,152.6mmol) in THF (150mL) at-78 ℃. Slowly dropwise adding I after reacting for 1h2Of THF (60 mL). The reaction was monitored after 30 min. After monitoring the completion of the reaction, saturated Na was added dropwise2SO3Quenching the aqueous solution, concentrating to remove THF, extracting with ethyl acetate, drying over anhydrous sodium sulfate, concentrating, and separating by column chromatography to obtain compound A-2(12.21g, yield 32%).1H NMR(300MHz,CDCl3):δ7.77(dd,J=5.2,0.9Hz,1H),7.67(d,J=5.2Hz,1H).
Step two: synthesis of 3-chloro-4-iodo-2-pyridylamine (A-3)
Reacting NH3·H2O (30mL) was added slowly dropwise to a solution of A-2(12.2g,48.6mmol) in DMSO (40 mL). After the addition, the reaction was carried out at 60 ℃ for 48 hours. After completion of the reaction was monitored, the reaction system was poured into water (200mL), stirred for 30min, and dried by suction filtration to obtain Compound A-3(11.0g, yield 91%).1H NMR(300MHz,CDCl3):δ7.57(d,J=5.2Hz,1H),7.12(d,J=5.2Hz,1H),5.05(s,2H).
Step three: synthesis of ethyl 3- ((2-amino-3-chloropyridin-4-yl) thio) propionate (A-4)
Compound A-3(5.50g,22.1mmol,1.0eq.), Pd (OAc)2(248.0mg,5 mol%) and XantPhos (767.2mg,6 mol%) were placed in a 250mL single neck flask, the nitrogen was replaced by applying a vacuum to the system, ethyl mercaptopropionate (3.26g,24.3mmol,1.1eq.), 1, 4-dioxane (45mL) and DIPEA (7.7mL,44.2mmol,2.0eq.) were added and reacted overnight in an oil bath at 100 ℃ with monitoring that the conversion of the starting material was complete. Filtering with diatomaceous earth, concentrating the filtrate, adding 30mL ethyl acetate and 10mL water, extracting and separating, washing the organic phase with saturated saline 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound A-4(4.10g, yield 81%).1H NMR(300MHz,CDCl3):δ7.87(d,J=5.5Hz,1H),6.53(d,J=5.5Hz,1H),4.89(s,2H),4.19(q,J=7.2Hz,2H),3.23(t,J=7.5Hz,2H),2.72(t,J=7.5Hz,2H),1.28(t,J=7.2Hz,3H).
Step four: synthesis of sodium 3-amino-2-chlorothiophenylate (A-5)
An ethanol solution of sodium ethoxide (5.88g,17.3mmol,1.1eq., ω ═ 20%) was slowly added dropwise to a solution of a-4(4.10g,15.7mmol) in THF (20 mL). The reaction was carried out at room temperature for 2 h. After the reaction was monitored to be complete, n-hexane (100mL) was added, and the mixture was slurried, filtered, and the solid was dried to give Compound A-5(3.20g, crude).
Step five: synthesis of 3-amino-2-chlorobenzenethiol (A)
Compound a-5(1.60g, crude) was dissolved in water (10mL) and hydrochloric acid (3M) was added dropwise to pH 6. The reaction solution was extracted with ethyl acetate, and the organic phase was washed with saturated sodium chloride solution 3 times, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography to give intermediate a (1.05g, yield in two steps 75%).1H NMR(300MHz,DMSO-d6):δ11.46(s,1H),7.09(d,J=6.9Hz,1H),6.76(s,2H),6.67(d,J=6.9Hz,1H).
Intermediates
Figure BDA0003526744790000051
(B) The synthesis of (2):
Figure BDA0003526744790000052
the method comprises the following steps: synthesis of ethyl 3- ((5-chloropyrazin-2-yl) thio) propionate (B-2) ethyl 3-mercaptopropionate (4.45mL,1.05eq.) was added slowly dropwise to 2, 5-dichloropyrazine (5.00g,33.6mmol) and K2CO3(4.64g,1.0eq.) in DMF (42 mL). The reaction was carried out at room temperature for 4 hours. After completion of the reaction was monitored, 200mL of ethyl acetate was added to dilute the reaction mixture, the mixture was washed with saturated brine 5 times, and the organic phase was dried over anhydrous sodium sulfate, concentrated and subjected to column chromatography to obtain Compound B-2(7.78g, yield 94%).1H NMR(300MHz,CDCl3):δ8.39(d,J=1.5Hz,1H),8.22(d,J=1.5Hz,1H),5.17(q,J=7.2Hz,2H),3.42(t,J=7.0Hz,2H),2.75(t,J=7.0Hz,2H),1.27(t,J=7.2Hz,3H).
Step two: synthesis of sodium 5-chloropyrazine-2-thiolate (B-3)
An ethanol solution of sodium ethoxide (11.22g,33.0mmol,1.1eq., ω ═ 20%) was slowly added dropwise to a solution of B-3(7.38g,30.0mmol) in THF (100 mL). The reaction was carried out at room temperature for 2 h. After the reaction was monitored to be complete, n-hexane (100mL) was added, and the mixture was slurried, filtered, and the solid was dried to give Compound B-3(5.13g, crude).
Step three: synthesis of 3-chloro-4- ((5-chloropyrazin-2-yl) thio) pyridin-2-amine (B)
Compound A-3(5.50g,22.1mmol,1.0eq.), compound B-3(4.47g,26.5mmol,1.2eq.), and compound Pd2dba3(303.5mg,1.5 mol%) and Xantphos (383.6mg,3 mol%) were placed in a 100mL single neck flask, the nitrogen was replaced by applying a vacuum to the system, 1, 4-dioxane (45mL) and DIPEA (7.7mL,44.2mmol,2.0eq.) were added and the reaction was allowed to proceed overnight in a 100 ℃ oil bath and monitored for complete conversion of the starting material. Filtering with celite, concentrating the filtrate, adding 50mL ethyl acetate and 10mL water, extracting and separating, washing the organic phase with saturated brine for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain intermediate compound B (4.4g, yield 73%).1H NMR(300MHz,CDCl3):δ8.52(d,J=1.5Hz,1H),8.36(d,J=1.5Hz,1H),7.90(d,J=5.2Hz,1H),6.58(d,J=5.2Hz,1H),5.04(brs,2H).
Example 1:
Figure BDA0003526744790000061
(1) the synthesis of (2):
Figure BDA0003526744790000062
the method comprises the following steps: synthesis of 3- (benzyl (tert-butoxycarbonyl) amino) propionic acid (1-2):
ethyl acrylate (5.34mL,50.0mmol,1.0eq.) was slowly added dropwise to a solution of 1-1(6.05g,50.0mmol) in ethanol (100 mL). The reaction was carried out at room temperature for 12 hours, after completion of the reaction was monitored, the reaction mixture was concentrated, and DCM (100mL), triethylamine (13.8mL,100.0mmol,2.0eq.) and Boc were added to the mixture in this order2O (12.01g,55.0mmol,1.1 eq.). Reacting for half an hour at room temperature, monitoring the reaction completion, concentrating, and adding THF/H into the mixture in sequence2O/EtOH (25/25/25mL) mixed solution and lithium hydroxide (3.62g, 150.0mmol,3.0 eq.). Reacting at room temperature for 6h, monitoring the reaction completion, concentrating, adding 30mL of water and 30mL of ethyl acetateThe mixture was extracted, separated, and the aqueous phase was adjusted to pH 3 with hydrochloric acid (3M), and then extracted twice with ethyl acetate. The organic phase was washed with an appropriate amount of saturated brine for 3 times, dried over anhydrous sodium sulfate, and concentrated to give compound 1-2(13.60g, crude).
Step two: synthesis of tert-butyl 5-oxo-1, 3,4, 5-tetrahydro-2H-benzo [ c ] aza-2-carboxylate (1-3):
thionyl chloride (10.8mL,150.0mmol,3.0eq.) was slowly added dropwise to a 1-2 solution of anhydrous DCM (100mL) at 0 ℃ for 2h at room temperature, after completion of the reaction was monitored, concentrated and pumped with an oil pump for half an hour. The mixture was dissolved in anhydrous DCM (100mL), and aluminum trichloride (19.85g,150.0mml,3.0eq.) was added at 0 ℃ to react overnight at room temperature. The reaction was poured into ice aqueous sodium hydroxide (6M,100mL), filtered, the filter residue was washed twice with DCM (20mL), the liquid separated and the organic phase washed 3 times with saturated brine. To the organic phase was added triethylamine (7.6mL,55.0mmol,1.1eq.) and Boc sequentially2O (10.92g,50.0mmol,1.0 eq.). Reacting for half an hour at room temperature, washing the reaction solution with an appropriate amount of saturated sodium chloride solution for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound 1-3(3.10g, yield 23% in six steps).1H NMR(300MHz,CDCl3):δ7.89(dd,J=7.8,1.5Hz,1H),7.49-7.44(m,1H),7.42-7.29(m,2H),4.70(d,J=16.0Hz,2H),3.79-3.66(m,2H),3.08-2.98(m,2H),1.39(s,9H).
Step three: synthesis of tert-butyl (S) -5- (((R) -tert-butylsulfinyl) amino) -1,3,4, 5-tetrahydro-2H-benzo [ c ] aza-2-carboxylate (1-4)
(R) -2-methylpropane-2-sulfinamide (2.86g,23.6mmol,2.0eq.) and tetraethyltitanate (5.38g,23.6mmol,2.0eq.) were added to a solution of 1-3(3.10g,11.8mmol) in THF (20mL) and reacted at 75 ℃ overnight. After monitoring the reaction completion, LiBH (iBu) was slowly added dropwise at-50 ℃3The reaction solution (23.6mL,1M,2.0eq.) was allowed to react at room temperature for 2 h. After completion of the reaction was monitored, water (1mL) was slowly added dropwise to the reaction system. Filtration was carried out, and the filter cake was washed with ethyl acetate (10mL), followed by liquid separation, and the organic phase was washed with saturated brine 3 times, dried over anhydrous sodium sulfate, concentrated, and separated by column chromatography to obtain compounds 1 to 4(2.85g, yield 66%).1H NMR(300MHz,CDCl3):δ7.42-7.36(m,1H),7.30-7.17(m,3H),4.76(dd,J=6.0,1.4Hz,2H),4.52(d,J=15.1Hz,1H),4.38(d,J=15.1Hz,1H),3.79-3.63(m,2H),2.32-2.19(m,1H),2.06-1.93(m,1H),1.39(s,9H),1.23(s,9H).
Step four: synthesis of (R) -N- ((S) -2- (5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropylamine-2-sulfinamide (1-5)
Trifluoroacetic acid (0.5mL) was slowly added dropwise to a solution of 1-4(118.6mg,0.33mmol,1.3eq.) in DCM (2mL) and reacted at room temperature for 2 h. After monitoring the reaction completion, concentrate. DMSO (1mL), DIPEA (1mL) and Compound B (68.3mg,0.25mmol) were added sequentially. Reacting for 2h at 100 ℃. After monitoring the reaction for completion, 25mL of ethyl acetate and 10mL of water were added. The organic phase was washed with saturated brine 5 times, dried over anhydrous sodium sulfate, concentrated, and separated by column chromatography to give compounds 1 to 6(42.0mg, yield 33%).1H NMR(300MHz,CDCl3):δ8.23(d,J=1.3Hz,1H),8.21(d,J=1.3Hz,1H),7.65(d,J=5.5Hz,1H),7.46-7.34(m,2H),7.33-7.23(m,2H),5.98(d,J=5.5Hz,1H),5.06(s,2H),4.87(d,J=15.5Hz,1H),4.76(d,J=15.5Hz,1H),4.38-4.23(m,1H),4.08-3.93(m,1H),3.73(d,J=4.1Hz,1H),2.46-2.35(m,1H),2.17-2.02(m,1H),1.23(s,9H).
Step five: synthesis of (S) -2- (5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-amine (1)
A2M ethyl acetate hydrogen chloride solution (1.0mL) was slowly added dropwise to a solution of 1-5(42.0mg,0.08mmol) in ethyl acetate (2mL) and allowed to react overnight at room temperature. After the reaction was monitored to be complete, 20mL of purified water and 10mL of ethyl acetate were added, extracted, the aqueous phase was adjusted to pH 6 with saturated sodium carbonate, and the aqueous phase was washed with ethyl acetate until all impurities were washed off. The aqueous phase was further adjusted to pH 8, extracted twice with 15mL of ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate, concentrated, and lyophilized to give compound 1(21.1mg, yield 63%).1H NMR(300MHz,CDCl3):δ8.24(d,J=1.4Hz,1H),8.20(d,J=1.4Hz,1H),7.67(d,J=5.5Hz,1H),7.44-7.33(m,2H),7.32-7.28(m,1H),7.26-7.18(m,1H),6.00(d,J=5.5Hz,1H),5.03(d,J=15.1Hz,1H),4.89(s,2H),4.73(d,J=15.1Hz,1H),4.43(dd,J=5.6,2.3Hz,1H),4.25-4.02(m,2H),2.23-2.09(m,1H),1.92-1.81(m,1H)。
Example 2:
Figure BDA0003526744790000081
(2) the synthesis of (2):
detailed procedures referring to the synthesis of compound 1, compound 2 is finally obtained by five-step reactions using 3-methoxybenzylamine as the starting material instead of 1-1.1H NMR(300MHz,CDCl3):δ8.24(d,J=1.4Hz,1H),8.20(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.29(d,J=8.4Hz,1H),6.92(d,J=2.7Hz,1H),6.77(dd,J=8.4,2.7Hz,1H),6.01(d,J=5.5Hz,1H),5.00(d,J=15.1Hz,1H),4.87(s,2H),4.69(d,J=15.1Hz,1H),4.38(dd,J=7.8,2.7Hz,1H),4.20-4.09(m,2H),3.81(s,3H),2.19-2.05(m,1H),1.96-1.77(m,1H)。
Example 3:
Figure BDA0003526744790000082
(3) the synthesis of (2):
detailed procedures referring to the synthesis of compound 1, compound 3 is finally obtained by five-step reactions using 3-methylbenzylamine as a starting material instead of 1-1.1H NMR(300MHz,CDCl3):δ8.24(d,J=1.4Hz,1H),8.21(d,J=1.5Hz,1H),7.68(d,J=5.4Hz,1H),7.27(d,J=8.0Hz,1H),7.16(d,J=1.2Hz,1H),7.08(dd,J=8.0,1.2Hz,1H),6.02(d,J=5.4Hz,1H),4.99(d,J=15.2Hz,1H),4.86(s,2H),4.68(d,J=15.2Hz,1H),4.39(dd,J=7.8,2.6Hz,1H),4.19-4.11(m,2H),2.33(s,3H),2.21-2.05(m,1H),1.92-1.79(m,1H)。
Example 4:
Figure BDA0003526744790000091
(4) the synthesis of (2):
detailed procedures referring to the synthesis of compound 1, compound 4 is finally obtained by five-step reactions using 3-fluorobenzylamine as the starting material instead of 1-1.1H NMR(300MHz,CDCl3):δ8.25(d,J=1.4Hz,1H),8.18(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.35(dd,J=8.5,5.6Hz,1H),7.10(dd,J=8.9,2.7Hz,1H),6.94(ddd,J=8.5,8.2,2.7Hz,1H),6.01(d,J=5.5Hz,1H),5.03(d,J=15.1Hz,1H),4.87(s,2H),4.71(d,J=15.1Hz,1H),4.42(dd,J=7.8,2.6Hz,1H),4.23-4.08(m,2H),2.24-2.09(m,1H),1.92-1.80(m,1H)。
Example 5:
Figure BDA0003526744790000092
(5) the synthesis of (2):
detailed operation steps refer to the synthesis of the compound 1, 3-chlorobenzylamine is used as a starting material to replace 1-1, and the compound 5 is finally obtained through five-step reaction.1H NMR(300MHz,CDCl3):δ8.26(d,J=1.4Hz,1H),8.18(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.38-7.31(m,2H),7.24(dd,J=8.2,2.2Hz,1H),6.02(d,J=5.5Hz,1H),5.02(d,J=15.1Hz,1H),4.96(s,2H),4.69(d,J=15.1Hz,1H),4.41(d,J=7.7Hz,1H),4.22-4.01(m,2H),2.22-2.08(m,1H),1.92-1.78(m,1H)。
Example 6:
Figure BDA0003526744790000093
(6) the synthesis of (2):
the detailed operation steps refer to the synthesis of the compound 1, 3-bromobenzylamine is used as a starting material to replace 1-1, and the compound 6 is finally obtained through five-step reaction.1H NMR(300MHz,CDCl3):δ8.26(d,J=1.4Hz,1H),8.18(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.51(d,J=2.1Hz,1H),7.40(dd,J=8.1,2.1Hz,1H),7.28(d,J=8.1Hz,1H),6.02(d,J=5.5Hz,1H),5.01(d,J=15.1Hz,1H),4.93(s,2H),4.69(d,J=15.1Hz,1H),4.39(dd,J=8.1,2.7Hz,1H),4.23-4.01(m,2H),2.21-2.08(m,1H),1.93-1.79(m,1H)。
Example 7:
Figure BDA0003526744790000101
(7) the synthesis of (2):
detailed procedures referring to the synthesis of compound 1, compound 7 was finally obtained by five-step reactions using 3-cyanobenzylamine as the starting material in place of 1-1.1H NMR(300MHz,DMSO-d6):δ8.50(d,J=1.4Hz,1H),8.28(d,J=1.4Hz,1H),7.99(d,J=1.7Hz,1H),7.75(dd,J=8.0,1.7Hz,1H),7.64(d,J=8.0Hz,1H),7.62(d,J=5.4Hz,1H),6.35(s,2H),5.75(d,J=5.4Hz,1H),5.08(d,J=15.3Hz,1H),4.81(d,J=15.3Hz,1H),4.45(dd,J=8.7,2.0Hz,1H),4.30-4.14(m,1H),4.04-3.85(m,1H),2.07-1.93(m,1H),1.77-1.61(m,1H)。
Example 8:
Figure BDA0003526744790000102
(8) the synthesis of (2):
detailed operation steps referring to the synthesis of the compound 1, 3-aminobenzylamine is used as a starting material to replace 1-1, and the compound 8 is finally obtained through five-step reaction.1H NMR(300MHz,CDCl3):δ8.23(d,J=1.4Hz,1H),8.19(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.14(d,J=8.1Hz,1H),6.69(d,J=2.5Hz,1H),6.56(dd,J=8.1,2.5Hz,1H),6.02(d,J=5.4Hz,1H),4.93(d,J=15.1Hz,1H),4.87(s,1H)4.62(d,J=15.1Hz,1H),4.32(dd,J=7.5,2.5Hz,1H),4.20-4.06(m,2H),3.67(s,2H),2.18-2.04(m,1H),1.91-1.77(m,1H)。
Example 9:
Figure BDA0003526744790000103
(9) the synthesis of (2):
detailed procedures refer to the synthesis of compound 1, replacing 1-1 with 3- (pyrrolidin-1-yl) benzylamine as starting material, and obtaining compound 9 through five-step reactions.1H NMR(300MHz,CDCl3):δ8.25(d,J=1.4Hz,1H),8.22(d,J=1.4Hz,1H),7.68(d,J=5.5Hz,1H),7.19(d,J=8.2Hz,1H),6.56(d,J=2.5Hz,1H),6.41(dd,J=8.2,2.5Hz,1H),6.02(d,J=5.5Hz,1H),5.00(d,J=15.1Hz,1H),4.86(s,2H),4.66(d,J=15.1Hz,1H),4.34(dd,J=7.6,2.4Hz,1H),4.27-4.10(m,2H),3.36-3.22(m,4H),2.18-2.05(m,1H),2.02-1.92(m,4H),1.93-1.81(m,1H)。
Example 10:
Figure BDA0003526744790000111
(10) the synthesis of (2):
detailed procedures refer to the synthesis of compound 1, and 3-dimethylaminobenzylamine is used as a starting material to replace 1-1, and compound 10 is finally obtained through five-step reactions.1H NMR(300MHz,CDCl3):δ8.23(s,2H),7.68(d,J=5.5Hz,1H),7.22(d,J=8.4Hz,1H),6.73(d,J=2.7Hz,1H),6.58(dd,J=8.4,2.7Hz,1H),6.02(d,J=5.5Hz,1H),5.00(d,J=15.1Hz,1H),4.86(s,2H),4.68(d,J=15.1Hz,1H),4.34(dd,J=7.4,2.5Hz,1H),4.22-4.09(m,2H),2.95(s,6H),2.19-2.05(m,1H),1.90-1.81(m,1H)。
Example 11:
Figure BDA0003526744790000112
(11) the synthesis of (2):
detailed description of the preferred embodimentsreferring to the synthesis of compound 1, compound 11 was finally obtained by a five-step reaction starting with 3- (4-fluorophenyl) benzylamine instead of 1-1.1H NMR(400MHz,CDCl3):δ8.25(d,J=1.4Hz,1H),8.23(d,J=1.4Hz,1H),7.65(d,J=5.4Hz,1H),7.58-7.50(m,3H),7.49-7.39(m,2H),7.18-7.07(m,2H),6.01(d,J=5.4Hz,1H),5.10(d,J=15.1Hz,1H),4.87(s,2H),4.79(d,J=15.1Hz,1H),4.76(brs,1H),4.47(dd,J=7.8,2.6Hz,1H),4.27-4.10(m,2H),2.25-2.14(m,1H),1.96-1.85(m,1H)。
Example 12:
Figure BDA0003526744790000113
(12) the synthesis of (2):
Figure BDA0003526744790000121
the method comprises the following steps: synthesis of N- (tert-butoxycarbonyl) -N-phenethylglycine (12-2):
ethyl bromopropionate (1.18mL,10.0mmol,1.0eq.) and triethylamine (2.78mL,20.0mmol,2.0eq.) were added dropwise in that order to a solution of 12-1(1.21g,10.0mmol) in THF (20 mL). The reaction was carried out at room temperature for 4h, after completion of the reaction was monitored, the reaction mixture was concentrated, and DCM (20mL), triethylamine (2.78mL,20.0mmol,2.0eq.) and Boc were added to the mixture in this order2O (2.40g,11.0mmol,1.1 eq.). Reacting for half an hour at room temperature, monitoring the reaction completion, concentrating, and adding THF/H into the mixture in sequence2O/EtOH (5/5/5mL) mixed solution and lithium hydroxide (0.72g, 30.0mmol, 3.0 eq.). After the reaction was completed for 6 hours at room temperature, the reaction mixture was concentrated, 20mL of water and 20mL of ethyl acetate were added, the mixture was extracted, the aqueous phase was separated, the pH was adjusted to 3 with hydrochloric acid (3M), and then an appropriate amount of ethyl acetate was added and the mixture was extracted twice.The organic phase was washed with an appropriate amount of saturated brine for 3 times, dried over anhydrous sodium sulfate, and concentrated to give compound 12-2(2.01g, crude).
Step two: synthesis of 1-oxo-1, 2,4, 5-tetrahydro-3H-benzo [ d ] aza-3-carboxylic acid tert-butyl ester (12-3):
referring to step two of the synthetic route of example 1, 12-2 was substituted for 1-2 to give compound 12-3(0.31g, 16% yield over six steps).1H NMR(300MHz,CDCl3):δ7.77(d,J=7.6,1.2Hz,1H),7.43(ddd,J=7.5,7.4,1.2Hz,1H),7.36(ddd,J=7.6,7.4,1.0Hz,1H),7.23(dd,J=7.5,1.0Hz,1H),4.17(d,J=17.7Hz,2H),3.79-3.68(m,2H),3.08-2.97(m,2H),1.22(s,9H).
Step three: synthesis of tert-butyl (R) -1- (((R) -tert-butylsulfinyl) amino) -1,2,4, 5-tetrahydro-3H-benzo [ d ] aza-3-carboxylate (12-4):
referring to step three of the synthetic route of example 1, 12-3 was substituted for 1-3 to give compound 12-4(0.14g, yield 32%). LC-MS (ESI):367.2(M + H)+.
Step four: synthesis of (R) -N- ((R) -3- (5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropylamine-2-sulfinamide (12-5):
referring to step four of the synthetic route of example 1, 12-4 was substituted for 1-4 to give compound 12-5(98.2mg, yield 66%).1H NMR(300MHz,CDCl3):δ8.36(d,J=1.4Hz,1H),8.30(d,J=1.4Hz,1H),7.70(d,J=5.4Hz,1H),7.46-7.39(m,1H),7.35-7.29(m,1H),7.25-7.19(m,1H),7.14-7.09(m,1H),5.98(d,J=5.4Hz,1H),5.14(dd,J=14.6,5.6Hz,1H),4.96(s,2H),4.83-4.78(m,1H),4.36(dd,J=14.6,5.3Hz,1H),4.26(d,J=2.2Hz,1H),3.61-3.20(m,3H),2.97-2.85(m,1H),0.98(s,9H).
Step five: synthesis of (R) -3- (5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ d ] azepin-1-amine (12):
referring to step five of the synthetic route of example 1, substituting 12-5 for 1-5, Compound 12(45.2mg, 58% yield) was obtained.1H NMR(300MHz,CDCl3):δ8.28(d,J=1.3Hz,1H),8.26(d,J=1.3Hz,1H),7.69(d,J=5.5Hz,1H),7.36-7.29(m,1H),7.26-7.19(m,2H),7.19-7.14(m,1H),6.00(d,J=5.5Hz,1H),4.92(s,2H),4.36(dd,J=7.2,2.9Hz,1H),4.20(dd,J=14.3,7.2Hz,1H),4.05(ddd,J=13.3,6.4,2.8Hz,1H),3.92(dd,J=14.3,2.9Hz,1H),3.69(ddd,J=13.3,9.7,2.5Hz,1H),3.37(ddd,J=15.2,9.7,2.8Hz,1H),2.99(ddd,J=15.2,6.4,2.5Hz,1H)。
Example 13:
Figure BDA0003526744790000131
(13) the synthesis of (2):
detailed procedures referring to the synthesis of compound 12, compound 13 was finally obtained by five-step reaction using 3-fluorophenethylamine as starting material instead of 12-1.1H NMR(300MHz,CDCl3)δ8.28(d,J=1.2Hz,1H),8.26(d,J=1.2Hz,1H),7.70(d,J=5.4Hz,1H),7.30(dd,J=8.2,7.0Hz,1H),6.98-6.82(m,2H),6.00(d,J=5.4Hz,1H),4.89(s,2H),4.35(dd,J=7.1,2.7Hz,1H),4.20(dd,J=14.3,7.1Hz,1H),4.08(ddd,J=14.5,6.5,2.6Hz,1H),3.88(dd,J=14.3,2.7Hz,1H),3.68(ddd,J=14.5,9.8,2.1Hz,1H),3.36(ddd,J=15.2,9.8,2.6Hz,1H),3.94(ddd,J=15.2,6.5,2.1Hz,1H)。
Example 14:
Figure BDA0003526744790000132
(14) the synthesis of (2):
detailed procedures referring to the synthesis of compound 12, compound 14 is finally obtained by five-step reactions using 3-methoxyphenethylamine as starting material instead of 12-1.1H NMR(300MHz,CDCl3):δ8.28(d,J=1.4Hz,1H),8.26(d,J=1.4Hz,1H),7.70(d,J=5.5Hz,1H),7.24(d,J=8.1Hz,1H),6.78-6.69(m,2H),6.01(d,J=5.5Hz,1H),4.87(s,2H),4.32(dd,J=6.9,2.6Hz,1H),4.22(dd,J=14.1,6.9Hz,1H),4.10(ddd,J=14.5,7.1,2.8Hz,1H),3.85(dd,J=14.1,2.6Hz,1H),3.80(s,3H),3.65(ddd,J=14.5,10.2,2.0Hz,1H),3.35(ddd,J=15.4,10.2,2.8Hz,1H),2.91(ddd,J=15.4,7.1,2.0Hz,1H)。
Example 15:
Figure BDA0003526744790000141
(15) the synthesis of (2):
Figure BDA0003526744790000142
the method comprises the following steps: synthesis of 3- ((2-amino-3-chloropyridin-4-yl) thio) -6-chloropyrazin-2-amino (15-2):
placing compounds 15-1(208.4mg,1.0mmol), a (192.7mg,1.2mmol,1.2eq.), cuprous iodide (38.9mg,0.2mmol,0.2eq.), 1, 10-phenanthroline (54.2mg,0.3mmol,0.3eq.) and potassium phosphate (424.5mg,2.0mmol,2.0eq.) in a 15mL sealed tube, vacuumizing the system to replace nitrogen, adding 1, 4-dioxane (4mL) to react in an oil bath at 85 ℃ overnight, and monitoring until the conversion of the raw materials is complete. Filtering with diatomaceous earth, concentrating the filtrate, adding 20mL ethyl acetate and 10mL water, extracting, separating, washing the organic phase with saturated saline solution for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound 15-2(216.2mg, yield 75%).1H NMR(300MHz,CDCl3):δ8.01(s,1H),7.77(d,J=5.4Hz,1H),6.09(d,J=5.4Hz,1H),5.23(s,2H),4.96(s,2H).
Step two: synthesis of (R) -N- ((S) -2- (6-amino-5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (15-3):
trifluoroacetic acid (0.5mL) was slowly added dropwise to a solution of 1-4(186.0mg,0.52mmol,1.3eq.) in DCM (2mL) and reacted at room temperature for 2 h. After monitoring the reaction completion, concentrate. NMP (1mL), potassium carbonate (220.8mg,1.6mmol,4.0eq.) and compound 15-3(115.3mg,0.4mmol) were added sequentially. The reaction is carried out for 10h at 100 ℃. After monitoring the reaction for completion, 25mL of ethyl acetate and 10mL of water were added. The organic phase was washed with saturated brine 5 times, dried over anhydrous sodium sulfate, concentrated, and separated by column chromatography to give compound 15-3(95.2mg, yield 46%).1H NMR(300MHz,CDCl3):δ8.24(d,J=1.3Hz,1H),8.20(d,J=1.3Hz,1H),7.67(d,J=5.5Hz,1H),7.45-7.34(m,2H),7.33-7.27(m,2H),5.99(d,J=5.5Hz,1H),5.02-4.72(m,6H),4.40-4.23(m,1H),4.05-3.93(t,J=11.5Hz,1H),3.46(d,J=4.0Hz,1H),2.37-2.31(m,1H),2.18-2.03(m,1H),1.22(s,9H).
Step three: synthesis of (S) -2- (6-amino-5- ((2-amino-3-chloropyridin-4-yl) thio) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-amine (15):
referring to step five of the synthetic route of example 1, substituting 15-3(80.9mg,0.16mmol) for 1-5 gave compound 15(44.2mg, yield 67%).1H NMR(300MHz,CDCl3):δ7.67(d,J=5.4Hz,1H),7.64(s,1H),7.41(d,J=7.8Hz,1H),7.36-7.28(m,2H),7.25-7.18(m,1H),6.01(d,J=5.4Hz,1H),4.92(d,J=15.2Hz,1H),4.87(s,2H),4.79(s,2H),4.68(d,J=15.2Hz,1H),4.40(dd,J=8.2,2.5Hz,1H),4.17-3.88(m,2H),2.23-2.10(m,1H),1.88-1.76(m,1H)。
Example 16:
Figure BDA0003526744790000151
(16) the synthesis of (2):
Figure BDA0003526744790000152
the method comprises the following steps: synthesis of 6-chloro-3- (2, 3-dichlorophenyl) pyrazin-2-amine (16-2):
compound 16-1(0.21g,1.0mmol), 2, 3-dichlorophenylboronic acid (0.21g,1.1mmol,1.1eq.), pd (dppf) Cl2(14.6mg,2 mol%) and potassium phosphate (0.42g,2.0mmol,2.0eq.) were placed in a 15mL sealed tube, the system was evacuated to replace nitrogen, 1, 4-dioxane (2mL) and water (0.2mL) were added and reacted overnight in a 120 ℃ oil bath with TLC monitoring until complete conversion of the starting material. Filtering with celite, concentrating the filtrate, extracting with 20mL ethyl acetate, washing with an appropriate amount of saturated brine for 3 times, drying with anhydrous sodium sulfate, concentrating, and separating by column chromatography to obtain compound 16-2(0.25g, yield 91%).1H NMR(300MHz,CDCl3):δ8.03(s,1H),7.60(dd,J=7.6,2.0Hz,1H),7.36(dd,J=7.7,7.6Hz,1H),7.32(dd,J=7.6,2.0Hz,1H),4.65(s,2H).
Step two: synthesis of 6-chloro-3- (2, 3-dichlorophenyl) pyrazine-2-amino-tert-butyl ester (16-3):
compound 17-2(0.25g,0.91mmol,1.0eq.) and DMAP (5.6mg,0.05mmol,0.05eq.) were placed in a 10mL single vial, DCM (5mL) and di-tert-butyl dicarbonate (0.44g,1.91mmol,2.1eq.) were added sequentially, after addition, the reaction was carried out at room temperature for 2h, and TLC was monitored as it wasThe conversion of the material is complete. Washing with saturated salt for 3 times, drying the organic phase with anhydrous sodium sulfate, concentrating, and separating by column chromatography to obtain compound 16-3(0.35g, yield 80%).1H NMR(300MHz,CDCl3):δ8.68(s,1H),7.57(d,J=7.5Hz,1H),7.46-7.24(m,2H),1.37(s,18H).
Step three: synthesis of tert-butyl (6- ((S) -5- (((R) -tert-butylsulfinyl) amino) -1,3,4, 5-tetrahydro-2H-benzo [ c ] azepin-2-yl) -3- (2, 3-dichlorophenyl) pyrazin-2-yl) carbamate (16-4)
Referring to step four of the synthetic route of example 1, substituting 16-3(0.19g,0.4mmol) for B gave compound 16-4(0.15g, yield 55%).1H NMR(300MHz,CDCl3):δ8.18(s,1H),7.46(dd,J=7.9,1.7Hz,1H),7.43-7.35(m,2H),7.34-7.23(m,3H),7.20(t,J=7.8Hz,1H),4.91(d,J=15.4Hz,1H)4.85-4.78(m,1H),4.74(d,J=15.4Hz,1H),4.44-4.23(m,1H),4.07-3.93(m,1H),3.47(d,J=3.8Hz,1H),2.47-2.30(m,1H),2.19-2.10(m,1H),1.34(s,18H),1.24(s,9H).
Step four: synthesis of (S) -2- (6-amino-5- (2, 3-dichlorophenyl) pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] aza-5-amine (16):
referring to step five of the synthetic route of example 1, substituting 16-4 for 1-5, Compound 16 was obtained (46.5mg, yield 52%).1H NMR(300MHz,CDCl3):δ7.58(s,1H),7.47(dd,J=7.0,2.6Hz,1H),7.41(d,J=7.4Hz,1H),7.36(dd,J=7.1,1.7Hz,1H),7.32-7.19(m,4H),4.89(d,J=15.6Hz,1H),4.67(d,J=15.6Hz,1H),4.40(dd,J=8.3,2.8Hz,1H),4.20(s,2H),4.10-3.90(m,2H),2.26-2.21(m,1H),1.91-1.83(m,1H)。
Example 17:
Figure BDA0003526744790000161
(17) the synthesis of (2):
Figure BDA0003526744790000162
the method comprises the following steps: synthesis of 2-chloro-N- (2, 2-dimethoxyethyl) -5-iodopyrimidin-4-amine (17-2): triethylamine (2.77mL,20.0mmol,2.0eq.) was added dropwise to a solution of 17-1(2.74g,10.0mmol) and 2, 2-dimethoxyethane-1-amine (2.10g,20.0mmol,2.0eq.) in ethanol (40mL) at 0 ℃. The reaction was allowed to proceed overnight at room temperature and monitored by TLC until complete conversion of starting material. After concentration, 30mL of ethyl acetate and 10mL of water were added, the mixture was separated, and the organic phase was washed 3 times with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain Compound 17-2(3.64g, crude product).
Step two: synthesis of 8-iodoimidazo [1,2-c ] pyrimidin-5-ol (17-3):
17-2(3.64g, crude) was added to concentrated sulfuric acid (40 mL). The reaction was allowed to proceed overnight at room temperature and monitored by TLC until complete conversion of starting material. The reaction solution was poured into ice water, pH was adjusted to 6 with sodium hydroxide, and filtered to give compound 17-3(2.55g, crude product).
Step three: synthesis of 5-chloro-8-iodoimidazo [1,2-c ] pyrimidine (17-4):
DIPEA (1mL) was added dropwise to a solution of 17-3(0.52g,2.0mmol) in phosphorus oxychloride (8 mL). The reaction was carried out at 120 ℃ for 5h and TLC monitored until complete conversion of starting material. After concentration, 20mL of dichloromethane was added, and the mixture was poured into 10mL of ice water, followed by liquid separation, the organic phase was washed with saturated brine 3 times, dried over anhydrous sodium sulfate, concentrated, and column chromatography to give compound 17-4(0.34g, 63%).1H NMR(300MHz,CDCl3):δ8.21(s,1H),7.89(d,J=1.5Hz,1H),7.82(d,J=1.5Hz,1H).
Step four: synthesis of (R) -N- ((S) -2- (8-iodoimidazo [1,2-c ] pyrimidin-5-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (17-5):
trifluoroacetic acid (0.5mL) was slowly added dropwise to a solution of 1-4(0.44g,1.2mmol,1.2eq.) in DCM (2mL) and reacted at room temperature for 2 h. After monitoring the reaction to completion, concentrate. Acetonitrile (2mL), DIPEA (0.35mL,2.0mmol,2.0eq.) and compound 17-5(279.5mg,1.0mmol) were added sequentially. The reaction was carried out at room temperature for 4 hours. After monitoring the reaction for completion, 25mL of ethyl acetate and 10mL of water were added. The organic phase was washed with saturated brine 3 times, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give compound 17-5(0.35g, yield 69%).1H NMR(300MHz,CDCl3):δ8.01(s,1H),7.66(d,J=1.5Hz,1H),7.58(d,J=1.5Hz,1H),7.47(dd,J=7.4,1.4Hz,1H),7.38(ddd,J=7.5,7.4,1.4Hz,1H),7.31(ddd,J=7.5,7.4,1.4Hz,1H),7.14(dd,J=7.4,1.4Hz,1H),4.90-4.83(m,1H),4.78(d,J=15.8Hz,1H),4.62(d,J=15.8Hz,1H),4.08-3.84(m,2H),3.72(d,J=5.9Hz,1H),2.64-2.51(m,1H),2.50-2.35(m,1H),1.17(s,9H).
Step five: synthesis of (R) -N- ((S) -2- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (17-6)
Compound 17-5(127.3mg,0.25mmol), A-5(54.5mg,0.30mmol,1.2eq.), Pd2dba3(23.0mg,10 mol%) and Xantphos (29.0mg,20 mol%) were placed in a 15mL sealed tube, the system was evacuated to replace nitrogen, 1, 4-dioxane (2mL) and DIPEA (87. mu.L, 0.5mmol,2.0eq.) were added and reacted in a 100 ℃ oil bath overnight, monitored until complete conversion of the starting material. Diluting with 10mL ethyl acetate, filtering with diatomaceous earth, concentrating the filtrate, adding 15mL ethyl acetate and 5mL water, extracting and layering, washing the organic phase with saturated saline solution for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound 17-6(42.1mg, yield 31%).1H NMR(300MHz,CDCl3):δ7.98(s,1H),7.63(d,J=1.3Hz,1H),7.60(d,J=5.4Hz,1H),7.57(d,J=1.3Hz,1H),7.50(dd,J=7.7,1.4Hz,1H),7.38(ddd,J=7.7,7.6,1.4Hz,1H),7.32(ddd,J=7.6,7.5,1.4Hz,1H),7.17(dd,J=7.5,1.4Hz,1H),5.84(d,J=5.4Hz,1H),4.94(d,J=15.4Hz,1H),4.91(s,2H),4.79(d,J=15.4Hz,1H),4.16-4.03(m,2H),3.66(d,J=5.4Hz,1H),2.71-2.58(m,1H),2.53-2.38(m,1H),1.19(s,9H).
Step six: synthesis of (S) -2- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-amine (17):
referring to step five of the synthetic route of example 1, substituting 17-6 for 1-5, Compound 17(25.0mg, yield 72%) was obtained.1H NMR(300MHz,CDCl3):δ7.97(s,1H),7.64-7.54(m,3H),7.48(dd,J=7.5,1.1Hz,1H),7.36(ddd,J=7.8,7.5,1.3Hz,1H),7.25(ddd,J=7.8,7.4,1.1Hz,1H),7.16(ddd,J=7.4,1.3Hz,1H),5.86(d,J=5.4Hz,1H),5.05(d,J=15.6Hz,1H),4.89(s,2H),4.73(d,J=15.6Hz,1H),4.48(dd,J=7.8,1.4Hz,1H),4.20-4.09(m,1H),4.01-3.89(m,1H),2.43-2.32(m,1H),2.28-2.15(m,1H)。
Example 18:
Figure BDA0003526744790000181
(18) the synthesis of (2):
Figure BDA0003526744790000182
the method comprises the following steps: synthesis of 6-amino-3-methylpyrimidine-2, 4(1H,3H) -dione (18-2)
Concentrated sulfuric acid (0.05mL) was added slowly dropwise to a solution of 4-amino-2, 6-dihydroxypyrimidine (0.52g,4.1mmol) in HMDS (2.5 mL). After 6h reaction at 130 ℃ the mixture was concentrated to remove HMDS. DMF (2.5mL) and methyl iodide (0.85mL,14.4mmol,3.5eq.) were added in this order and reacted at room temperature overnight. After completion of the reaction was monitored, a saturated solution of sodium hydrogencarbonate was added dropwise until no bubble was generated, followed by suction filtration, water washing of the cake and drying to obtain Compound 18-2(0.36g, yield 63%).1H NMR(300MHz,DMSO-d6):δ10.43(s,1H),6.23(brs,2H),4.59(s,1H),3.00(s,3H).
Step two: synthesis of 6-amino-5-iodo-3-methylpyrimidine-2, 4(1H,3H) -dione (18-3)
Compound 18-2(0.19g,1.4mmol) was dissolved in DMF (2mL) and AcOH (4mL) and NIS (0.37g,1.68mmol,1.2eq.) was added. Reacting for 2h at room temperature, monitoring the reaction, filtering, washing the filter cake with water, and drying to obtain a compound 18-3(0.30g, crude product).
Step three: synthesis of (R) -2-methyl-N- ((S) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) propane-2-sulfinamide (18-4)
Trifluoroacetic acid (0.5mL) was added to a solution of 1-5(0.36g,1.0mmol) in DCM (2mL) and reacted at room temperature for 2 h. The reaction was monitored for completion, concentrated, 5mL of water was added and the pH was adjusted to 9 with saturated aqueous sodium carbonate solution. Ethyl acetate (10mL) was extracted twice and concentrated to give 18-4 which was directly used for the next reaction.
Step four: synthesis of (R) -N- ((S) -2- (4-amino-5-iodo-1-methyl-6-oxo-1, 6-dihydropyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (18-5)
DBU (0.8mL,5.25mmol,7.0eq.) was added dropwise to 18-3(0.2g, 0.7.)5mmol), 18-4 (from step three), and Bop (0.66g,1.5mmol,2.0eq.) in DMF (2mL) at room temperature for 8 h. After completion of the reaction was monitored, 20mL of ethyl acetate and 5mL of water were added, followed by liquid separation, and the organic phase was washed with an appropriate amount of saturated brine 5 times, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography to give 18-5(0.11g, yield 28%). LC-MS (ESI) 516.2(M + H)+.
Step five: synthesis of (R) -N- ((S) -2- (4-amino-5- ((2-amino-3-chloropyridin-4-yl) thio) -1-methyl-6-oxo-1, 6-dihydropyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (18-6):
compounds 18-5(0.1g,0.19mmol), a (62.6mg,0.39mmol,2.0eq.), cuprous iodide (7.4mg,0.04mmol,0.2eq.), TMEDA (12 μ L,0.08mmol,0.4eq.), and potassium phosphate (120.9mg,0.57mmol,3.0eq.) were placed in a 15mL sealed tube, the system was evacuated to replace nitrogen, 1, 4-dioxane (1mL) was added and reacted in a 100 ℃ oil bath overnight, monitored until the conversion of the starting material was complete. Diluting with 10mL of ethyl acetate, filtering with celite, concentrating the filtrate, adding 10mL of ethyl acetate and 10mL of water, extracting and separating, washing the organic phase with saturated brine for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound 18-6(46.7mg, yield 40%).1H NMR(300MHz,CDCl3):δ7.70(d,J=5.4Hz,1H),7.45(dd,J=7.5,1.4Hz,1H),7.39-7.31(m,2H),7.20(dd,J=8.0,1.5Hz,1H),6.15(d,J=5.4Hz,1H),4.86(s,2H),4.84-4.74(m,1H),4.63(d,J=15.7Hz,1H),4.50(d,J=15.7Hz,1H),3.87-3.73(m,2H),3.57(d,J=5.9Hz,1H),3.42(s,3H),2.60-2.44(m,1H),2.41-2.27(m,1H),1.23(s,9H).
Step six: synthesis of (S) -6-amino-2- (5-amino-1, 3,4, 5-tetrahydro-2H-benzo [ c ] azepin-2-yl) -5- ((2-amino-3-chloropyridin-4-yl) thio) -3-methylpyrimidin-4 (3H) -one (18):
referring to step five of the synthetic route of example 1, substituting 18-6 for 1-5, Compound 18(17.6mg, yield 46%) was obtained.1H NMR(300MHz,CDCl3):δ7.67(d,J=5.4Hz,1H),7.43(dd,J=7.6,1.4Hz,1H),7.33(ddd,J=7.6,7.4,1.4Hz,1H),7.23(ddd,J=7.6,7.4,1.0Hz,1H),7.16(dd,J=7.6,1.0Hz,1H)6.14(d,J=5.4Hz,1H),5.23(s,2H),4.93(s,2H),4.72(d,J=15.5Hz,1H),4.46(d,J=15.5Hz,1H),4.41(dd,J=8.1,2.0Hz,1H),3.91-3.77(m,1H),3.70-3.57(m,1H),3.41(s,3H),2.31-3.16(m,1H),2.16-2.06(m,1H)。
Example 19:
Figure BDA0003526744790000201
(19) the synthesis of (2):
Figure BDA0003526744790000202
the method comprises the following steps: synthesis of 5-bromothiazolo [5,4-b ] pyridin-2-amine (19-2)
19-1(1.0g,5.78mmol) was slowly added to a solution of KSCN (2.2g,22.8mmol,4.0eq.) in acetic acid (10 mL). After stirring at room temperature for fifteen minutes, a solution of bromine (0.38mL,7.51mmol,1.3eq.) in acetic acid (10mL) was slowly added dropwise. The reaction was carried out at room temperature for 3h and the completion of the reaction was monitored. The reaction was quenched dropwise with water, filtered, and the filtrate was concentrated, adjusted to pH 7 with saturated aqueous sodium carbonate solution, extracted twice with ethyl acetate (20mL), the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give compound 19-2(1.33g, crude product).
Step two: synthesis of 5-bromo-2-chlorothiazolo [5,4-b ] pyridine (19-3)
The chloroidene (0.65g,4.9mmol,1.5eq.) was added to a solution of 19-2(0.75g,3.2mmol) in acetonitrile (10mL) at 0 ℃. After stirring at this temperature for fifteen minutes, a solution of tBuONO (0.57mL,4.9mmol,1.5eq.) in acetonitrile (2mL) was slowly added dropwise. The reaction was allowed to proceed overnight at room temperature and was monitored for completion. After concentration, ethyl acetate (20mL) and water (5mL) were added to separate the layers, and the organic layer was washed with saturated sodium bicarbonate and saturated brine, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography to give compound 19-3(0.63g, 44% yield in two steps).1H NMR(300MHz,CDCl3):δ8.06(d,J=8.5Hz,1H),7.63(d,J=8.5Hz,1H).
Step three: synthesis of 3-chloro-4- ((2-chlorothiazolo [5,4-b ] pyridin-5-yl) thio) pyridin-2-amine (19-4)
Mixing the compound 19-3(0.63g,2.5mmol), A-5(0.59g,3.25mmol,1.3eq.), Pd2dba3(0.23g,10 mol%) and Xantphos (0.29g,20 mol%) were placed at 15mIn an L sealed tube, the system was evacuated to replace nitrogen, 1, 4-dioxane (2mL) and DIPEA (0.85mL,5.0mmol,2.0eq.) were added, reacted overnight in an oil bath at 100 ℃ and monitored until complete conversion of the starting material. Diluting with 10mL of ethyl acetate, filtering with celite, concentrating the filtrate, adding 15mL of ethyl acetate and 5mL of water, extracting and layering, washing the organic phase with saturated brine for 3 times, drying with anhydrous sodium sulfate, concentrating, and purifying by column chromatography to obtain compound 19-4(0.15g, yield 18%). LC-MS (ESI) 329.0(M + H)+.
Step four: synthesis of (R) -N- ((S) -2- (5- ((2-amino-3-chloropyridin-4-yl) thio) thiazole [5,4-b ] pyridin-2-yl-2, 3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-yl) -2-methylpropane-2-sulfinamide (19-5):
trifluoroacetic acid (0.5mL) was slowly added dropwise to a solution of 1-4(0.22g,0.59mmol,1.3eq.) in DCM (2mL) and reacted at room temperature for 2 h. After monitoring the reaction completion, concentrate. DMF (2mL), cesium carbonate (0.24g,0.9mmol,2.0eq.) and compound 19-4(0.15g,0.45mmol) were added sequentially. The reaction was carried out at room temperature for 4 hours. After monitoring the reaction for completion, 25mL of ethyl acetate and 10mL of water were added. The organic phase was washed with saturated brine 3 times, dried over anhydrous sodium sulfate, concentrated, and isolated by column chromatography to give compound 19-5(56.2mg, yield 27%).1H NMR(300MHz,CDCl3):δ7.70(d,J=8.3Hz,1H),7.59(d,J=5.6Hz,1H),7.50(d,J=8.3Hz,1H),7.46-7.39(m,2H),7.38-7.30(m,2H),6.04(d,J=5.6Hz,1H),5.54(s,2H),4.93(d,J=15.4Hz,1H),4.87-4.79(s,1H),4.71(d,J=15.4Hz,1H),4.36-4.19(m,2H),3.77(d,J=4.0Hz,1H),2.54-2.40(m,2H),1.25(s,9H).
Step five: synthesis of (S) -2- (5- ((2-amino-3-chloropyridin-4-yl) thio) thiazolo [5,4-b ] pyridin-2-yl) -2,3,4, 5-tetrahydro-1H-benzo [ c ] azepin-5-amine (19):
referring to step five of the synthetic route of example 1, substituting 19-5 for 1-5, Compound 19(33.0mg, yield 72%) was obtained.1H NMR(300MHz,CDCl3):δ7.68(d,J=8.2Hz,1H),7.66(d,J=5.4Hz,1H),7.46(d,J=8.2Hz,1H),7.40(dd,J=7.3,1.3Hz,2H),7.30(ddd,J=7.6,7.3,1.3Hz,1H),7.25(ddd,J=7.6,7.3,1.6Hz,1H),6.04(d,J=5.4Hz,1H),5.10(d,J=15.1Hz,1H),4.90(s,2H),4.68(d,J=15.1Hz,1H),4.45(dd,J=7.9,2.6Hz,1H),4.29-4.16(m,1H),4.15-3.99(m,1H),2.31-2.15(m,1H),1.99-1.84(m,1H)。
The present invention is further described and explained below in conjunction with test examples, which are not intended to limit the scope of the present invention.
Example 21 in vitro SHP2 enzyme level Activity assay
The compounds of the above examples were tested for activity at the SHP2 enzyme level by the following specific procedures:
1 preparation of the Compound
The compound was dissolved in DMSO to prepare a 30mM stock solution, which was stored in a-20 ℃ freezer under dark conditions.
2SHP2 reaction process
(1) A1 XReactionbuffer was prepared.
(2) Preparation of compound concentration gradient: test compounds were tested at an initial concentration of 30 μ M, 3-fold dilution, 10 concentrations, single well assay. 100% DMSO solution diluted to 100-fold final concentration in 384source plates, compounds were diluted 3-fold with Precision, 10 concentrations. 250nL of 100-fold final concentration of compound was transferred to the target 384 plates using a dispenser Echo 550. 250nL DMSO was added to the positive control, and 250nL of 1mM SHP099 was added to the negative control.
(3) 5-fold final concentration of the activated peptide solution was prepared using 1 × ReactionBuffer, and 5 μ L of the solution was added to each reaction plate and centrifuged at 1000rpm for 1 min.
(4) A2.5-fold final concentration of enzyme solution was prepared using 1 XReactionBuffer, and 10. mu.L of each solution was added to the reaction plate, centrifuged at 1000rpm for 1min, and incubated at room temperature for 60 minutes.
(5) A substrate solution of 2.5 times final concentration was prepared using 1 XReactionBuffer, and 10. mu.L of each was added to the reaction plate, centrifuged at 1000rpm for 1min, and incubated at room temperature for 20 minutes.
(6) Ex355/Em460 fluorescence values were read with EnSight
3 data analysis
Formula for calculation
Figure BDA0003526744790000221
Wherein: RFU: fluorescence value of the sample; mean (nc): mean fluorescence values for control wells containing 10 μ M SHP 099;
mean (pc): mean fluorescence values of positive control wells.
Fitting dose-effect curve
The log value of the concentration is taken as an X axis, the percent inhibition rate is taken as a Y axis, and a dose-effect curve is fitted by adopting the log (inhibitor) vs. response-Variable slope of the GraphPad Prism 5 analysis software, so as to obtain the IC of each compound on the enzyme activity50The value is obtained.
The calculation formula is Y ═ Bottom + (Top-Bottom)/(1+10^ ((LogIC)50-X)*HillSlope))
4. Results of the experiment
The specific results are shown in the table:
compound numbering SHP2IC50(nM) Compound numbering SHP2IC50(nM)
1 A 10 A
2 A 11 A
3 A 12 C
4 B 13 C
5 B 14 C
6 A 15 C
7 B 16 A
8 B 17 C
9 A 18 C
SHP099 107 19 C
A<50nM,50nM≤B≤100nM,C>100nM。
And (4) experimental conclusion: the above data show that the examples of the invention have allosteric inhibitory effects on SHP2 phosphatase, and that many of the examples are significantly superior to the positive control SHP 099.
EXAMPLE 22 in vitro antiproliferative Activity of Compounds
1. Experimental procedure
(1) The PBS solution was autoclaved and stored in a refrigerator at 4 ℃.
(2) Weighing trypsin and pancreatin digestive juice, adding ultrapure water to fully dissolve, filtering with a microporous filter to obtain liquid, and storing in a refrigerator at-20 ℃.
(3) Respectively weighing culture medium powder and NaHCO3Adding ultrapure water for full dissolution, adding 10% double antibody, filtering with microporous membrane to obtain culture solution, storing at 4 deg.C in refrigerator, and adding 10% fetal calf serum before use.
(4) Taking the NCI-H358 cells or KYSE520 cells out of the liquid nitrogen tank, immediately placing the cells in a constant-temperature water bath kettle at 37 ℃, shaking the cells to melt the cells, pouring the cells into a culture bottle, and adding a culture solution (containing 10% fetal calf serum) for dilution. Transferring the diluted culture medium into a centrifuge tube, centrifuging for 5 minutes at 1000r/min, discarding the supernatant, adding fresh culture medium, blowing, mixing, transferring into a culture bottle, and culturing in 5% CO2And cultured in an incubator at 37 ℃. When the cells are attached to the wall and spread to the bottom of the bottle, passage is started, a small amount of fresh culture medium (containing 10% fetal calf serum) is added to stop digestion, the liquid in the culture bottle is poured out, PBS is washed twice, the fresh culture medium is added to blow and beat uniformly, and the cells are evenly distributed into two culture bottles to continue culture.
(5) Taking the logarithmic phase cells, pouring off the old culture medium, adding a trypsin solution for digestion for 3 minutes, adding a fresh culture medium containing 10% fetal calf serum to stop digestion, transferring the solution to a centrifuge tube, centrifuging for 5 minutes at 1000r/min, and discarding the supernatant. Adding culture medium to prepare cell suspension, and counting cells. After counting was complete, cells were plated in 96-well plates at 5000-. Placing the 96-well plate with the cells at 37 ℃ and 5% CO2The incubator continues to culture for 24 h. Gradient dilution of drug with culture mediumThey were then added to 96 well plates at 90. mu. mol/L, 30. mu. mol/L, 10. mu. mol/L, 3.3. mu. mol/L, 1.1. mu. mol/L, 0.37. mu. mol/L, 100. mu.L per well, with three duplicate wells per concentration. Adding corresponding concentration of culture medium containing solvent into control group, adding blank culture medium with the same volume into zero setting hole, and placing in 5% CO2And incubating in an incubator at 37 ℃ for 48 h. Adding 20. mu.L of CCK8 reagent into each well, mixing well, adding 5% CO2And culturing in a 37 ℃ incubator for 1h in a dark place. The 96-well plate was then placed in a microplate reader for detection and absorbance was measured at 450 nm.
2. Data processing
Drawing a curve and calculating the inhibition rate of the drug on cells and IC50
The inhibition ratio was [ (control average OD value-experimental average OD value)/(control average OD value-blank control average OD value) ] × 100%.
3. Results of the experiment
The inhibitory activity of the compound on non-small cell lung cancer cell strain NCI-H358 cells and esophageal cancer cell strain KYSE520 cells is as follows:
compound numbering NCI-H358(IC50,μM) KYSE520(IC50,μM)
1 8.3 9.7
3 8.6 6.3
9 11.5 6.3
11 3.2 1.2
SHP099 33.4 38.1
And (4) experimental conclusion: the data show that the compounds of the embodiment of the invention have good inhibition effect on the proliferation of NCI-H358 and KYSE520 cells. Various embodiments of the present invention have novel structures and superior in vitro anti-proliferative activity compared to SHP 099.

Claims (6)

1. An aromatic aza-heptacyclic compound shown as a general formula I, and pharmaceutically acceptable salts, enantiomers, non-isomers, tautomers, solvates, polymorphs or prodrugs thereof,
Figure FDA0003526744780000011
ring A is
Figure FDA0003526744780000012
X is N or CR4
R1、R2And R4Independently hydrogen, halogen, amino;
l is a bond, C1-C2Carbon chain of (3), O or S;
when n is 1, m is 1;
when n is 2, m is 0;
R3is hydrogen, deuterium, halogen, ammoniaRadical, hydroxyl, nitro, cyano, amido, carboxyl, sulfonyl, -C (O) NR5R6、-C(O)R5、-C(O)OR5、-NR5C(O)R6、-NR5C(O)NR6R7、-NR5C(O)OR6、-NR5R6、-OC(O)R5、-OC(O)NR5R6、-SR5、-S(O)2R5、-S(O)R5、-S(O)2NR5R6、C1-C6Alkyl radical, C1-C6Haloalkyl, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C3-C7Epoxy group, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10Heteroaromatic rings, mono-heterocycles, spirocycles, bridged rings, and fused rings; wherein R is5、R6And R7Independently hydrogen, deuterium, halogen, C1-C7Alkyl radical, C1-C7Haloalkyl, C1-C7Alkoxy radical, C1-C7Haloalkoxy, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10A heteroaromatic ring.
2. The aromatic aza heptacyclic compound of claim 1, wherein:
wherein ring A is
Figure FDA0003526744780000013
X is N or CR4
R1、R2And R4Independently hydrogen, halogen, amino;
l is a bond, C1-C2Carbon chain of (3), O or S;
when n is 1, m is 1;
when n is 2, m is 0;
R3is hydrogen, deuterium, halogen, amino, hydroxyl, nitro, cyano, amido, carboxyl, sulfonyl, -C (O) NR5R6、-C(O)R5、-C(O)OR5、-NR5C(O)R6、-NR5R6、-OC(O)R5、-SR5、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C6-C10Aromatic ring heterocycle, C6-C10A heteroaromatic ring; wherein R is5And R6Independently hydrogen, deuterium, halogen, C1-C6Alkyl radical, C3-C7Cycloalkyl, C2-C6Alkenyl radical, C2-C6Alkynyl.
3. The aromatic aza-heptacyclic compound according to claims 1-2, characterized in that the aromatic aza-heptacyclic compound is any one of the following structural formulas:
Figure FDA0003526744780000021
4. a pharmaceutical composition comprising the aryloazepine heptacyclic compound according to any one of claims 1 to 3 and a pharmaceutically acceptable excipient.
5. The pharmaceutical composition of claim 3, wherein the pharmaceutical composition is in the form of a tablet, capsule, injection, or lyophilized powder.
6. Use of the aryloazepine heptacyclic compound according to any one of claims 1 to 3, the pharmaceutical composition according to claim 4 or 5 for the preparation of a therapeutic antitumor agent, as a prodrug of an antitumor agent or as an intermediate of an antitumor agent.
CN202210194719.8A 2022-03-01 2022-03-01 Aryl-containing aza-seven-membered ring compound and preparation method and application thereof Active CN114539223B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210194719.8A CN114539223B (en) 2022-03-01 2022-03-01 Aryl-containing aza-seven-membered ring compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210194719.8A CN114539223B (en) 2022-03-01 2022-03-01 Aryl-containing aza-seven-membered ring compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN114539223A true CN114539223A (en) 2022-05-27
CN114539223B CN114539223B (en) 2024-04-09

Family

ID=81662458

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210194719.8A Active CN114539223B (en) 2022-03-01 2022-03-01 Aryl-containing aza-seven-membered ring compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114539223B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115974841A (en) * 2023-01-09 2023-04-18 中国药科大学 Substituted benzoazepine amino compound and preparation method and application thereof
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113646049A (en) * 2019-04-08 2021-11-12 默克专利有限公司 Pyrimidone derivatives as SHP2 antagonists

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113646049A (en) * 2019-04-08 2021-11-12 默克专利有限公司 Pyrimidone derivatives as SHP2 antagonists

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
CN115974841A (en) * 2023-01-09 2023-04-18 中国药科大学 Substituted benzoazepine amino compound and preparation method and application thereof

Also Published As

Publication number Publication date
CN114539223B (en) 2024-04-09

Similar Documents

Publication Publication Date Title
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
JP6600365B2 (en) JAK inhibitor
CN114539223B (en) Aryl-containing aza-seven-membered ring compound and preparation method and application thereof
US10160771B2 (en) Hepatitis C virus inhibitors and uses thereof in preparation of drugs
RU2763898C9 (en) Lsd1 inhibitor, as well as its production method and its application
CA2829939C (en) Tricyclic gyrase inhibitors
CN109415341B (en) Benzotriazole-derived alpha, beta unsaturated amide compounds as TGF-beta R1 inhibitor
WO2015073767A1 (en) Substituted 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine derivatives as casein kinase 1 d/e inhibitors
NO332005B1 (en) New pyridopyrazines, their use, medicament containing at least one such compound and process for the preparation of such a medicament
CN112292374B (en) Novel phosphoinositide 3-kinase inhibitor and preparation method and application thereof
EP3157925A1 (en) Imidazo-pyridazne derivatives as casein kinase 1 delta/epsilon inhibitors
JP7168149B2 (en) Pyrazin-2(1H)-one compounds as FGFR inhibitors
KR20220066074A (en) Triazolopyrimidines as A2A/A2B inhibitors
KR20210113287A (en) Halo-allylamine compounds and uses thereof
TW202204351A (en) Compounds having a macrocyclic structure and uses thereof
JP2011529085A (en) Antiviral compounds
JP6900406B2 (en) Dihydropyrazoloazepine compounds as Akt inhibitors
CN116768861A (en) SOS1 protein degradation targeting chimeric and composition, preparation and application thereof
AU2017359819B2 (en) FGFR4 inhibitor and preparation method and use thereof
CN114478403A (en) Compound containing aromatic guanidyl as well as preparation method and application thereof
CN110283174B (en) PI3K delta inhibitor and application thereof
CN116456987A (en) Compounds useful as CDK kinase inhibitors and uses thereof
CN115974841A (en) Substituted benzoazepine amino compound and preparation method and application thereof
TW202334132A (en) Sarm1 enzyme activity inhibitor and its application
CN117003734A (en) Pyrimidine ring derivative and preparation method and application thereof

Legal Events

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