CN116348466A - Pyrazine sulfur biphenyl compound and application thereof - Google Patents

Pyrazine sulfur biphenyl compound and application thereof Download PDF

Info

Publication number
CN116348466A
CN116348466A CN202180065019.2A CN202180065019A CN116348466A CN 116348466 A CN116348466 A CN 116348466A CN 202180065019 A CN202180065019 A CN 202180065019A CN 116348466 A CN116348466 A CN 116348466A
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
added
compounds
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180065019.2A
Other languages
Chinese (zh)
Inventor
付志飞
罗妙荣
帅斌
张杨
李秋
黎健
陈曙辉
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.)
Shanghai Qilu Pharmaceutical Research and Development Centre Ltd
Original Assignee
Medshine Discovery Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medshine Discovery Inc filed Critical Medshine Discovery Inc
Publication of CN116348466A publication Critical patent/CN116348466A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • 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
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/20Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/54Improvements relating to the production of bulk chemicals using solvents, e.g. supercritical solvents or ionic liquids

Abstract

A pyrazinethion biphenyl compound and application thereof, in particular discloses a compound of a formula (II) or pharmaceutically acceptable salt thereof.

Description

Pyrazine sulfur biphenyl compound and application thereof
The present application claims priority as follows:
CN202011008309.7, filing date: 09 month 23 of 2020;
CN202110182212.6, filing date: 2021, 02, 09;
CN202110412812.7, filing date: 2021, 04, 16;
CN202110590591.2, filing date: 2021, 05, 28.
Technical Field
The invention relates to pyrazinethion biphenyl compounds and application thereof, in particular to a compound of a formula (II) or pharmaceutically acceptable salts thereof.
Background
Phosphorylation of tyrosine kinases and dephosphorylation of tyrosine phosphatases are well known in the art as signal transduction mechanisms that are common to organisms that regulate the tyrosine phosphorylation levels of intracellular proteins. Shp2 (SH 2 domain-containing protein-tyrosine phosphatase-2), which is a non-transmembrane protein tyrosine phosphatase with dephosphorylation function, is one of the important members of the Protein Tyrosine Phosphatase (PTP) family, and its molecule is encoded by the PTPN11 gene, and can positively regulate downstream signal transduction pathway by the catalytic activity of phosphatase, can also act as a phosphatase-independent adaptor protein to positively regulate, and can also act as a negative regulation under specific conditions, thereby being widely involved in the regulation of biological functions such as differentiation and migration of cells and related signal transduction processes. The PTPN11 mutation is considered to be a high risk factor for juvenile myelomonocytic leukemia (JMML) and is also considered to be a leukemia proto-oncogene due to its abnormal activation and mutation of Shp2 in different types of leukemia; shp2 is also reported to be in an overactivated state in prostate cancer, breast cancer, pancreatic cancer, gastric cancer and glioma; shp2 is used as an oncogene in lung cancer to promote the occurrence and development of tumors by regulating various mechanisms. However, in the course of liver cancer, shp2 plays a role of an oncogene under the influence of a specific environment. In conclusion, shp2 plays an important regulatory role as an important node molecule in the processes of tumorigenesis and development, and is a potential therapeutic target.
Disclosure of Invention
The present invention provides a compound of formula (II) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021119985-APPB-000001
wherein,
structural unit
Figure PCTCN2021119985-APPB-000002
Is that
Figure PCTCN2021119985-APPB-000003
E 1 Is O or CH 2
T 1 Is N or CH;
R 1 is that
Figure PCTCN2021119985-APPB-000004
R 11 、R 13 And R is 14 Each independently is C 1-3 An alkyl group;
R 12 is H or C 1-3 An alkyl group;
R 2 is F, cl, br or I;
R 3 is C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R a Substitution;
R 4 h, F, cl, br, I or C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R b Substitution;
R a and R is b F, cl, br, I, OH or NH respectively and independently 2
n is 0, 1, 2 or 3;
m is 1, 2 or 3;
when n is 0, the structural unit
Figure PCTCN2021119985-APPB-000005
Is that
Figure PCTCN2021119985-APPB-000006
Structural unit
Figure PCTCN2021119985-APPB-000007
Is that
Figure PCTCN2021119985-APPB-000008
When n is 1, 2 or 3, the structural unit
Figure PCTCN2021119985-APPB-000009
Is a structural unit
Figure PCTCN2021119985-APPB-000010
Structural unit
Figure PCTCN2021119985-APPB-000011
Is that
Figure PCTCN2021119985-APPB-000012
The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021119985-APPB-000013
wherein,
structural unit
Figure PCTCN2021119985-APPB-000014
Is that
Figure PCTCN2021119985-APPB-000015
R 1 Is that
Figure PCTCN2021119985-APPB-000016
R 11 、R 13 And R is 14 Each independently is C 1-3 An alkyl group;
R 12 is H or C 1-3 An alkyl group;
R 2 is F, cl, br or I;
R 3 is C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R a Substitution;
R 4 h, F, cl, br, I or C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R b Substitution;
R a and R is b F, cl, br, I, OH or NH respectively and independently 2
m is 1, 2 or 3.
In some aspects of the invention, R is as described above 1 Is that
Figure PCTCN2021119985-APPB-000017
The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, R is as described above 11 、R 13 And R is 14 Each independently CH 3 The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, R is as described above 12 Is H or CH 3 The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, R is as described above 3 Is CH 3 The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, R is as described above 4 F, cl, br or I, and other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, the structural units described above
Figure PCTCN2021119985-APPB-000018
Is that
Figure PCTCN2021119985-APPB-000019
Figure PCTCN2021119985-APPB-000020
The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, the structural units described above
Figure PCTCN2021119985-APPB-000021
Is that
Figure PCTCN2021119985-APPB-000022
Figure PCTCN2021119985-APPB-000023
The other variables are defined in any of the embodiments of the present invention.
In some aspects of the invention, the structural units described above
Figure PCTCN2021119985-APPB-000024
Is that
Figure PCTCN2021119985-APPB-000025
Figure PCTCN2021119985-APPB-000026
Figure PCTCN2021119985-APPB-000027
The other variables are defined in any of the embodiments of the present invention.
The invention is thatIn some aspects of (a) the above structural unit
Figure PCTCN2021119985-APPB-000028
Is that
Figure PCTCN2021119985-APPB-000029
Figure PCTCN2021119985-APPB-000030
The other variables are defined in any of the embodiments of the present invention.
Still other embodiments of the present invention are derived from any combination of the variables described above.
In some embodiments of the invention, the above-described compounds have the structure of formula (II-1)
Figure PCTCN2021119985-APPB-000031
Wherein,
R 11 、R 12 、R 2 、n、E 1 And structural unit
Figure PCTCN2021119985-APPB-000032
As defined in any of the claims of the present invention.
In some embodiments of the invention, the above-described compounds have the structure of formula (I-1) or (I-2)
Figure PCTCN2021119985-APPB-000033
Wherein,
R 11 、R 12 、R 13 、R 14 、R 2 and structural unit
Figure PCTCN2021119985-APPB-000034
As defined in any of the claims of the present invention.
In some embodiments of the invention, the above-described compounds have the structure of formula (I-1A), (I-1B), (I-2A), (II-1A) or (II-1B)
Figure PCTCN2021119985-APPB-000035
Figure PCTCN2021119985-APPB-000036
Wherein,
n、m、E 1 、T 1 、R 11 、R 12 、R 13 、R 14 、R 2 、R 3 and R is 4 As defined in any of the claims of the present invention.
In some embodiments of the invention, the above-described compounds have the structure of formula (I-1A-1), (I-2A-1), (II-1A-1) or (II-1B-1)
Figure PCTCN2021119985-APPB-000037
Wherein,
m、n、E 1 、T 1 、R 11 、R 12 、R 13 、R 14 、R 2 、R 3 and R is 4 As defined in any of the claims of the present invention.
The present invention also provides the following compounds or pharmaceutically acceptable salts thereof,
Figure PCTCN2021119985-APPB-000038
Figure PCTCN2021119985-APPB-000039
in some embodiments of the invention, the above-described compounds are
Figure PCTCN2021119985-APPB-000040
Figure PCTCN2021119985-APPB-000041
Figure PCTCN2021119985-APPB-000042
In one embodiment of the present invention, the above-mentioned compounds of the present invention are:
Figure PCTCN2021119985-APPB-000043
in one embodiment of the present invention, the above compound of the present invention is compound 004,
Figure PCTCN2021119985-APPB-000044
in one embodiment of the invention, the compound of the invention is a stereoisomer of compound 004, or a formate of said stereoisomer, which stereoisomer, or formate thereof, has a retention time of from 3.1 to 3.5 minutes, preferably from 3.2 to 3.4 minutes, more preferably about 3.3 minutes, after chiral supercritical fluid chromatography; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: chiralpak AS-3 (100 mm. Times.4.6 mm,3 μm); mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ]:40% -40%.
In one embodiment of the present invention, the above compound of the present invention is another stereoisomer of compound 004, which has a retention time of 4.3 to 4.7min, preferably 4.4 to 4.6min, more preferably about 4.5min after chiral supercritical fluid chromatography; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: chiralpak AS-3 (100 mm. Times.4.6 mm,3 μm); mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ]:40% -40%.
In one embodiment of the present invention, the above-mentioned compounds of the present invention are
Figure PCTCN2021119985-APPB-000045
In one embodiment of the invention, the compound of the invention is a stereoisomer of compound 005; the stereoisomer has a retention time of 4.8-5.2min, preferably 4.9-5.1min, more preferably about 5.0min after chiral supercritical fluid chromatography; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 48% -78% of ethanol and 7min.
In one embodiment of the invention, the compound of the invention is another stereoisomer of compound 005; the stereoisomer has a retention time of 5.6-6.0min, preferably 5.7-5.9min, more preferably about 5.8min after chiral supercritical fluid chromatography; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 48% -78% of ethanol and 7min.
In one embodiment of the invention, the compounds of the invention are
Figure PCTCN2021119985-APPB-000046
In one embodiment of the invention, the compound is a stereoisomer of compound 006, which has a retention time of 4.9 to 5.3 minutes, preferably 5.0 to 5.2 minutes, more preferably about 5.1 minutes after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-methanol ]; 40-40% of methanol.
In one embodiment of the invention, the above compound is another stereoisomer of compound 006, which has a retention time of 6.6 to 7.0min, preferably 6.7 to 6.9min, more preferably about 6.8min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-methanol ]; 40-40% of methanol.
In one embodiment of the invention, the compounds of the invention are
Figure PCTCN2021119985-APPB-000047
In one embodiment of the invention, the compound of the invention is a stereoisomer of compound 007; the stereoisomer has a retention time of 6.6-7.0min, preferably 6.7-6.9min, more preferably about 6.8min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are chromatographic columns: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 48% -78% of ethanol and 10min.
In one embodiment of the invention, the compound of the invention is another stereoisomer of compound 007; the stereoisomer has a retention time of 7.1-7.5min, preferably 7.2-7.4min, more preferably about 7.4min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are chromatographic columns: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 48% -78% of ethanol and 10min.
In one embodiment of the invention, the compounds of the invention are
Figure PCTCN2021119985-APPB-000048
In one embodiment of the invention, the compound of the invention is a stereoisomer of compound 008; the stereoisomer has a retention time of 2.7-3.1min, preferably 2.8-3.0min, more preferably about 2.9min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 45% -45% of ethanol.
In one embodiment of the invention, the compound of the invention is another stereoisomer of compound 008; the stereoisomer has a retention time of 3.6-4.0min, preferably 3.7-3.9min, more preferably about 3.9min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; 45% -45% of ethanol.
In one embodiment of the invention, the compounds of the invention are
Figure PCTCN2021119985-APPB-000049
In one embodiment of the invention, the compound of the invention is a stereoisomer of compound 010; the stereoisomer has a retention time of 2.3-2.7min, preferably 2.4-2.6min, more preferably about 2.6min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; ethanol accounts for 40-40%.
In one embodiment of the invention, the compound of the invention is another stereoisomer of compound 010; the stereoisomer has a retention time of 3.2-3.6min, preferably 3.3-3.5min, more preferably about 3.4min after chiral supercritical fluid chromatography; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-ethanol ]; ethanol accounts for 40-40%.
The second aspect of the present invention also provides a pharmaceutical composition comprising a compound as defined in any of the above claims or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
The present invention also provides a method of treating a SHP 2-associated disease in a subject in need thereof, comprising providing to the subject an effective dose of a compound as defined in any of the above-described claims, or a pharmaceutically acceptable salt or pharmaceutical composition thereof.
The invention also provides application of the compound, the isomer or the pharmaceutically acceptable salt or the pharmaceutical composition thereof in preparing medicaments for treating SHP2 related diseases.
Technical effects
The compound of the invention has better inhibition activity on protein tyrosine phosphatase SHP2, and has excellent treatment effect in SHP2 abnormal tumor patients.
Definition and description
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular term or phrase, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
The term "pharmaceutically acceptable" as used herein is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" refers to salts of the compounds of the present invention prepared from the compounds of the present invention which have the specified substituents found herein with relatively non-toxic acids or bases. When the compounds of the present invention contain relatively acidic functional groups, base addition salts may be obtained by contacting such compounds with a sufficient amount of base in pure solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salts or similar salts. When the compounds of the present invention contain relatively basic functional groups, the acid addition salts may be obtained by contacting such compounds with a sufficient amount of acid in pure solution or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and organic acid salts including acids such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and methanesulfonic acid; also included are salts of amino acids (e.g., arginine, etc.), and salts of organic acids such as glucuronic acid. Certain specific compounds of the invention contain basic and acidic functionalities that can be converted to either base or acid addition salts.
Pharmaceutically acceptable salts of the invention can be synthesized from the parent compound containing an acid or base by conventional chemical methods. In general, the preparation of such salts is as follows: prepared via reaction of these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
The compounds of the invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, and racemic mixtures and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers and mixtures thereof are included within the scope of the present invention.
Unless otherwise indicated, the term "enantiomer" or "optical isomer" refers to stereoisomers that are mirror images of each other.
Unless otherwise indicated, the term "cis-trans isomer" or "geometric isomer" is caused by the inability of a double bond or a single bond of a ring-forming carbon atom to rotate freely.
Unless otherwise indicated, the term "diastereoisomer" refers to stereoisomers of a molecule having two or more chiral centers and having a non-mirror relationship between the molecules.
Unless otherwise indicated, "(+)" means dextrorotation, "(-)" means levorotatory, "(±)" means racemization.
Unless otherwise indicated, with solid wedge bonds
Figure PCTCN2021119985-APPB-000050
And a wedge-shaped dotted bond
Figure PCTCN2021119985-APPB-000051
Representing the absolute configuration of a solid centre by straight solid keys
Figure PCTCN2021119985-APPB-000052
And straight dotted line key
Figure PCTCN2021119985-APPB-000053
Representing the relative configuration of the three-dimensional center by wavy lines
Figure PCTCN2021119985-APPB-000054
Solid key representing wedge shape
Figure PCTCN2021119985-APPB-000055
Or wedge-shaped dotted bond
Figure PCTCN2021119985-APPB-000056
Or by wave lines
Figure PCTCN2021119985-APPB-000057
Representing straight solid keys
Figure PCTCN2021119985-APPB-000058
And straight dotted line key
Figure PCTCN2021119985-APPB-000059
Unless otherwise indicated, the term "tautomer" or "tautomeric form" refers to the fact that at room temperature, different functional group isomers are in dynamic equilibrium and are capable of rapid interconversion. If tautomers are possible (e.g., in solution), chemical equilibrium of the tautomers can be reached. For example, proton tautomers (also known as proton tautomers) (prototropic tautomer) include interconversions by proton transfer, such as keto-enol isomerisation and imine-enamine isomerisation. Valence isomer (valance tautomer) includes the interconversion by recombination of some of the bond-forming electrons. A specific example of where keto-enol tautomerization is the interconversion between two tautomers of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one.
Unless otherwise indicated, the atoms with "#" or "#" are chiral atoms or chiral centers, either in (R) or (S) single enantiomer form or enriched in one enantiomer form; for example, the number of the cells to be processed,
Figure PCTCN2021119985-APPB-000060
expressed as
Figure PCTCN2021119985-APPB-000061
Unless otherwise indicated, the terms "enriched in one isomer", "enriched in one enantiomer" or "enantiomerically enriched" mean that the content of one isomer or enantiomer is less than 100% and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.
Unless otherwise indicated, the term "isomer excess" or "enantiomeric excess" refers to the difference between the relative percentages of two isomers or enantiomers. For example, where one isomer or enantiomer is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomer excess (ee value) is 80%.
Optically active (R) -and (S) -isomers and D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound of the invention is desired, it may be prepared by asymmetric synthesis or derivatization with chiral auxiliary wherein the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl), a diastereomeric salt is formed with an appropriate optically active acid or base, and then the diastereomeric resolution is carried out by conventional methods well known in the art, and then the pure enantiomer is recovered. Furthermore, separation of enantiomers and diastereomers is typically accomplished by the use of chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (e.g., carbamate formation from amine).
The compounds of the present invention may contain non-natural proportions of atomic isotopes on one or more of the atoms comprising the compounds. For example, compounds can be labeled with radioisotopes, such as tritium @, for example 3 H) Iodine-125% 125 I) Or C-14% 14 C) A. The invention relates to a method for producing a fibre-reinforced plastic composite For example, deuterium can be substituted for hydrogen to form a deuterated drug, and the bond between deuterium and carbon is stronger than the bond between normal hydrogen and carbon, so that the deuterated drug has the advantages of reducing toxic and side effects, increasing the stability of the drug, enhancing the curative effect, prolonging the biological half-life of the drug and the like compared with the non-deuterated drug. All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
The term "optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is substituted with a substituent, which may include deuterium and variants of hydrogen, provided that the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxygen (i.e., =o), it means that two hydrogen atoms are substituted. Oxygen substitution does not occur on the aromatic group.
The term "optionally substituted" means that the substituents may or may not be substituted, and the types and numbers of substituents may be arbitrary on the basis that they can be chemically achieved unless otherwise specified.
When any variable (e.g., R) occurs more than once in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 0 to 2R, the group may optionally be substituted with up to two R's, and R's in each case have independent options. Furthermore, combinations of substituents and/or variants thereof are only permissible if such combinations result in stable compounds.
When one of the variables is selected from a single bond, the two groups to which it is attached are indicated as being directly linked, e.g., when L in A-L-Z represents a single bond, it is indicated that the structure is actually A-Z.
When a substituent is absent, it is meant that the substituent is absent, e.g., X in A-X is absent, meaning that the structure is actually A. When the listed substituents do not indicate which atom is attached to the substituted group, such substituents may be bonded through any atom thereof, for example, a pyridyl group may be attached to the substituted group as a substituent through any carbon atom on the pyridine ring.
When the exemplified linking group does not indicate its linking direction, its linking direction is arbitrary, for example,
Figure PCTCN2021119985-APPB-000062
The linking group L is-M-W-, in which case-M-W-may be a group formed by linking the rings A and B in the same direction as the reading order from left to right
Figure PCTCN2021119985-APPB-000063
The ring A and the ring B may be connected in a direction opposite to the reading order from left to right
Figure PCTCN2021119985-APPB-000064
Combinations of such linking groups, substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
Unless otherwise specified, when a group has one or more bondable sites, any one or more of the sites of the group may be bonded to other groups by chemical bonds. When the bond is not located and an H atom is present in the ligatable site, then the bond is attached to the siteThe number of H atoms of (2) is correspondingly reduced with the number of chemical bonds connected to the group with corresponding valence. The chemical bond of the site and other groups can be a straight solid line bond
Figure PCTCN2021119985-APPB-000065
Straight dotted line key
Figure PCTCN2021119985-APPB-000066
Or wave lines
Figure PCTCN2021119985-APPB-000067
And (3) representing. For example-OCH 3 The straight solid line bond in (a) represents the connection to other groups through the oxygen atom in the group;
Figure PCTCN2021119985-APPB-000068
the straight dashed bonds in (a) represent the attachment to other groups through both ends of the nitrogen atom in the group;
Figure PCTCN2021119985-APPB-000069
the wavy line means that the carbon atoms at positions 1 and 2 in the phenyl group are attached to other groups;
Figure PCTCN2021119985-APPB-000070
It means that any of the ligatable sites on the piperidinyl group may be attached to other groups by 1 chemical bond, including at least
Figure PCTCN2021119985-APPB-000071
These 4 connection modes, even though H atom is drawn on-N-, are
Figure PCTCN2021119985-APPB-000072
Still include
Figure PCTCN2021119985-APPB-000073
The group of this linkage is only when 1 chemical bond is linked, the H at this site will be correspondingly reduced by 1 to the corresponding monovalent piperidinyl group.
Unless otherwise specified, the term "C 1-3 Alkyl "is used to denote a straight or branched saturated hydrocarbon group consisting of 1 to 3 carbon atoms. The C is 1-3 Alkyl includes C 1-2 And C 2-3 Alkyl groups, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C (C) 1- 3 Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
Unless otherwise specified, C n-n+m Or C n -C n+m Comprising any one of the specific cases of n to n+m carbons, e.g. C 1-12 Comprises C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 And C 12 Also included is any one of the ranges n to n+m, e.g. C 1-12 Comprises C 1-3 、C 1-6 、C 1-9 、C 3-6 、C 3-9 、C 3-12 、C 6-9 、C 6-12 And C 9-12 Etc.; similarly, n-membered to n+m-membered means that the number of atoms on the ring is n to n+m, for example, 3-12 membered ring includes 3-membered ring, 4-membered ring, 5-membered ring, 6-membered ring, 7-membered ring, 8-membered ring, 9-membered ring, 10-membered ring, 11-membered ring, and 12-membered ring, and any one of n to n+m is also included, for example, 3-12-membered ring includes 3-6-membered ring, 3-9-membered ring, 5-6-membered ring, 5-7-membered ring, 6-8-membered ring, 6-10-membered ring, and the like.
The term "leaving group" refers to a functional group or atom that may be substituted with another functional group or atom by a substitution reaction (e.g., a nucleophilic substitution reaction). For example, representative leaving groups include triflate; chlorine, bromine, iodine; sulfonate groups such as methanesulfonate, toluenesulfonate, p-bromophenylsulfonate, p-toluenesulfonate and the like; acyloxy groups such as acetoxy, trifluoroacetoxy, and the like.
The term "protecting group" includes, but is not limited to, "amino protecting group", "hydroxy protecting group" or "mercapto protecting group". The term "amino protecting group" refers to a protecting group suitable for preventing side reactions at the amino nitrogen position. Representative amino protecting groups include, but are not limited to: a formyl group; acyl groups such as alkanoyl (e.g., acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl groups such as t-butoxycarbonyl (Boc); arylmethoxycarbonyl groups such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl groups such as benzyl (Bn), trityl (Tr), 1-bis- (4' -methoxyphenyl) methyl; silyl groups such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like. The term "hydroxy protecting group" refers to a protecting group suitable for use in preventing side reactions of a hydroxy group. Representative hydroxyl protecting groups include, but are not limited to: alkyl groups such as methyl, ethyl and t-butyl; acyl groups such as alkanoyl (e.g., acetyl); arylmethyl groups such as benzyl (Bn), p-methoxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (benzhydryl, DPM); silyl groups such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like.
Unless otherwise indicated, conditions for supercritical fluid chromatography, such as supercritical fluid chromatography (column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm), mobile phase: [0.1% ammonia-ethanol ]; ethanol%: 48% -78%,7min, where 7min represents the time required for the concentration of ethanol to increase from 48% to 78%.
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, embodiments formed by combining with other chemical synthetic methods, and equivalent alternatives well known to those skilled in the art, preferred embodiments including but not limited to the examples of the present invention.
The compounds of the present invention may be structured by conventional methods well known to those skilled in the art, and if the present invention relates to the absolute configuration of a compound, the absolute configuration may be confirmed by conventional means in the art. For example, single crystal X-ray diffraction (SXRD), the grown single crystal is collected from diffraction intensity data using a Bruker D8vent diffractometer, and the light source is cukα radiation, scanning:
Figure PCTCN2021119985-APPB-000074
after scanning and collecting the relevant data, the absolute configuration can be confirmed by further analyzing the crystal structure by a direct method (Shellxs 97).
The solvent used in the present invention is commercially available. The invention adopts the following abbreviations: aq represents water; eq represents equivalent, equivalent; naCNBH 3 Represents sodium cyanoborohydride; r.t. stands for room temperature; mp represents the melting point; DCM represents dichloromethane; meOH represents methanol; SFC represents supercritical fluid chromatography; MMS indicates intrinsic liver clearance.
Compounds are either prepared according to the general nomenclature of the art or are used
Figure PCTCN2021119985-APPB-000075
Software naming, commercial compounds are referred to by vendor catalog names.
Detailed Description
The following examples illustrate the invention in detail, but are not meant to limit the invention in any way. The invention has been described in detail herein, and specific embodiments thereof are disclosed, it will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the invention without departing from the spirit and scope of the invention, and still fall within the scope of the invention.
Example 1
Figure PCTCN2021119985-APPB-000076
Figure PCTCN2021119985-APPB-000077
Step 1: synthesis of Compound 001-2:
compound 001-1 (5 g,24.22mmol,1 eq) was dissolved in acetonitrile (50 mL), diiodomethane (9.73 g,36.33mmol,2.93mL,1.5 eq) was added, tert-butyl nitrite (3.75 g,36.33mmol,4.32mL,1.5 eq) was slowly added at 0deg.C, the mixture was stirred while heating to 20deg.C, and then stirred while heating to 80deg.C for 1 hour. The reaction solution was concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 80mL petroleum ether, filtered, and finally the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. Column chromatography: separating the crude product by column chromatography (petroleum ether) to obtain compound 001-2.
Step 2: synthesis of Compound 001-3:
compounds 001-2 (5 g,15.76mmol,1 eq) and dimethylphosphine (1.23 g,15.76mmol,1 eq) were dissolved under nitrogen to dioxane (50 mL), tris (dibenzylideneacetone) dipalladium (721.38 mg, 787.78. Mu. Mol,0.05 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (455.82 mg, 787.78. Mu. Mol,0.05 eq), N, N-diisopropylethylamine (5.09 g,39.39mmol,6.86mL,2.5 eq) were added in one portion, and then stirred at 60℃for 16 hours. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 30mL of water and 50mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (30 mL ). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. High performance liquid chromatography (column: phenomenex Genimi NX C18150. Times.40 mm. Times.5 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 22-32 percent for 10 min) to obtain the compound 001-3.MS (ESI) m/z 269.0[ M+H ]] +
Step 3: synthesis of Compounds 001-4:
under the protection of nitrogen, the compound 001-3 (2.3 g,8.60mmol,1 eq) and the compound 001-3A (2.25 g,10.32mmol,1.2 eq) were dissolved in dioxane (25 mL), N-diisopropylethylamine (3.33 g,25.80mmol,4.49mL,3 eq), tris (dibenzylideneacetone) dipalladium (787.38 mg, 859.86. Mu. Mol,0.1 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (497.53 mg, 859.86. Mu. Mol,0.1 eq) were added in one portion, and then stirred at 110℃for 5 hours. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 50mL of water and 50mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (50 mL,30 mL). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. The crude product was separated by column chromatography (0-10% methanol in dichloromethane). To give compound 001-4.MS (ESI) m/z 405.2[ M+H ] ] +
Step 4: synthesis of Compounds 001-5:
compound 001-4 (0.5 g,1.23mmol,1 eq) was dissolved in tetrahydrofuran (5 mL), -potassium tert-butoxide (1M, 1.85mL,1.5 eq) dissolved in tetrahydrofuran (5 mL) was added (dropwise) at-78deg.C, and the mixture was stirred at-78deg.C for 1 hour. And directly spin-drying the reaction solution to obtain a crude product. The crude product obtained was used directly in the next reaction without further purification. To give compound 001-5.MS (ESI) m/z 221.0[ M+H ]] +
Step 5: synthesis of Compounds 001-6:
compounds 001-5 (270 mg,1.22mmol,1 eq) and 001-5A (382.59 mg,1.84mmol,1.5 eq) were dissolved under nitrogen to dioxane (3 mL), tris (dibenzylideneacetone) dipalladium (112.05 mg, 122.36. Mu. Mol,0.1 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (70.80 mg, 122.36. Mu. Mol,0.1 eq), N, N-diisopropylethylamine (474.43 mg,3.67mmol, 639.40. Mu.L, 3 eq) were added in one portion, and then stirred for 5 hours at 110 ℃. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 30mL of water and 30mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (30 mL ). MergingThe organic phase was washed once with saturated sodium chloride solution (30 mL), and finally the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was isolated by column chromatography (0-10% methanol in dichloromethane) (TLC detection DCM: meoh=10:1). To give the compound 001-6.MS (ESI) m/z 347.9[ M+H ] ] +
Step 6: synthesis of Compounds 001-7:
compound 001-6 (40 mg, 114.88. Mu. Mol,1 eq) and compound 001-6A (47.29 mg, 172.32. Mu. Mol,1.5 eq) were dissolved in a mixed solvent of dimethylacetamide (2 mL) and water (2 mL), and potassium carbonate (79.39 mg, 574.40. Mu. Mol,5 eq) was added in one portion, followed by stirring at a temperature of 100℃for 16 hours. And directly spin-drying the reaction solution to obtain a crude product. The obtained compound 001-7 was used directly in the next reaction. MS (ESI) m/z 586.3[ M+H ]] +
Step 7: synthesis of Compound 001:
compounds 001-7 (45 mg, 76.77. Mu. Mol,1 eq) were dissolved in hydrochloric acid/dioxane (4M, 479.83. Mu.L, 25 eq) and the mixture was stirred at 20℃for 1 hour. And directly spin-drying the reaction solution to obtain a crude product. High performance liquid chromatography (column: phenomenex Gemini-NX 18X 30mm X3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 0-30 percent and 7 min) to obtain formate of the compound 001. MS (ESI) m/z 482.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 1.32-1.35(m,3H),1.71(br d,J=12.80Hz,1H),1.80-1.90(m,3H),1.95(s,3H),1.99(s,3H),3.09-3.25(m,2H),3.41(br d,J=4.02Hz,1H),3.87(d,J=9.29Hz,1H),4.00(d,J=9.03Hz,1H),4.18-4.38(m,3H),6.95(d,J=7.53Hz,1H),7.36(m,1H),7.62(s,1H),7.75(m,1H)。
Example 2
Figure PCTCN2021119985-APPB-000078
Step 1: synthesis of Compound 002-1:
compound 001-1 (200 mg, 968.68. Mu. Mol,1 eq) was dissolved in acetonitrile (5 mL) under nitrogen, and two additions were made at a timeMethyl iodide (73.00 mg, 774.94. Mu. Mol, 69.52. Mu.L, 0.8 eq) was warmed to 60℃and tert-butyl nitrite (149.83 mg,1.45mmol, 172.82. Mu.L, 1.5 eq) was slowly added, followed by stirring for 1 hour at 80 ℃. The reaction solution was concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 80mL petroleum ether, filtered, and finally the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. Column chromatography: the crude product was isolated by column chromatography (petroleum ether=100%). Compound 002-1 was obtained. 1 H NMR(400MHz,CDCl 3 )δppm 7.33(dd,J=7.15,2.13Hz,1H),6.95-7.05(m,2H),2.40(s,3H)。
Step 2: synthesis of compound 002-2:
compound 002-1 (2.8 g,11.79mmol,1 eq) was dissolved in methanol (40 mL), and amine acetate (1.82 g,23.57mmol,2 eq) and iodobenzene acetate (9.49 g,29.47mmol,2.5 eq) were added in one portion and the mixture was stirred at 20℃for 1 hour. And directly spin-drying the reaction solution to obtain a crude product. The crude product is separated by column chromatography (0-10% methanol in dichloromethane) to give compound 002-2.MS (ESI) m/z 269.7[ M+H ]] +
Step 3: synthesis of compound 002-3:
compound 002-2 (1 g,3.72mmol,1 eq) and compound 001-3A (894.37 mg,4.10mmol,1.1 eq) were dissolved under nitrogen to dioxane (10 mL), diisopropylethylamine (1.44 g,11.17mmol,1.95mL,3 eq), tris (dibenzylideneacetone) dipalladium (340.98 mg, 372.36. Mu. Mol,0.1 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (215.45 mg, 372.36. Mu. Mol,0.1 eq) were added in one portion, and then stirred for 5 hours at 110 ℃. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 50mL of water and 50mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (50 mL,30 mL). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. Column chromatography: the crude product was separated by column chromatography (0-10% methanol in dichloromethane). Compound 002-3 was obtained. MS (ESI) m/z 406.1[ M+H ] ] +
Step 4: synthesis of compound 002-4:
compound 002-4 (100 mg, 246.30. Mu. Mol,1 eq) was dissolved in tetrahydrofuran (1.23 mL), potassium tert-butoxide (1M, 1.23mL, eq) was added at-78deg.C in one portion, and the mixture was stirred at-78deg.C for 1 hour under nitrogen. And directly spin-drying the reaction solution to obtain a crude product. The crude product obtained was used directly in the next reaction without further purification. Compound 002-4 was obtained. MS (ESI) m/z 221.9[ M+H ] +.
Step 5: synthesis of compound 002-5:
compound 002-4 (50 mg, 225.50. Mu. Mol,1 eq) and compound 001-5A (70.51 mg, 338.25. Mu. Mol,1.5 eq) were dissolved in dioxane (3 mL) under nitrogen, diisopropylethylamine (87.43 mg, 676.51. Mu. Mol, 117.84. Mu.L, 3 eq), tris (dibenzylideneacetone) dipalladium (20.65 mg, 22.55. Mu. Mol,0.1 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (13.05 mg, 22.55. Mu. Mol,0.1 eq) were added in one portion, and then stirred at 110℃for 5 hours. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 30mL of water and 30mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (30 mL. Times.3). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. The crude product was separated by column chromatography (0-10% methanol in dichloromethane). Compound 002-5 was obtained. MS (ESI) m/z 348.9[ M+H ] +.
Step 6: synthesis of Compound 002-6:
under the protection of nitrogen, compound 002-5 (50 mg, 143.16. Mu. Mol,1 eq) and compound 001-6A (39.29 mg, 143.16. Mu. Mol,1 eq) were dissolved in dimethylacetamide (2 mL), water (2 mL) and potassium carbonate (59.36 mg, 429.48. Mu. Mol,3 eq) was added in one portion, followed by stirring at 100℃for 16 hours. The reaction solution was directly spin-dried to obtain compound 002-6.MS (ESI) m/z 587.1[ M+H ] +.
Step 7: synthesis of compound 002:
compound 002-6 (50 mg, 85.15. Mu. Mol,1 eq) was dissolved in hydrochloric acid/methanol (4M, 532.17. Mu.L, 25 eq) and the mixture was stirred at 20℃for 1 hour. And directly spin-drying the reaction solution to obtain a crude product. High performance liquid chromatography (column: phenomenex Gemini-NX 18X 30mm X3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 0-30 percent, 7 min). Obtaining the compound002 formate. The formate of 002 was added to ethyl acetate (30 mL), washed with saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was dried by spin-drying to give compound 002.MS (ESI) m/z 483.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 7.96(d,J= 7.75Hz,1H),7.63(s,1H),7.37(t,J=8.00Hz,1H),6.97(d,J=8.50Hz,1H),4.63(br s,2H),4.20-4.37(m,4H),3.99(m,1H),3.87(m,1H),3.36(s,3H),3.10-3.24(m,1H),1.77-1.91(m,3H),1.70(m,1H),1.32(m,3H)。
Example 3
Figure PCTCN2021119985-APPB-000079
Step 1: synthesis of Compound 003-1:
compound 002-2 (1.9 g,7.07mmol,1 eq) was dissolved in N, N-dimethylformamide (20 mL) under nitrogen, sodium hydride (424.45 mg,10.61mmol,60% purity, 1.5 eq) was added in one portion at 0deg.C, the mixture was stirred at 0deg.C for 10min, and then methyl iodide (2.01 g,14.15mmol, 880.86. Mu.L, 2 eq) was added and the mixture was warmed to 25deg.C and stirred for hours. The reaction solution was dropwise added to 10mL of ice water, diluted with 50mL of ethyl acetate, separated, washed with aqueous ethyl acetate (50 mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by spinning the filtrate under reduced pressure. The crude product was separated by column chromatography (petroleum ether: ethyl acetate=0-50%) to give compound 003-1.MS (ESI) m/z 283.9[ M+H ] ] +
Step 2: synthesis of Compound 003-2:
compound 003-1 (1.5 g,5.31mmol,1 eq) and compound 001-3A (1.27g,5.84mmol,1.1 eq) were dissolved in dioxane (20 mL) under nitrogen, diisopropylethylamine (2.06 g,15.92mmol,2.77mL,3 eq), tris (dibenzylideneacetone) dipalladium (486.08 mg, 530.81. Mu. Mol,0.1 eq), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (307.14 mg, 530.81. Mu. Mol,0.1 eq) were added in one portion, and then stirred at 110℃for 5 hours. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 50mL of water and 50mL of ethyl acetate, and the layers were separated by extraction whileThe aqueous phase was extracted three times with ethyl acetate (50 mL,30 mL). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. Column chromatography: the crude product was separated by column chromatography (0-10% methanol in dichloromethane). Compound 003-2 is obtained. MS (ESI) m/z 420.0[ M+H ]] +
Step 3: synthesis of Compound 003-3:
compound 003-2 (100 mg, 238.08. Mu. Mol,1 eq) was dissolved in tetrahydrofuran (1 mL), potassium tert-butoxide (1M, 1.19mL,5 eq) was added at 78℃in one portion, and the mixture was stirred at-78℃for 1 hour under nitrogen. And directly spin-drying the reaction solution to obtain the compound 003-3.MS (ESI) m/z 235.9[ M+H ] +.
Step 4: synthesis of Compound 003-4:
compound 003-3 (56 mg, 237.54. Mu. Mol,1 eq) and compound 001-5A (74.27 mg, 356.30. Mu. Mol,1.5 eq) were dissolved in dioxane (3 mL) under nitrogen, diisopropylethylamine (92.10 mg, 712.61. Mu. Mol, 124.12. Mu.L, 3 eq), tris (dibenzylideneacetone) dipalladium (21.75 mg, 23.75. Mu. Mol,0.1 eq), 4, 5-bisdiphenylphosphine-9, 9-dimethylxanthene (13.74 mg, 23.75. Mu. Mol,0.1 eq) were added in one portion, and then stirred at 110℃for 5 hours. The reaction solution was cooled to 20℃and concentrated under reduced pressure at 43 ℃. The concentrate was dissolved in 30mL of water and 30mL of ethyl acetate, the layers were separated by extraction, and the aqueous phase was extracted three times with ethyl acetate (30 mL. Times.3). The organic phases were combined, washed once with saturated sodium chloride solution (30 mL) and finally the organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. The crude product is separated by column chromatography (0-10% methanol in dichloromethane) to obtain compound 003-4.MS (ESI) m/z 362.9[ M+H ]] +
Step 5: synthesis of Compound 003-5:
compound 003-4 (85 mg, 233.98. Mu. Mol,1 eq) and compound 001-6A (77.05 mg, 280.77. Mu. Mol,1.2 eq) were dissolved in dimethylacetamide (2 mL), water (2 mL) under nitrogen, and potassium carbonate (97.01 mg, 701.93. Mu. Mol,3 eq) was added in one portion, followed by stirring at a temperature of 100℃for 16 hours. And directly spin-drying the reaction solution to obtain a crude product. Compound 003-5 was obtained. MS (ESI) m/z 601.1[ M+H ] ] +
Step 6: synthesis of compound 003:
compound 003-5 (50 mg, 83.16. Mu. Mol,1 eq) was dissolved in hydrogen chloride/methanol (4M, 20.79. Mu.L, 1 eq) and the mixture was stirred at 20℃for 1 hour. And directly spin-drying the reaction solution to obtain a crude product. High performance liquid chromatography (Phenomenex Gemini-NX C18 75X 30mm X3 μm; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 5 to 35 percent, 7 minutes) and separating and purifying to obtain formate of the compound 003. Formate of 003 was added to ethyl acetate (30 mL), washed with saturated sodium hydrogencarbonate solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was dried by spin-drying to obtain compound 003.MS (ESI) m/z 497.0[ M+H ] ]+1 H NMR(400MHz,CD 3 OD)δppm 7.92(dd,J=7.91,1.38Hz,1H), 7.63(s,1H),7.40(t,J=7.91Hz,1H),7.00(dd,J=8.03,1.51Hz,1H),4.25-4.38(m,3H),4.02(d,J=9.29Hz,1H),3.90(d,J=9.29Hz,1H),3.44(d,J=4.27Hz,1H),3.36(s,3H),3.07-3.24(m,2H),2.57(s,3H),1.79-1.90(m,3H),1.68-1.76(m,1H),1.35(s,3H)。
Example 4
Figure PCTCN2021119985-APPB-000080
Figure PCTCN2021119985-APPB-000081
Step 1: synthesis of compound 004-2:
compound 004-1 (5.0 g,19.43mmol,1 eq) was dissolved in anhydrous tetrahydrofuran (60 mL) under nitrogen, and after three nitrogen substitutions, the temperature was reduced to-78 ℃. To the reaction mixture was slowly added dropwise a tetrahydrofuran solution of lithium diisopropylamide (2.0 m,10.69ml,1.1 eq) and the mixture was stirred at-78 ℃ for 1 hour, after which a tetrahydrofuran solution of compound 004-2A was slowly added dropwise to the system and reacted at-78 ℃ for 30 minutes, and then the reaction system was slowly warmed to-25 ℃ and reacted for 15 hours. Quenched with 100mL of saturated ammonium chloride solution after the reaction The ethyl acetate was extracted three times (50 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was separated by column chromatography (petroleum ether: ethyl acetate=0-10%) to give compound 004-2.MS (ESI) m/z 388.2[ M+Na ]] +
Step 2: synthesis of compound 004-3:
compound 004-2 (6.91 g,18.91mmol,1 eq) was dissolved in dioxane (80 mL) and methanol (32 mL) under nitrogen, and after further addition of aqueous sodium hydroxide (6M, 16mL,5.08 eq), the temperature was raised to 100deg.C and the reaction was refluxed for 15 hours. Cooling to room temperature after the reaction is finished, decompressing and removing the organic solvent, then regulating the pH value to 3-4 by using dilute hydrochloric acid (1.0M), filtering, washing a filter cake by using water, dissolving the washed filter cake in ethyl acetate again, drying by using anhydrous sodium sulfate, and spin-drying to obtain a compound 004-3, MS (ESI) M/z is 360.1[ M+Na ]] +
Step 3: synthesis of compound 004-4:
compound 004-4 (6.10 g,18.08mmol,1 eq) and polyphosphoric acid (40 ml,1.0 eq) were added to a single vial under nitrogen and reacted at 120 ℃ for 1 hour. After the reaction was completed, the reaction mixture was cooled to room temperature, quenched by pouring into ice water, and the pH was slowly adjusted to 9 with an aqueous sodium hydroxide solution (6M) under an ice bath. Ethyl acetate was extracted three times (50 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation and the crude product was dissolved in dichloromethane (100 mL). The reaction mixture was reacted at 25℃for 2 hours after addition of di-tert-butyl dicarbonate (6.12 g,28.05mmol,6.44mL,3.0 eq) and triethylamine (5.68 g,56.10mmol,7.81mL,6.0 eq). After the completion of the reaction, water was added, the mixture was separated, the aqueous phase was extracted with ethyl acetate (50 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give Compound 004-4.MS (ESI) m/z 342.1[ M+Na ] +
Step 4: synthesis of compound 004-5:
under the protection of nitrogen, compound 004-4 (2 g,6.26mmol,1 eq) and compound 004-5A (2.28 g,18.79mol,3.0 eq) were added to a single-necked flask followed by tetraethyltitanate (5 mL) at 100deg.C for 18 hours under reflux. After the reaction was completed, the mixture was cooled to room temperature and poured intoQuench in ice water, add ethyl acetate (50 mL) and stir for 1 hour, separate the liquid and extract the aqueous phase with ethyl acetate (50 mL ×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered and the solvent was removed by rotary evaporation. Column chromatography: the crude product was separated by column chromatography (petroleum ether: ethyl acetate=0-20%) to give compound 004-5.MS (ESI) m/z 445.1[ M+Na] +
Step 5: synthesis of compound 004-6:
compound 004-5 (2.54 g,6.01mmol,1 eq) was dissolved in tetrahydrofuran (25 mL) under nitrogen, cooled to-20deg.C and sodium borohydride (45 mg,12.02mmol,2.0 eq) was added. The reaction system was gradually returned to 25℃for 12 hours. After the reaction, the reaction mixture was quenched with water in an ice bath, extracted with ethyl acetate (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give compound 004-6.MS (ESI) m/z 447.1[ M+Na ]] +
Step 6: synthesis of compound 004-7:
Compound 004-6 (34 mg, 80. Mu. Mol,1 eq) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (91.2 mg, 800. Mu. Mol, 59.23. Mu.L, 10 eq) was added. The reaction mixture reacts for 2 hours at 25 ℃, then potassium carbonate is added to neutralize the reaction system to be neutral, and the solvent is dried by spinning to obtain the compound 004-7.MS (ESI) m/z 325.1[ M+H ]] +
Step 7: synthesis of compound 004-8:
compound 004-7 (25.9 mg, 80. Mu. Mol,1 eq) and compound 002-5 (27.6 mg, 80. Mu. Mol,1 eq) were dissolved in N, N-dimethylacetamide (4 mL) and water (4 mL), and reacted at 100℃for 48 hours after adding potassium carbonate (57.38 mg, 415. Mu. Mol,5 eq). After completion of the reaction, the mixture was cooled to room temperature, water (10 mL) was added, the mixture was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was isolated by column chromatography (dichloromethane: methanol=0-10%) to give compound 004-8.MS (ESI) m/z 637.1[ M+H ]] +
Step 8: synthesis of compound 004:
compound 004-8 (23 mg, 36.1. Mu. Mol,1 eq) was dissolved in methanol (2 mL) and a hydrochloric acid methanol solution (4M, 530. Mu.L, 58.5 eq) was added). The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain crude product, and the crude product is sent to a high performance liquid chromatography (chromatographic column: phenomenex Gemini-NX C18: 75 x 30mm x 3 μm; mobile phase: water (0.225% formic acid) -acetonitrile) ]The method comprises the steps of carrying out a first treatment on the surface of the 48% -78% of acetonitrile, 7 min) to obtain formate of the compound 004. The formate of 004 was added to ethyl acetate (30 mL), washed with saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, filtered, and the filtrate was dried by spin-drying to give compound 004.MS (ESI) m/z 533.2[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δppm 7.95(dd,J=7.78,1.25Hz,1H),7.68(s,1H),7.28(m,2H),7.04(dd,J=7.91,1.38Hz,1H),6.91–6.96(m,2H),4.88(s,2H),4.21(m,2H),3.98(s,1H),3.34(s,3H),3.17–3.27(m,2H),3.05-3.14(d,J=15.56Hz,1H),2.75(d,J=15.56Hz,1H),1.30-1.34(m,2H)。
Step 9: synthesis of compounds 004-8A and 004-8B:
separating compound 004-8 by chiral supercritical fluid chromatography (chromatographic column: chiralpak AS-3 (100 mm. Times.4.6 mm,3 μm), mobile phase: carbon dioxide, [0.05% triethylamine, ethanol ]]40% -40%) to obtain two isomers, wherein isomer 1 is 004-8A (retention time 2.557 min). MS (ESI) m/z 637.1[ M+H ]] + . Isomer 2 was 004-8B (retention time 3.028 min), MS (ESI) m/z 637.1[ M+H ]] +
Step 10: synthesis of compound 004A:
compound 004-8A (23 mg, 36.1. Mu. Mol,1 eq) was dissolved in methanol (2 mL), and hydrochloric acid methanol solution (4M, 530. Mu.L, 58.5 eq) was added. The reaction mixture is directly dried by spin drying after reacting for 1 hour at 25 ℃ to obtain crude product, and the crude product is sent to high performance liquid chromatography (chromatographic column: welch Xtimate C18 is 150 mm by 25mm by 5 mu m; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile%: 10% -40%,7 min) to obtain formate of compound 004A. MS (ESI) m/z 533.2[ M+H ] ] +
1 H NMR(400MHz,CD 3 OD)δppm 7.95(dd,J=7.78,1.25Hz,1H),7.64(s,1H),7.50dd,J=8.28,5.27Hz,1H),7.38(t,J=7.91,1H),7.15–7.02(m,2H),6.99(dd,J=8.03,1.51Hz,1H),4.45–4.25(m,3H),3.36(s,3H),3.32–3.22(m,2H),3.20–3.10(m,2H),2.00-1.50(m,4H)。
1 H NMR(400MHz,CDCl 3 )δppm 7.95(dd,J=7.78,1.25Hz,1H),7.68(s,1H),7.28(m,2H),7.04(dd,J=7.91,1.38Hz,1H),6.91–6.96(m,2H),4.88(s,2H),4.21(m,2H),3.98(s,1H),3.34(s,3H),3.17–3.27m,2H),3.05-3.14(d,J=15.56Hz,1H),2.75(d,J=15.56Hz,1H),1.30-1.34(m,2H)。
004A formate is sent to chiral supercritical fluid chromatography for analysis: (column: chiralpak AS-3 (100 mm. Times. 4.6mm,3 μm; mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ].
Step 11: synthesis of compound 004B:
compound 004-8B (23 mg, 36.1. Mu. Mol,1 eq) was dissolved in methanol (2 mL), and hydrochloric acid methanol solution (4M, 530. Mu.L, 58.5 eq) was added. The reaction mixture is directly dried by spin drying after reacting for 1 hour at 25 ℃ to obtain crude product, and the crude product is sent to high performance liquid chromatography (chromatographic column: welch Xtimate C18 is 150 mm by 25mm by 5 mu m; mobile phase: [ water (0.225% formic acid) -acetonitrile)]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 10 to 70 percent, 7 minutes) and separating to obtain formate of the compound 004B. MS (ESI) m/z 533.2[ M+H ]] +
1 H NMR(400MHz,CD 3 OD)δppm 7.95(dd,J=7.78,1.25Hz,1H),7.64(s,1H),7.54(dd,J=8.53,5.02Hz,1H),7.38(t,J=7.91,1H),7.18–7.05(m,2H),6.99(dd,J=8.03,1.25Hz,1H),4.45–4.25(m,3H),3.36(s,3H),3.32–3.22(m,2H),3.22–3.15(m,2H),2.00–1.50(m,4H)。
1 H NMR(400MHz,CDCl 3 )δppm 7.95(dd,J=7.78,1.25Hz,1H),7.68(s,1H),7.28(m,2H),7.04(dd,J=7.91,1.38Hz,1H),6.91–6.96(m,2H),4.88(s,2H),4.21(m,2H),3.98(s,1H),3.34(s,3H),3.17–3.27(m,2H),3.05-3.14(d,J=15.56Hz,1H),2.75(m,1H),1.30-1.34(m,2H)。
004B formate is sent to chiral supercritical fluid chromatography for analysis: (column: chiralpak AS-3 (100 mm. Times. 4.6mm,3 μm; mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ].
Example 5
Figure PCTCN2021119985-APPB-000082
Step 1: synthesis of Compound 005-2:
compound 005-1 (500 mg,1.17mmol,1 eq) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (3.0 mL) was added. The reaction mixture reacts for 2 hours at 25 ℃, then potassium carbonate is added to neutralize the reaction system to be neutral, and the solvent is dried by spin to obtain the compound 005-2.MS (ESI) m/z 327.1[ M+H ] ] +
Step 2: synthesis of Compound 005-3:
compound 005-2 (380 mg,1.16mmol,1 eq) and compound 002-5 (405 mg,1.16mmol,1 eq) were dissolved in N, N-dimethylacetamide (10 mL) and water (10 mL), and after addition of potassium carbonate (995 mg,5.8mmol,5 eq) the mixture was reacted at 100℃for 48 hours. After completion of the reaction, the mixture was cooled to room temperature, water (10 mL) was added, the mixture was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was isolated by column chromatography (dichloromethane: methanol=0-10%) to give compound 005-3.MS (ESI) m/z 639.1[ M+H ]] +
Step 3: synthesis of Compound 005:
compound 005-3 (250 mg, 391. Mu. Mol,1 eq) was dissolved in methanol (10 mL) and hydrochloric acid in methanol (4M, 2.5mL, 58.5 eq) was added. The reaction mixture is directly spin-dried to obtain crude product after reacting for 1 hour at 25 ℃, and the crude product is sent to supercritical fluid chromatography (chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm) and mobile phase: [0.1% ammonia-ethanol ]]The method comprises the steps of carrying out a first treatment on the surface of the 48% -78% of ethanol and 7min. Compound 005A (5.032 min) and compound 005B (5.823 min) were isolated. Compound 005A: MS (ESI) m/z 535.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD) delta ppm 7.94 (dd, j=7.78, 1.25hz, 1H), 7.65 (s, 1H) 7.38-7.34 (m, 2H), 7.00-6.97 (m, 1H), 6.91-6.55 (m, 2H), 4.48-4.40 (m, 1H), 4.37-4.26 (m, 1H), 4.09 (s, 1H), 3.45-3.37 (m, 2H), 3.35 (s, 3H), 1.96-1.93 (m, 2H), 1.85-1.82 (m, 2H). Compound 005B: MS (ESI) m/z 535.1[ M+H ] ] +1 H NMR(400MHz,CD 3 OD)δppm 7.94(dd,J=7.78,1.25Hz,1H),7.65(s,1H),7.38–7.34(m,2H),7.00–6.97(m,1H),6.91–6.66–6.55(m,2H),4.48–4.40(m,1H),4.37–4.26(m,1H),4.09(s,1H),3.45–3.37(m,2H),3.35(s,3H),1.96–1.93(m,2H),1.85–1.82(m,2H)。
Example 6
Figure PCTCN2021119985-APPB-000083
Step 1: synthesis of compound 006-3:
compound 006-1 (7.13 g,33.93mmol,1 eq) was dissolved in tetrahydrofuran (160 mL), lithium diisopropylamide (2M, 22.06mL,1.3 eq) was added to the reaction mixture after the temperature had dropped to-78℃and the reaction mixture was reacted at-78℃for 1 hour, 006-2 (10 g,37.32mmol,1.1 eq) was added, the reaction mixture was reacted at-78℃for another 1 hour, and then the temperature was slowly raised to 25℃with stirring. After the reaction was completed, it was quenched with 25mL of saturated ammonium chloride solution, extracted three times with ethyl acetate (150 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was isolated by column chromatography (petroleum ether: ethyl acetate=0-10%) to give compound 006-3.MS (ESI) m/z 296.8[ M+H ]] +
Step 2: synthesis of compound 006-4:
compound 006-3 (12.00 g,30.21mmol,1 eq) was dissolved in a mixed solution of N-N dimethylacetamide (100 mL) and water (10 mL) under nitrogen, and dichlorobis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine was added]Palladium (2.14 g,3.02mmol,2.14mL,0.1 eq) and triethylamine (12.23 g,120.82mmol,16.82mL,4 eq) were reacted by pumping nitrogen three times and heating to 130℃for 5 hours. The reaction solution was cooled to room temperature, 150mL of water was added, extraction was performed with ethyl acetate (200 mL. Times.3), the organic phases were combined, concentrated under reduced pressure to a concentrated solution volume of about 150mL, washed with water 4 times, washed with saturated brine 2 times, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. Separating crude product by column chromatography (30% -35% ethyl acetate in petroleum ether) to obtain compound 006-4, MS (ESI) m/z 263.9[ M+H ] ] +
Step 3: synthesis of compound 006-5:
compound 006-4 (8.83 g,27.65mmol,1 eq) was dissolved in tetraethyltitanate (85 ml) and 004-5A (10.05 g,82.94mmol,3 eq) was added and the system was purged with nitrogen three times before warming to 130℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, the reaction mixture was added to ice water, stirred for 40 minutes, the supernatant was added to a separating funnel, extraction was performed by adding ethyl acetate (200 mL. Times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is separated by column chromatography (25% -35% ethyl acetate in petroleum ether) to obtain compound 006-5.MS (ESI) m/z 322.9[ M ] t Bu] +
Step 4: synthesis of compound 006-6:
compound 006-5 (7.34 g,17.37mmol,1 eq) was dissolved in tetrahydrofuran (70 ml), cooled to 0deg.C, sodium borohydride (1.31 g,34.74mmol,2 eq) was added with stirring and the reaction was gradually brought back to 25deg.C for 16 hours. 150mL of water was added to the reaction mixture to quench the unreacted NaBH 4 Ethyl acetate extraction (200 ml×3), and the combined organic phases were dried over anhydrous sodium sulfate and the crude product concentrated under reduced pressure. The crude product was purified by SFC to afford compound 006-6.MS (ESI) m/z 325.0[ M ] t Bu] +
Step 5: synthesis of compound 006-7:
compound 006-6 (1.10 g,2.59mmol,1 eq) was dissolved in dichloromethane (10 mL) and trifluoroacetic acid (3.84 g,33.68mmol,2.49mL,13 eq) was added and the mixture reacted at 25℃for 50min. After the completion of the reaction, part of trifluoroacetic acid was concentrated under reduced pressure, 30mL of water was added to the concentrated solution, 3g of potassium carbonate was added to remove excess trifluoroacetic acid, ethyl acetate was used for extraction (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give compound 006-7.MS (ESI) m/z 325.1[ M+H ]] +
Step 6: synthesis of compound 006-8:
compounds 006-7 (380 mg,1.16mmol,1 eq) and 002-5 (40)5mg,1.16mmol,1 eq) in N, N-dimethylacetamide (10 mL) and water (10 mL) potassium carbonate (995 mg,5.8mmol,5 eq) was added and reacted at 100℃for 48 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, water (50 mL) was added, the mixture was extracted with ethyl acetate (50 mL. Times.3), and the organic phase was combined and washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was isolated by column chromatography (dichloromethane: methanol=0-10%) to give compound 006-8.MS (ESI) m/z 637.2[ M+H ]] +
Step 7: synthesis of compound 006:
Compound 006-8 (308 mg, 483. Mu. Mol,1 eq) was dissolved in methanol (10 mL) and hydrochloric acid in methanol (4M, 2.5mL,58.5 eq) was added. The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain 006 crude product, and the crude product is sent to chiral supercritical fluid chromatography (chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm) and mobile phase: [0.1% ammonia water-methanol)]The method comprises the steps of carrying out a first treatment on the surface of the 40-40% of methanol. Compound 006A (5.115 min) and compound 006B (6.812 min) were isolated. Compound 006A: MS (ESI) m/z 533.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD) delta ppm 7.94 (d, j=7.78 hz, 1H), 7.65 (s, 1H), 7.37 (t, j=7.91 hz, 1H), 7.24 (dd, j=7.91, 5.14hz, 1H), 7.19-7.10 (m, 1H), 7.00-6.93 (m, 2H), 4.39-4.25 (m, 2H), 3.98 (s, 1H), 3.35 (s, 3H), 3.30-3.10 (m, 3H), 2.79 (d, j=15.31 hz, 1H), 1.90-1.70 (m, 2H), 1.64-1.59 (m, 1H), 1.45-1.35 (m, 1H). Compound 006B: MS (ESI) m/z 533.1[ M+H ]] + 。δppm 7.94(d,J=7.78Hz,1H),7.65(s,1H),7.37(t,J=7.91Hz,1H),7.24(dd,J=7.91,5.14Hz,1H),7.19–7.10(m,1H),7.00–6.93(m,2H),4.39–4.25(m,2H),3.98(s,1H),3.35(s,3H),3.30-3.10(m,3H),2.79(d,J=15.31Hz,1H),1.90–1.70(m,2H),1.64–1.59(m,1H),1.45–1.35(m,1H)。
Example 7
Figure PCTCN2021119985-APPB-000084
Figure PCTCN2021119985-APPB-000085
Step 1: synthesis of Compound 007-2:
compound 007-1 (500 mg,1.23mmol,1 eq) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (3.0 mL) was added. The reaction mixture is reacted for 2 hours at 25 ℃, then potassium carbonate is added to neutralize the reaction system to be neutral, and the solvent is dried by spinning to obtain the compound 007-2.MS (ESI) m/z 306.1[ M+H ]] +
Step 2: synthesis of Compound 007-3:
Compound 007-2 (365 mg,1.19mmol,1 eq) and compound 002-5 (420 mg,1.20mmol,1 eq) were dissolved in N, N-dimethylacetamide (10 mL) and water (10 mL) and reacted at 100℃for 48 hours after the addition of potassium carbonate (832 mg,5.8mmol,5 eq). After completion of the reaction, the mixture was cooled to room temperature, water (10 mL) was added, the mixture was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was isolated by column chromatography (dichloromethane: methanol=0-10%) to give compound 007-3.MS (ESI) m/z 621.1[ M+H ]] +
Step 3: synthesis of Compound 007:
compound 007-3 (238 mg, 391. Mu. Mol,1 eq) was dissolved in methanol (10 mL) and a solution of hydrochloric acid in methanol (4M, 2.5mL,58.5 eq) was added. The reaction mixture is directly spin-dried to obtain crude product after reacting for 1 hour at 25 ℃, and the crude product is sent to supercritical fluid chromatography (chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm) and mobile phase: [0.1% ammonia-ethanol ]]The method comprises the steps of carrying out a first treatment on the surface of the 48% -78% of ethanol and 10min. Compound 007A (6.802 min) and compound 007B (7.384 min) were isolated. Compound 007A: MS (ESI) m/z 516.1[ M+H ]] + . Compound 007A: MS (ESI) m/z 516.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm8.35(d,J=5.14Hz,1H)7.95(dd,J=7.91Hz,1.38,1H),7.84(d,J=7.53Hz,2H),7.63(s,1H),7.37(t,J=7.91Hz,1H),7.28(dd,J=7.40,5.14Hz,1H),7.00(dd,J=7.91,1.38Hz,1H),4.37–4.26(m,2H),4.04(s,1H),3.35(s,3H),3.20–3.30(m,3H),2.97–2.82(m,1H),1.95–1.75(m,2H),1.70–1.60(m,1H),1.45–1.40(m,1H)。Compound 007B: MS (ESI) m/z 516.1[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm8.35(d,J=5.14Hz, 1H),7.95(dd,J=7.91Hz,1.38,1H),7.84(d,J=7.53Hz,2H),7.63(s,1H),7.37(t,J=7.91Hz,1H),7.28(dd,J=7.40,5.14Hz,1H),7.00(dd,J=7.91,1.38Hz,1H),4.37–4.26(m,2H),4.04(s,1H),3.35(s,3H),3.20–3.30(m,3H),2.97–2.82(m,1H),1.95–1.75(m,2H),1.70–1.60(m,1H),1.45–1.40(m,1H)。
Example 8
Figure PCTCN2021119985-APPB-000086
Step 1: synthesis of Compound 008-2:
Compound 006-1 (19 g,70.92mmol,1.1 eq) was dissolved in tetrahydrofuran (160 mL), lithium diisopropylamide (2M, 38.6mL,1.2 eq) was added to the reaction mixture after the temperature had dropped to-78℃and the reaction mixture was reacted at-78℃for 1 hour, 008-1 (13.6 g,64.50mmol,1.0 eq) was added, the reaction mixture was reacted at-78℃for another 1 hour, and then the temperature was slowly raised to 25℃with stirring. After the reaction was completed, it was quenched with 250mL of saturated ammonium chloride solution, extracted three times with ethyl acetate (150 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was separated by column chromatography (petroleum ether: ethyl acetate=0-10%) to give compound 008-2.MS (ESI) m/z 297.9[ M ] t Bu] +
Step 2: synthesis of Compound 008-3:
compound 008-2 (12.00 g,30.21mmol,1 eq) was dissolved in a mixed solution of N-N dimethylacetamide (100 mL) and water (10 mL) under the protection of nitrogen, and dichlorobis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine was added]Palladium (2.14 g,3.02mmol,2.14mL,0.1 eq) and triethylamine (12.23 g,120.82mmol,16.82mL,4 eq) were reacted by pumping nitrogen three times and heating to 130℃for 5 hours. The reaction mixture was cooled to room temperature, 150mL of water was added, extraction was performed with ethyl acetate (200 mL. Times.3), the organic phases were combined, concentrated under reduced pressure to a concentrated solution volume of about 150mL, washed with saturated brine (200 mL. Times.6), and dried over anhydrous sodium sulfate Drying and concentrating under reduced pressure to obtain a crude product. Separating the crude product by column chromatography (30-35% ethyl acetate in petroleum ether) to obtain 008-3, MS (ESI) m/z 264.1[ M ] t Bu] +
Step 3: synthesis of Compound 008-4:
008-3 (18 g,56.36mmol,1 eq) was dissolved in tetraethyl titanate (180 mL), 004-5A (20.49 g,169.08mmol,3 eq) was added, nitrogen was purged three times, and then the temperature was raised to 130℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, the reaction mixture was added to ice water, stirred for 40 minutes, the supernatant was added to a separating funnel, extraction was performed by adding ethyl acetate (200 mL. Times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is separated by column chromatography (25% -35% ethyl acetate in petroleum ether) to obtain compound 008-4.MS (ESI) m/z 323.2[ M-Boc ]] +
Step 4: synthesis of Compound 008-5:
008-4 (18 g,42.60mmol,1 eq) was dissolved in tetrahydrofuran (70 mL), cooled to 0℃and sodium borohydride (3.22 g,85.20mmol,2 eq) was added with stirring and the reaction was gradually brought back to 25℃for 16 hours. 150mL of water was added to the reaction mixture to quench the unreacted NaBH 4 Ethyl acetate extraction (200 ml×3), and the combined organic phases were dried over anhydrous sodium sulfate and the crude product concentrated under reduced pressure. The crude product was purified by SFC (column DAICEL CHIRALPAK AD (250 mm. Times.50 mm,10 μm); mobile phase: [0.1% ammonia-ethanol) ]The method comprises the steps of carrying out a first treatment on the surface of the 25% -25% of ethanol. The compound 008-5 was isolated. MS (ESI) m/z 325.2[ M-Boc ]] +
Step 5: synthesis of Compound 008-6:
008-5 (800 mg,1.88mmol,1 eq) was dissolved in methylene chloride (10 mL), trifluoroacetic acid (3.84 g,33.68mmol,2.49mL,13 eq) was added, and the mixture was reacted at 25℃for 50 minutes. After the completion of the reaction, a part of trifluoroacetic acid was concentrated under reduced pressure, 30mL of water was added to the concentrated solution, 3g of potassium carbonate was added to remove excess trifluoroacetic acid, ethyl acetate was used for extraction (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered and the solvent was removed by rotary evaporation to give compound 008-6.MS (ESI) m/z:325.2[M+H] +
Step 6: synthesis of Compound 008-7:
compound 008-6 (570 mg,1.76mmol,1 eq) and compound 002-5 (767 mg,2.20mmol,1.2 eq) were dissolved in N, N-dimethylacetamide (10 mL) and water (10 mL), and after addition of potassium carbonate (607 mg,4.39mmol,2.5 eq) the mixture was reacted at 100℃for 48 hours. After completion of the reaction, the mixture was cooled to room temperature, water (50 mL) was added, the mixture was extracted with ethyl acetate (50 mL. Times.3), and the organic phase was combined and washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give Compound 008-7.MS (ESI) m/z 637.2[ M+H ]] +
Step 7: synthesis of compound 008:
Compound 008-7 (1.04 g,1.63mmol,1 eq) was dissolved in methanol (10 mL) and a solution of hydrochloric acid in methanol (4M, 2.5mL,58.5 eq) was added. The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain 008 crude product, and the crude product is sent to supercritical fluid chromatography (chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm) and mobile phase: [0.1% ammonia-ethanol ]]The method comprises the steps of carrying out a first treatment on the surface of the 45% -45% of ethanol. Compound 008A (2.921 min) and compound 008B (3.872 min) were isolated. Compound 008A: MS (ESI) m/z 533.1[ M+H ]]+。 1 H NMR(400MHz,CD 3 OD) delta ppm 7.94 (dd, j=7.91, 1.38hz, 1H), 7.63 (s, 1H), 7.38 (t, j=7.91 hz, 1H), 7.30-7.20 (m, 2H), 7.00-6.95 (m, 2H), 4.35-4.28 (m, 2H), 4.03 (s, 1H), 3.36 (s, 3H), 3.32-3.25 (m, 2H), 3.21 (d, j=16.06 hz, 1H), 2.85 (d, j=16.06 hz, 1H), 1.92-1.77 (m, 2H), 1.65-1.60 (m, 1H), 1.50-1.43 (m, 1H). Compound 008B: MS (ESI) m/z 533.1[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δppm 7.84(dd,J=7.91,1.38Hz,1H),7.63(s,1H),7.42(t,J=7.91Hz,1H),7.30–7.20(m,2H),6.99(t,J=8.53Hz,1H),6.86(dd,J=8.53,1.51Hz,1H),4.25–4.18(m,2H),3.89(s,1H),3.36(s,3H),3.32–3.15(m,2H),3.10(d,J=16.06Hz,1H),2.65(d,J=16.06Hz,1H),1.80–1.65(m,2H),1.60–1.50(m,1H),1.20–1.05(m,1H)。
Example 9
Figure PCTCN2021119985-APPB-000087
Figure PCTCN2021119985-APPB-000088
Step 1: synthesis of Compound 009-3:
potassium tert-butoxide (17.43 g,155.36mmol,3.5 eq) was dissolved in ethylene glycol dimethyl ether (500 mL) at 0deg.C, and after three nitrogen substitutions, a solution of 009-2 (13.00 g,66.58mmol,1.5 eq) in ethylene glycol dimethyl ether (500 mL) was slowly added dropwise at 0deg.C. The temperature is controlled not to be higher than 5 ℃. After the mixture was stirred at 0℃for 1 hour, isopropyl alcohol (3.73 g,62.14mmol,4.76mL,1.4 eq) was added at 0℃and the mixture was stirred for 30 minutes. A solution of 009-1 (10 g,44.39mmol,1 eq) in DME (500 mL) was added. The mixture was stirred at 0℃for 1 hour and then returned to 25℃for 12 hours. After the reaction was completed, it was quenched with 200mL of saturated ammonium chloride solution, extracted with ethyl acetate (500 mL x 3), and the organic phases were combined, dried over anhydrous sodium sulfate and spun dry. The crude product was purified by flash column chromatography (petroleum ether: ethyl acetate=20:1-10:1) to give compound 009-3.
Step 2: synthesis of Compound 009-5:
compound 009-3 (6.3 g,26.66mmol,1 eq) was dissolved in tetrahydrofuran (250 mL), lithium diisopropylamide (2M, 22.06mL,1.3 eq) was added to the reaction system after the temperature had dropped to-78℃and reacted at-78℃for 1 hour, a solution of 009-4 (7.50 g,27.99mmol,1.05 eq) in tetrahydrofuran was added to the reaction system for 50mL, the system was reacted at-78℃for another 1 hour, and then the temperature was slowly raised to 25℃with stirring. After the reaction was completed, it was quenched with 250mL of saturated ammonium chloride solution, extracted three times with ethyl acetate (450 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. The crude product was separated by column chromatography (petroleum ether: ethyl acetate=10-20%) to give compound 009-5.MS (ESI) m/z 369.0[ M ] t Bu] +
Step 3: synthesis of Compound 009-6:
compound 009-5 (5)1g,12.05mmol,1 eq) in a mixed solution of N-N dimethylacetamide (200 mL) and water (20 mL) was added dichlorobis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine]Palladium (0.85 g,1.2mmol,0.1 eq) and triethylamine (4.88 g,48.19mmol,6.71mL,4 eq) were reacted by pumping nitrogen three times and raising the temperature to 130℃for 5 hours. The reaction solution was cooled to room temperature, 150mL of water was added, extraction was performed with ethyl acetate (200 mL. Times.3), the organic phases were combined, concentrated under reduced pressure to a concentrated solution volume of about 150mL, washed with saturated brine (200 mL. Times.6), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. Separating the crude product by column chromatography (30-35% ethyl acetate in petroleum ether) to obtain 009-6, MS (ESI) m/z 290.1[ M ] t Bu] +
Step 4: synthesis of Compound 009-7:
009-6 (4.2 g,12.16mmol,1 eq) was dissolved in tetraethyltitanate (100 mL), 004-5A (4.42 g,36.48mmol,3 eq) was added, the system was purged with nitrogen three times, and then warmed to 130℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, the reaction mixture was added to ice water, stirred for 40 minutes, the supernatant was added to a separating funnel, extraction was performed by adding ethyl acetate (200 mL. Times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is separated by column chromatography (10 to 20 percent of ethyl acetate in petroleum ether) to obtain the compound 009-7.MS (ESI) m/z 449.1[ M+H ]] +
Step 5: synthesis of Compound 009-8:
009-7 (4.5 g,10.03mmol,1 eq) was dissolved in tetrahydrofuran (70 mL), cooled to 0℃and sodium borohydride (379.51 mg,10.03mmol,1 eq) was added with stirring and the reaction was gradually brought back to 25℃for 4 hours. 150mL of water was added to the reaction mixture to quench the unreacted NaBH 4 Ethyl acetate extraction (200 ml×3), and the combined organic phases were dried over anhydrous sodium sulfate and the crude product concentrated under reduced pressure. The crude product was purified by high performance liquid chromatography (column Phenomenex Genimi NX C (150 mm. Times.40 mm,5 μm); mobile phase [ water (0.225% formic acid) -acetonitrile) ]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 42-72 percent) and separating to obtain the compound 009-8A and the compound 009-8B. MS (ESI) m/z 451.1[ M+H ]] + Step 6: compounds of formula (I)Synthesis of 009-9A:
009-8A (0.4 g,0.89mmol,1 eq) was dissolved in dichloromethane (10 mL), trifluoroacetic acid (3.84 g,33.68mmol,2.49mL,13 eq) was added and the mixture was reacted at 25℃for 50 min. After the reaction, part of trifluoroacetic acid was concentrated under reduced pressure, 30mL of water was added to the concentrated solution, 3g of potassium carbonate was added to remove excess trifluoroacetic acid, ethyl acetate was used for extraction (50 mL. Times.3), the organic phases were combined and dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to obtain Compound 009-9A. MS (ESI) m/z 351.1[ M+H ]] +
Step 7: synthesis of Compound 009-10A:
compound 009-9A (300 mg,0.860mmol,1 eq) and compound 002-5 (300 mg,0.860mmol,1 eq) were dissolved in N, N-dimethylacetamide (10 mL) and water (10 mL), and after the addition of potassium carbonate (0.591 mg,4.3mmol,5 eq) the mixture was reacted at 100℃for 48 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, water (50 mL) was added, the mixture was extracted with ethyl acetate (50 mL. Times.3), and the organic phase was combined and washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was separated by column chromatography (dichloromethane: methanol=0-10%) to give compound 009-10A. MS (ESI) m/z 351.1[ M+H ] ] +
Step 8: synthesis of Compound 009A:
compound 009-10A (40 mg, 60.3. Mu. Mol,1 eq) was dissolved in methanol (5 mL) and a solution of hydrochloric acid in methanol (4M, 2.5mL,58.5 eq) was added. The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain 009A crude product, and the crude product is subjected to high performance liquid chromatography (chromatographic column: phenomenex Gemini-NX C18 (75 mm. Times.30 mm,3 μm) and mobile phase: [ water (0.225% formic acid) -acetonitrile ]]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 5 to 35 percent) and 7 minutes. The formate of compound 009A was isolated: MS (ESI) m/z 560.0[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 7.98–7.86(m,1H),7.51(s,1H),7.48–7.34(m,2H),7.00–6.97(m,1H),6.91–6.55(m,2H),4.71–4.65(m,1H),4.61(s,2H),4.35(s,1H),3.37(s,3H),3.10–3.02(m,1H),2.44–2.34(m,1H),2.27–2.15(m,2H),2.12–2.01(m,3H),1.91–1.83(m,1H),1.64–1.57(m,1H)。
Step 9: synthesis of Compound 009-9B:
009-8B (0.4 g,0.89mmol,1 eq) was dissolved in dichloromethane (10 mL) and trifluoroacetic acid (3.84 g,33.68mmol,2.49mL,13 eq) was added and the mixture reacted at 25℃for 50 min. After the completion of the reaction, a part of trifluoroacetic acid was concentrated under reduced pressure, 30mL of water was added to the concentrated solution, 3g of potassium carbonate was added to remove excess trifluoroacetic acid, ethyl acetate was used for extraction (50 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered and the solvent was removed by rotary evaporation to give Compound 009-9B. MS (ESI) m/z 351.1[ M+H ]] +
Step 10: synthesis of Compound 009-10B:
compound 009-9B (300 mg,0.860mmol,1 eq) and compound 002-5 (300 mg,0.860mmol,1 eq) were dissolved in N, N-dimethylacetamide (10 mL) and water (10 mL), and after the addition of potassium carbonate (0.591 mg,4.3mmol,5 eq) the mixture was reacted at 100℃for 48 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, water (50 mL) was added, the mixture was extracted with ethyl acetate (50 mL. Times.3), and the organic phase was combined and washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product was separated by column chromatography (dichloromethane: methanol=0-10%) to give compound 009-10B. MS (ESI) m/z 351.1[ M+H ] ] +
Step 11: synthesis of Compound 009B:
compound 009-10B (20 mg, 60.3. Mu. Mol,1 eq) was dissolved in methanol (5 mL) and a solution of hydrochloric acid in methanol (4M, 2.5mL,58.5 eq) was added. The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain 009B crude product, the crude product is prepared into high performance liquid chromatography (chromatographic column: phenomenex Gemini-NX C18 (75 mm. Times.30 mm,3 μm), mobile phase: [ water (0.225% formic acid) -acetonitrile]The method comprises the steps of carrying out a first treatment on the surface of the Acetonitrile percent of 5 to 35 percent) and 7 minutes. Separating to obtain formate of compound 009B, MS (ESI) m/z:5,60.0[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 7.98–7.86(m,1H),7.51(s,1H),7.48–7.34(m,2H),7.00–6.97(m,1H),6.91–6.55(m,2H),4.71–4.65(m,1H),4.61(s,2H),4.35(s,1H),3.37(s,3H),3.10–3.02(m,1H),2.44–2.34(m,1H),2.27–2.15(m,2H),2.12–2.01(m,3H),1.91–1.83(m,1H),1.64–1.57(m,1H)。
Example 10
Figure PCTCN2021119985-APPB-000089
Figure PCTCN2021119985-APPB-000090
Step 1: synthesis of Compound 010-2:
compound 010-1 (8.5 g,41.06mmol,1 eq) was dissolved in chloroform (100 mL), N-bromosuccinimide (7.31 g,41.06mmol,1 eq) was added, benzoyl peroxide (596.75 mg,2.46mmol,0.06 eq) was purged three times with nitrogen, and then the temperature was slowly raised to 80℃for reaction for 5hr. After the completion of the reaction, the reaction mixture was cooled to room temperature, filtered, 200mL of water was added to the filtrate, the mixture was extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (200 mL. Times.1), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. The crude product is separated by flash column chromatography (3 to 5 percent of ethyl acetate in petroleum ether) to obtain the compound 010-2.
Step 2: synthesis of Compound 010-3:
compound 006-1 (4.35 g,20.67mmol,1 eq) was dissolved in tetrahydrofuran (120 mL), the system was purged with nitrogen three times, lithium diisopropylamide (2M, 12.40mL,1.2 eq) was added to the reaction system at-78℃for 1 hour, and 010-2 (6.5 g,22.73mmol,1.1 eq) in tetrahydrofuran (30 mL) was added. The system was reacted at-78℃for a further 1 hour, then slowly warmed to 25℃with stirring for a further 10 hours. After the reaction was completed, it was quenched with 100mL of saturated ammonium chloride solution, extracted three times with ethyl acetate (100 mL. Times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation. Column chromatography: the crude product is separated by column chromatography (10 to 15 percent of ethyl acetate in petroleum ether) to obtain the compound 010-3.MS (ESI) m/z=358.9 [ m ] t Bu] +
Step 3: synthesis of Compound 010-4:
compound 010-3 (4.41 g,10.62mmol,1 eq) was dissolved in a mixed solution of N-N dimethylacetamide (40 mL) and water (4 mL) and dichlorobis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine was added]Palladium (751.94 mg,1.06mmol, 751.94. Mu.L, 0.1 eq) and triethylamine (4.30 g,42.48mmol,5.91mL,4 eq) were reacted by pumping nitrogen three times and heating to 130℃for 5 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, 150mL of water was added, extraction was performed with ethyl acetate (200 mL. Times.3), the organic phases were combined, concentrated under reduced pressure to a concentrated solution volume of about 150mL, washed with saturated brine (200 mL. Times.6), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is separated by column chromatography (15 to 25 percent of ethyl acetate in petroleum ether) to obtain the compound 010-4, MS (ESI) m/z: =282.1 [ M ] t Bu] +
Step 4: synthesis of Compound 010-5:
compound 010-4 (2.35 g,6.97mmol,1 eq) was dissolved in tetraethyltitanate (30 mL), 004-5A (2.53 g,20.90mmol,3 eq) was added, the system was purged with nitrogen three times, and then warmed to 130℃for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, the reaction mixture was added to ice water, stirred for 40 minutes, the supernatant was added to a separating funnel, extraction was performed by adding ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product. The crude product is separated by column chromatography (25% -35% ethyl acetate in petroleum ether) to obtain compound 010-5.MS (ESI) m/z 341.1[ M-Boc ]] +
Step 5: synthesis of Compound 010-6:
compound 010-5 (2.68 g,6.08mmol,1 eq) was dissolved in tetrahydrofuran (30 mL), cooled to 0deg.C, sodium borohydride (460.30 mg,12.17mmol,2 eq) was added with stirring and the reaction was gradually brought back to 25deg.C for 16 hours. 100mL of water was added to the reaction mixture to quench the unreacted NaBH 4 Ethyl acetate extraction (100 ml×3), combining the organic phases and drying over anhydrous sodium sulfate, concentrating under reduced pressure to give the crude product. The crude product is subjected to column chromatography (45 to 55 percent of ethyl acetate in petroleum ether) and separated to obtain a compound 010-6.MS (ESI) m/z 343.1[ M-Boc ] +
Step 6: synthesis of Compound 010-7:
compound 010-6 (435.50 mg, 984.04. Mu. Mol,1 eq) was dissolved in methylene chloride (5 mL), trifluoroacetic acid (1.57 g,13.78mmol,1.02mL,14 eq) was added, and the mixture was reacted at 25℃for 50 minutes. After the reaction, part of trifluoroacetic acid was concentrated under reduced pressure, 30mL of water was added to the concentrated solution, 3g of potassium carbonate was added to remove excess trifluoroacetic acid, ethyl acetate was used for extraction (50 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give compound 010-7.MS (ESI) m/z 343.1[ M+H ]] +
Step 7: synthesis of Compound 010-8:
compound 001-7 (330.00 mg, 963.66. Mu. Mol,1 eq) and compound 002-5 (403.88 mg,1.16mmol,1.2 eq) were dissolved in N, N-dimethylacetamide (7 mL) and water (7 mL), and then reacted at 80℃for 16 hours after adding potassium carbonate (399.55 mg,2.89mmol,3 eq). After completion of the reaction, the reaction mixture was cooled to room temperature, water (50 mL) was added, the mixture was extracted with ethyl acetate (50 mL. Times.3), and the organic phase was combined and washed with saturated brine (50 mL. Times.6), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to give a crude product. The crude product is subjected to column chromatography (4 to 8 percent of methylene dichloride in methanol) and separated to obtain the compound 010-8.MS (ESI) m/z 655.2[ M+H ] ] +
Step 8: synthesis of Compound 010:
compound 010-8 (160 mg, 244.18. Mu. Mol,1 eq) was dissolved in methanol (2 mL) and hydrochloric acid in methanol (4M, 1.86mL,30.43 eq) was added. The reaction mixture is directly spin-dried after reacting for 1 hour at 25 ℃ to obtain 010 crude product, and the crude product is sent to supercritical fluid chromatography (chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm) and mobile phase: [0.1% ammonia-ethanol ]]The method comprises the steps of carrying out a first treatment on the surface of the Ethanol accounts for 40-40%. Compound 010A (retention time: 2.561 min) and compound 010B (retention time: 3.389 min) were isolated. Compound 010A: MS (ESI) m/z 551.0[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 7.94(dd,J=7.91,1.38Hz,1H),7.61(s,1H),7.35(t,J=7.91Hz,1H),7.30–7.23(m,1H),7.18–7.10(m,1H),7.00–6.95(m,1H),4.35–4.28(m,2H),3.94(s,1H),3.36(s,3H),3.30–3.11(m,3H),2.82–2.75(m,1H),1.90–1.71(m, 2H), 1.64-1.55 (m, 1H), 1.45-1.37 (m, 1H). Compound 010B: MS (ESI) m/z 551.0[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δppm 7.94(dd,J=7.91,1.38Hz,1H),7.61(s,1H),7.35(t,J=7.91Hz,1H),7.30–7.23(m,1H),7.18–7.10(m,1H),7.00–6.95(m,1H),4.35–4.28(m,2H),3.94(s,1H),3.36(s,3H),3.30–3.11(m,3H),2.82–2.75(m,1H),1.9–1.71(m,2H),1.64–1.55(m,1H),1.45–1.37(m,1H)。
Biological testing
Experimental example 1: in vitro evaluation
Reaction buffer:
60mM hydroxyethylpiperazine ethylene sulfate (HEPES) (pH 7.4), 1mM ethylenediamine tetraacetic acid (EDTA), 75mM KCl,75mM NaCl,0.01%Brij-35,5mM Dithiothreitol (DTT) and 10% DMSO (final).
Enzyme: PTPN11/SHP2-FL (RBC production, no CAS number)
Recombinant human PTPN11 full length (Genbank accession # NM-002834; aa 2-597,
isoform 1 (specification)) is expressed in e.coli, with a tag of N-terminal strep ii-TEV, C-terminal histidine. Mw= 71.93kDa.
Activation peptide: H2N-LN (pY) IDLDLV (dPEG 8) LST (pY) ASINFQK-amide (based on publications)
A substrate: diFMUP [6, 8-difluoro-7-hydroxy-4-methylcoumarin ]
Final concentration in assay:
0.35 mu M activating peptide
100. Mu.M DiFMUP (6, 8-difluoro-4-methylumbelliferone phosphate)
The steps are as follows:
1. preparing the specified enzyme/peptide and substrate in a freshly prepared reaction buffer;
2. adding an enzyme/peptide solution to the reaction well;
3. compounds were provided to enzyme solutions (Echo 550; nanoliter range) in 100% dmso by acoustic technique, incubated for 30 minutes at room temperature;
4. adding a substrate solution to the reaction well to initiate a reaction;
5. enzyme activity (Ex/Em 355/460) was monitored as a time course measurement of 60 minutes increase in fluorescent signal of fluorogenic substrate at room temperature;
6. data analysis: taking the slope of the linear portion measured over time x (signal/min) and calculating% enzyme activity relative to DMSO control; the background slope of the basal enzyme activity (no peptide) was subtracted.
The results of the in vitro screening test of the compounds of the present invention are shown in the reference numeral 1.
TABLE 1 in vitro screening test results for the compounds of the invention
Numbering of compounds PTPN11/SHP2-FL(IC 50 nM)
Formate of compound 001 21.8
Formate of Compound 002 12.6
Formate of Compound 003 72.4
Formate of compound 004 2.84
Formate of compound 004A 3.21
Formate of compound 004B 2.61
Compound 005A 5.35
Compound 005B 3.66
Compound 006A 2.95
Compound 006B 2.22
Compound 008A 2.78
Compound 008B 2.43
Compound 010A 6.01
Compound 010B 5.81
Conclusion: the compound has certain inhibition activity on PTPN11/SHP 2-FL.
Experimental example 2: evaluation of Compound H358 cell Activity
The purpose of the experiment is as follows:
this experiment was conducted to verify the inhibitory effect of the compounds of the present invention on proliferation of KRAS G12C mutated NCI-H358 human non-small cell lung cancer cells.
Experimental materials:
cell line NCI-H358 (from Punoxel), RPMI1640 medium, penicillin/streptomycin antibiotics from Vison's, and fetal bovine serum from Biosera. CellTiter-Glo (cell viability chemiluminescent detection reagent) reagent was purchased from Promega.
The experimental method comprises the following steps:
NCI-H358 cells were seeded in white 96-well plates, 80. Mu.L of cell suspension per well, containing 4000 NCI-H358 cells. Cell plates were placed in a carbon dioxide incubator overnight for culture. The test compounds were diluted 5-fold to the 9 th concentration, i.e. from 2000 μm to 5.12nM, using a row gun and a double multiplex assay was set up. 78. Mu.L of medium was added to the intermediate plate, and 2. Mu.L of the gradient diluted compound per well was transferred to the intermediate plate at the corresponding position, and 20. Mu.L of the gradient diluted compound per well was transferred to the cell plate after mixing. The concentration of compound transferred into the cell plate ranged from 10. Mu.M to 0.026nM. The cell plates were placed in a carbon dioxide incubator for 5 days. A cell plate was also prepared and the signal value read on the day of dosing as the maximum value (Max value in the following equation) was used in the data analysis. To this plate, 25. Mu.L of cell viability chemiluminescent detection reagent was added per well and incubated at room temperature for 10 minutes to stabilize the luminescent signal. After incubation of cell plates with compounds added, a multi-label analyzer reading was used.
Data analysis:
the raw data was converted to inhibition rate using the equation (Sample-Min)/(Max-Min) ×100%, and the IC50 values were obtained by curve fitting four parameters (obtained in the "log (inhibitor) vs. response-Variable slope" mode in GraphPad Prism).
The results of the cell activity screening assay for compound H358 of the present invention are shown in Table 2.
TABLE 2 in vitro screening test results for the compounds of the invention
Numbering of compounds H358(IC 50 nM)
Formate of compound 004A 56.5
Formate of compound 004B 19.2
Compound 006A 30
Compound 006B 18
Compound 010A 11.9
Compound 010B 7.9
Conclusion: the compounds of the present invention have good inhibitory activity on H358 cells.
Experimental example 3: pharmacokinetic evaluation of Compounds
The purpose of the experiment is as follows: test compounds pharmacokinetic in CD-1 mice
Experimental materials: CD-1 mouse (Male, 32-33 g)
Experimental operation:
the rodent drug substitution profile of the compounds after intravenous and oral administration was tested in standard protocols, and the candidate compounds in the experiments were formulated as clear solutions for single intravenous and oral administration to mice. The intravenous injection and oral solvent is hydroxypropyl beta cyclodextrin water solution or physiological saline solution with a certain proportion. Collecting whole blood sample within 24 hours, centrifuging at 3000g for 15 minutes, separating supernatant to obtain plasma sample, adding 4 times volume of acetonitrile solution containing internal standard to precipitate protein, centrifuging to obtain supernatant, adding equal volume of water, centrifuging to obtain supernatant, sampling, quantitatively analyzing blood concentration by LC-MS/MS analysis method, and calculating drug substitution parameters such as peak concentration, peak time, clearance, half life, area under curve of drug time, bioavailability, etc.
The pharmacokinetic test results of the compounds of the present invention are shown in table 3.
TABLE 3 pharmacokinetic test results
Figure PCTCN2021119985-APPB-000091
Conclusion: the compound of the invention can obviously improve single or partial indexes of the pharmacokinetics of mice.
Experimental example 4: human liver microsome CYP inhibition assay
The objective of the study was to evaluate the inhibition of human liver microsomal cytochrome P450 isozymes (CYP 1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3 A4) by the test sample using a 5 in 1 probe substrate for the CYP isozymes.
Mixed Human Liver Microsomes (HLM) were purchased from Corning Inc. (Steuben, new York, USA) or XenoTech, LLC (Lenexa, KS, USA) or other suppliers, and were stored at less than-70℃prior to use.
The diluted serial concentration working solution of the test sample is added into an incubation system containing human liver microsomes, a probe substrate and auxiliary factors of a circulating system, and a control containing no test sample and solvent is taken as an enzyme activity control (100%). The concentration of the metabolite produced by the probe substrate in the sample is determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Non-linear regression analysis was performed on the mean percent activity versus concentration of the test samples using SigmaPlat (V.11). Calculation of IC by three-parameter or four-parameter sigmoid logarithmic equation 50 Values.
The test results are shown in table 4:
TABLE 4 Table 4
Figure PCTCN2021119985-APPB-000092
Figure PCTCN2021119985-APPB-000093
Conclusion: the compounds of the present invention have a weak inhibition of five CYP isozymes.
Experimental example 5: inhibition assay of hERG Potassium ion channel
1. The purpose of the experiment is as follows:
the effect of test example 1 on hERG potassium ion channel was examined by full-automatic patch clamp.
2. Experimental method
2.1. Cell culture
Cells stably expressing hERG potassium ion channel used in the experiments were derived from CHO-hERE of Aviva Biosciences, cultured in 5% CO 2 Under 37 ℃. CHO hERG medium is shown in table 5.
TABLE 5 CHO hERG culture broth
Reagent Supplier Catalog Number Volume(mL)
F12Hams Invitrogen 31765-092 500
FBS Invitrogen 10099-141 50
G418/Geneticin Invitrogen 10131-027 1
Hygromycin B Invitrogen 10687-010 1
2.2. Prophase preparation of cells
CHO-hERG cells prepared for the experiments were cultured for at least two days and the cell density reached more than 75%. Prior to the start of the experiment, cells were digested with TrypLE and then resuspended with extracellular fluid for cell collection.
2.3. Preparation of intracellular and extracellular fluids
Extracellular fluid needs to be prepared once a month. The intracellular fluid should be frozen at-20deg.C. The intracellular and extracellular fluid compositions are shown in Table 6.
TABLE 6 intracellular and extracellular fluid compositions
Composition of components Extracellular fluid (mM) Intracellular fluid (mM)
NaCl 145 -
KCl 4 120
KOH - 31.25
CaCl 2 2 5.374
MgCl 2 1 1.75
Glucose 10 -
Na 2 ATP - 4
HEPES 10 10
EGTA - 10
pH 7.4with NaOH 7.2with KOH
Osmotic pressure 295mOsm 285mOsm
2.4. Preparation of the Compounds
Dissolving a compound to be tested and a positive control Amitriptyline into stock solution with a certain concentration by using DMSO, then diluting according to different gradients, and finally adding the stock solution into extracellular solution according to a certain proportion to dilute the extracellular solution into the concentration to be tested. Visual inspection was used to see if there was any precipitate before the experiment started. Finally, the concentration of DMSO in the test solution and the positive control Amitriptyline cannot exceed 0.3% at maximum.
2.5. Voltage stimulation scheme
Maintaining the clamp potential at-80 mv, firstly applying voltage stimulus of-50 mv for 80ms to record the cell leakage current value, then depolarizing to +20mv for 4800ms, opening the hERG channel, then repolarizing to-50 mv for 5000ms, leading out the hERG tail current and recording, and finally, recovering the voltage to the clamp potential of-80 mv for 3100ms. The above voltage stimulus is repeated every 15000 ms.
2.6.QPatch HTX Whole cell patch clamp recording
hERG QPatch HTX The experiment was performed at room temperature. Whole cell protocols, voltage stimulation protocols and compound detection protocols were established on the software of QPatch Assay Software 5.2.5 (Sophion Bioscience).
The voltage stimulus was repeated 30 times first, this zone being the baseline area for the subsequent analysis, followed by the addition of 5 μl of extracellular fluid, three times. The concentrations of the individual compounds applied were added sequentially and were still repeated three times at a 5. Mu.L addition volume. Cells were incubated at least no less than 5mins per test concentration. In the whole recording process, each index needs to reach the data analysis receiving standard, if the index does not reach the standard, the cell does not count into the analysis range, the compound is tested again, and the recording process is automatically operated by QPatch analysis software. The concentrations tested were 0.24. Mu.M, 1.20. Mu.M, 6.00. Mu.M, 30.00. Mu.M for each compound, and at least two cells were replicated for each concentration.
2.7. Data analysis
In each complete current record, the percent inhibition of each compound concentration was calculated based on the percentage of peak current in the negative control. And fitting by using a standard Highway equation to obtain a dose-response relation curve, wherein the specific equation is as follows:
I (C) =I b +(I fr -I b )*c n /(IC 50 n +c n )
c is the test concentration of the compound, n is the slope
The curve fitting and the inhibition rate calculation are both completed by analysis of QPatch analysis software, and if the inhibition rate exceeds half inhibition at the lowest concentration or the inhibition rate does not reach half inhibition at the highest concentration, the compound corresponds to the IC 50 Below the minimum concentration or IC 50 The value is greater than the highest concentration.
2.8. Test results
Results for hERG IC50 values for the compounds of the examples are shown in table 7.
TABLE 7 example compound hERG IC 50 Value results
Sample for sample hERG IC 50 (nM)
Formate of compound 004A 8.20
Formate of compound 004B 9.82
Compound 006A 10.3
Compound 006B 14.0
Conclusion: the compounds of the invention are not apparent to hERG inhibition.
Experimental example 6: in vitro microsome stability assay
6.1, experimental materials:
6.1.1 liver microsomes
Human and animal microsomes were purchased from Corning or Xenotech and stored in a-80 ℃ freezer.
6.1.2 reduced Nicotinamide Adenine Dinucleotide Phosphate (NADPH), supplier: chem-impex international, cat: 00616
6.1.3 control compound: testosterone, diclofenac, propafenone
6.2 Experimental procedure
6.2.1 preparation of working fluid
Stock solution: 10mM DMSO solution
Working concentration preparation: 100% acetonitrile to 100. Mu.M (organic phase content: 99% ACN,1% DMSO)
6.2.2 Experimental procedure
2 96 Kong Fuyo plates, designated T60 incubation plate and NCF60 incubation plate, respectively, were prepared.
445. Mu.L of microsomal working solution (liver microsomal protein concentration 0.56 mg/mL) was added to each of the T60 and NCF60 incubation plates, and the plates were then placed in a 37℃water bath for pre-incubation for about 10 minutes.
After the pre-incubation, 5. Mu.L of test or control compound working solution was added to each of the T60 and NCF60 incubation plates, and mixed well. The reaction was initiated by adding 50 μl of potassium phosphate buffer per well on NCF60 incubation plates; 180. Mu.L of stop solution (acetonitrile solution containing 200ng/mL tolbutamide and 200ng/mL labetalol) and 6. Mu.L of NADPH regeneration system working solution were added to the T0 stop plate, and 54. Mu.L of sample was removed from the T60 incubation plate to the T0 stop plate (T0 sample was produced). The reaction was started by adding 44. Mu.L of NADPH regeneration system working solution per well on the T60 incubation plate. Only 54. Mu.L of microsomal working solution, 6. Mu.L of NADPH regenerating system working solution and 180. Mu.L of stop solution were added to the Blank plate. Thus, in the samples of the test or control compounds, the final concentrations of the compounds, testosterone, diclofenac and propafenone were 1. Mu.M, the concentrations of liver microsomes were 0.5mg/mL, and the final concentrations of DMSO and acetonitrile in the reaction system were 0.01% (v/v) and 0.99% (v/v), respectively.
After incubation for a suitable period of time (e.g., 5, 15, 30, 45, and 60 minutes), 180. Mu.L of stop solution (acetonitrile solution containing 200ng/mL tolbutamide and 200ng/mL labetalol) was added to each of the sample wells of each of the stop plates, and then 60. Mu.L of sample was removed from the T60 incubation plate to terminate the reaction.
All sample plates were shaken and centrifuged at 3220 Xg for 20 minutes, and 80. Mu.L of supernatant was then diluted into 240. Mu.L of pure water per well for liquid chromatography tandem mass spectrometry.
The MMS results for the compounds of the invention are shown in Table 8.
TABLE 8 MMS results for the inventive compounds
Sample for sample MMS(mL/min/kg),H,M
Formate of compound 004A 19.7,67.9
Formate of compound 004B 33.0,61.1
Compound 006A 31.2,100.8
Compound 006B 47.9,57.3

Claims (22)

  1. A compound of formula (II) or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2021119985-APPB-100001
    wherein,
    structural unit
    Figure PCTCN2021119985-APPB-100002
    Is that
    Figure PCTCN2021119985-APPB-100003
    E 1 Is O or CH 2
    T 1 Is N or CH;
    R 1 is that
    Figure PCTCN2021119985-APPB-100004
    R 11 、R 13 And R is 14 Each independently is C 1-3 An alkyl group;
    R 12 is H or C 1-3 An alkyl group;
    R 2 is F, cl, br or I;
    R 3 is C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R a Substitution;
    R 4 h, F, cl, br, I or C 1-3 Alkyl, said C 1-3 Alkyl is optionally substituted with 1, 2 or 3R b Substitution;
    R a and R is b F, cl, br, I, OH or NH respectively and independently 2
    n is 0, 1, 2 or 3;
    m is 1, 2 or 3;
    when n is 0, the structural unit
    Figure PCTCN2021119985-APPB-100005
    Is that
    Figure PCTCN2021119985-APPB-100006
    Structural unit
    Figure PCTCN2021119985-APPB-100007
    Is that
    Figure PCTCN2021119985-APPB-100008
    When n is 1, 2 or 3, the structural unit
    Figure PCTCN2021119985-APPB-100009
    Is a structural unit
    Figure PCTCN2021119985-APPB-100010
    Structural unit
    Figure PCTCN2021119985-APPB-100011
    Is that
    Figure PCTCN2021119985-APPB-100012
  2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 11 、R 13 And R is 14 Each independently CH 3
  3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 12 Is H or CH 3
  4. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R 1 Is that
    Figure PCTCN2021119985-APPB-100013
  5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 3 Is CH 3
  6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 4 Is F, cl, br or I.
  7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the structural unit
    Figure PCTCN2021119985-APPB-100014
    Is that
    Figure PCTCN2021119985-APPB-100015
  8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein the structural unit
    Figure PCTCN2021119985-APPB-100016
    Is that
    Figure PCTCN2021119985-APPB-100017
  9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein the structural unit
    Figure PCTCN2021119985-APPB-100018
    Is that
    Figure PCTCN2021119985-APPB-100019
    Figure PCTCN2021119985-APPB-100020
  10. The compound according to claim 1, which has the structure of formula (II-1)
    Figure PCTCN2021119985-APPB-100021
    Wherein,
    R 11 、R 12 、R 2 、n、E 1 and structural unit
    Figure PCTCN2021119985-APPB-100022
    As defined in claim 1.
  11. The compound according to claim 1, which has the structure of formula (II-1A) or (II-1B),
    Figure PCTCN2021119985-APPB-100023
    Wherein n, m, T 1 、R 11 、R 12 、R 2 And R is 4 As defined in claims 1-10.
  12. The compound according to claim 1, which has the structure of formula (I-1A), (I-1B) or (I-2A), or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2021119985-APPB-100024
    Figure PCTCN2021119985-APPB-100025
    wherein n, R 11 、R 12 、R 13 、R 14 、R 2 、R 3 And R is 4 As defined in claims 1-10.
  13. The compound according to claim 11-12, which has the structure of formula (I-1A-1), (I-2A-1), (II-1A-1) or (II-1B-1), or a pharmaceutically acceptable salt thereof
    Figure PCTCN2021119985-APPB-100026
    Wherein m, n, E 1 、T 1 、R 11 、R 12 、R 13 、R 14 、R 2 、R 3 And R is 4 As defined in claims 11-12.
  14. The following compounds or pharmaceutically acceptable salts thereof,
    Figure PCTCN2021119985-APPB-100027
    Figure PCTCN2021119985-APPB-100028
  15. a compound according to claim 14, or a pharmaceutically acceptable salt thereof, which compound is
    Figure PCTCN2021119985-APPB-100029
    Figure PCTCN2021119985-APPB-100030
  16. Stereoisomers of the compounds of the formula, or pharmaceutically acceptable salts of said stereoisomers,
    Figure PCTCN2021119985-APPB-100031
    wherein the stereoisomer has a retention time of 3.1-3.5min, preferably 3.2-3.4min, more preferably 3.3min after chiral supercritical fluid chromatography; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: chiralpak AS-3 (100 mm. Times.4.6 mm,3 μm); mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ]:40% -40%.
  17. Stereoisomers of the compounds of the formula, or pharmaceutically acceptable salts of said stereoisomers,
    Figure PCTCN2021119985-APPB-100032
    Wherein the retention time of the compound after chiral supercritical fluid chromatography is 4.3-4.7min, preferably 4.4-4.6min, more preferably 4.5min; the conditions of the chiral supercritical fluid chromatographic analysis are as follows: chromatographic column: chiralpak AS-3 (100 mm. Times.4.6 mm,3 μm); mobile phase: carbon dioxide; [0.05% triethylamine, ethanol% ]:40% -40%.
  18. Stereoisomers of the compounds of the formula, or pharmaceutically acceptable salts of said isomers,
    Figure PCTCN2021119985-APPB-100033
    wherein the retention time of the compound after chiral supercritical fluid chromatography is 4.9-5.3min, preferably 5.0-5.2min, more preferably 5.1min; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-methanol ]; 40-40% of methanol.
  19. Stereoisomers of the compounds of the formula, or pharmaceutically acceptable salts of said stereoisomers,
    Figure PCTCN2021119985-APPB-100034
    wherein the retention time of the compound after chiral supercritical fluid chromatography is 6.6-7.0min, preferably 6.7-6.9min, more preferably 6.8min; the conditions for chromatographic separation of the chiral supercritical fluid are as follows: chromatographic column: DAICEL CHIRALPAK AS (250 mm. Times.30 mm,10 μm); mobile phase [0.1% ammonia-methanol ]; 40-40% of methanol.
  20. A pharmaceutical composition comprising a compound of any one of claims 1-19, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  21. Use of a compound according to any one of claims 1 to 19, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 20, for the manufacture of a medicament for the treatment of SHP 2-associated diseases.
  22. The use of claim 21, wherein the SHP 2-associated disease is a solid tumor.
CN202180065019.2A 2020-09-23 2021-09-23 Pyrazine sulfur biphenyl compound and application thereof Pending CN116348466A (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
CN2020110083097 2020-09-23
CN202011008309 2020-09-23
CN202110182212 2021-02-09
CN2021101822126 2021-02-09
CN2021104128127 2021-04-16
CN202110412812 2021-04-16
CN202110590591 2021-05-28
CN2021105905912 2021-05-28
PCT/CN2021/119985 WO2022063190A1 (en) 2020-09-23 2021-09-23 Pyrazine thiobiphenyl compound and application thereof

Publications (1)

Publication Number Publication Date
CN116348466A true CN116348466A (en) 2023-06-27

Family

ID=80844949

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180065019.2A Pending CN116348466A (en) 2020-09-23 2021-09-23 Pyrazine sulfur biphenyl compound and application thereof

Country Status (2)

Country Link
CN (1) CN116348466A (en)
WO (1) WO2022063190A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115210232A (en) * 2020-01-22 2022-10-18 南京明德新药研发有限公司 Pyrazolo heteroaromatic ring compound and application thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20240089A1 (en) 2021-05-05 2024-01-16 Revolution Medicines Inc RAS INHIBITORS FOR CANCER TREATMENT
EP4334324A1 (en) 2021-05-05 2024-03-13 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
IL308193A (en) 2021-05-05 2024-01-01 Revolution Medicines Inc Ras inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JO3517B1 (en) * 2014-01-17 2020-07-05 Novartis Ag N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
EP3310774B1 (en) * 2015-06-19 2020-04-29 Novartis AG Compounds and compositions for inhibiting the activity of shp2
CN112409334B (en) * 2018-05-09 2022-12-02 北京加科思新药研发有限公司 Heterocyclic derivatives useful as SHP2 inhibitors
CN112839935A (en) * 2018-09-26 2021-05-25 北京加科思新药研发有限公司 Novel heterocyclic derivatives useful as SHP2 inhibitors
TW202028183A (en) * 2018-10-10 2020-08-01 大陸商江蘇豪森藥業集團有限公司 Nitrogen-containing heteroaryl derivative regulators, preparation method and application thereof
WO2020156243A1 (en) * 2019-01-31 2020-08-06 贝达药业股份有限公司 Shp2 inhibitor and application thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115210232A (en) * 2020-01-22 2022-10-18 南京明德新药研发有限公司 Pyrazolo heteroaromatic ring compound and application thereof
CN115210232B (en) * 2020-01-22 2024-03-01 上海齐鲁制药研究中心有限公司 Pyrazolo heteroaromatic ring compound and application thereof

Also Published As

Publication number Publication date
WO2022063190A1 (en) 2022-03-31

Similar Documents

Publication Publication Date Title
CN116348466A (en) Pyrazine sulfur biphenyl compound and application thereof
CN115210232B (en) Pyrazolo heteroaromatic ring compound and application thereof
WO2022199587A1 (en) Pyrimidine heterocyclic compound and application thereof
JP7323748B2 (en) Pyrazolopyrimidine derivative and use thereof
CN114761411B (en) Spiro compounds as ERK inhibitors and application thereof
KR102345208B1 (en) Quinolino-pyrrolidin-2-one derivatives and their applications
CN112672994B (en) Heterospiro compounds serving as LSD1 inhibitor and application thereof
CN114555600B (en) Aminopyrimidines as CDK2/4/6 triple inhibitors
WO2021164742A1 (en) Quinoline compounds
WO2021129817A1 (en) Pyrimidine-based compound having inhibitory effect of ketohexokinase (khk)
WO2022156765A1 (en) Pyrazolopyrazine-linked tricyclic compound and application thereof
CN114072401B (en) Oxazole compounds as IRAK4 and BTK multi-target inhibitors
JP7296017B2 (en) Compounds containing benzosultam
CN117337278A (en) Alkyl carboxylic acid compounds and uses thereof
WO2021139814A1 (en) Quinoline imidazole compound and application thereof
TW202246279A (en) Pyrazinethiobiphenyl compounds and application thereof
CN113286594B (en) Application of pyridopyrimidine compounds in preparation of medicines for treating nasopharyngeal carcinoma
WO2022121900A1 (en) Pyrrolopyridine compound and application thereof
CN117279923A (en) Derivative of six-membered heteroaromatic urea ring and application thereof
CN116867771A (en) Oxadiazole substituted spiro compounds and application thereof
TW202246272A (en) Pyrazolopyrazine tricyclic compounds and uses thereof
CN117337286A (en) Methylsulfonyl urea compound and application thereof
CN117083285A (en) Pyrazine compound containing Se and application thereof
US20210253576A1 (en) Pyrazolopyrimidine derivative, use thereof and pharmaceutical composition

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
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20230721

Address after: 201203 Building 1, No. 56 Faraday Road and 576 Libing Road, China (Shanghai) Pilot Free Trade Zone, Pudong New Area, Shanghai

Applicant after: Shanghai Qilu Pharmaceutical Research Center Co.,Ltd.

Address before: 210032 room 218, business office building, No.9 Gaoxin Road, Jiangbei new district, Nanjing City, Jiangsu Province

Applicant before: Nanjing Mingde New Drug Development Co.,Ltd.