CN112566907A - Pyrazole derivatives as RET inhibitors - Google Patents

Pyrazole derivatives as RET inhibitors Download PDF

Info

Publication number
CN112566907A
CN112566907A CN201980053423.0A CN201980053423A CN112566907A CN 112566907 A CN112566907 A CN 112566907A CN 201980053423 A CN201980053423 A CN 201980053423A CN 112566907 A CN112566907 A CN 112566907A
Authority
CN
China
Prior art keywords
compound
isomer
pharmaceutically acceptable
acceptable salt
added
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
CN201980053423.0A
Other languages
Chinese (zh)
Inventor
陈正霞
张杨
戴美碧
李婕
龚珍
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.)
Medshine Discovery Inc
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 CN112566907A publication Critical patent/CN112566907A/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Abstract

A series of compounds with pyrazole structures and application thereof in preparing RET kinase inhibitors. The compound is specifically a derivative compound shown as a formula (III), an isomer thereof or a pharmaceutically acceptable salt thereof.

Description

Pyrazole derivatives as RET inhibitors
Reference to related applications
The following priority is claimed in the present application:
CN201810947337.1, App. No. 2018-08-17;
CN201811322012.0, application date 2018-11-07;
CN201811501055.5, application date 2018-12-07;
cn201910482535.x, application No. 2019-06-04;
CN201910487945.3, application date 2019-06-05.
Technical Field
The invention relates to a pyrazole derivative and application thereof in preparing a medicament for treating diseases related to RET kinase inhibitors. In particular to a compound shown as a formula (III) and pharmaceutically acceptable salts thereof.
Background
The RET protein is a receptor tyrosine kinase RTK and is also a transmembrane glycoprotein, expressed by the proto-oncogene RET (rearranged reduced transfection) located on chromosome 10, plays an important role in the development of the renal and enteric nervous systems during the embryonic stage, and is also critical in various tissue homeostasis, such as neurons, neuroendocrine, hematopoietic tissues and male germ cells. Unlike other RTKs, RET does not bind directly to the ligand molecule: such as neurotropic hormone (artemin), glial cell line-derived neurotrophic factor (GDNF), neurturin and persephin, which are ligands belonging to the GNDF Family (GFLs). These ligand GFLs generally bind to GDNF family receptor alpha (GFR alpha), and the resulting GFLs-GFR alpha complex mediates the auto-dimerization of RET proteins, causing a trans autophosphorylation reaction of tyrosine on the intracellular domain, recruitment of related adaptor proteins, activation of a cascade of signaling such as cell proliferation, and related signaling pathways including MAPK, PI3K, JAK-STAT, PKA, PKC, and the like.
There are two major oncogenic activation mechanisms of RET: one is that chromosomal rearrangements produce new fusion proteins, usually a fusion of the kinase domain of RET and a protein comprising a self-dimerization domain; secondly, the RET mutation directly or indirectly activates the kinase activity of RET. These alterations at the somatic or germ cell level are involved in the pathogenesis of a variety of cancers. RET chromosomal rearrangements are present in 5% -10% of papillary thyroid carcinoma patients; 60 percent of medullary thyroid medullary carcinoma has RET point mutation; among all NSCLC patients, there is probably 1-2% with RET fusion proteins, with KIF5B-RET being the most common.
In summary, aberrant RET expression or activation is found in a variety of tumors and gastrointestinal disorders such as irritable bowel syndrome. RET inhibitors are therefore of potential clinical value in neoplastic or bowel disorder diseases.
Disclosure of Invention
The invention provides a compound shown in a formula (III), an isomer thereof or a pharmaceutically acceptable salt thereof,
Figure PCTCN2019101121-APPB-000001
wherein the content of the first and second substances,
a ring is selected from
Figure PCTCN2019101121-APPB-000002
The above-mentioned
Figure PCTCN2019101121-APPB-000003
Optionally substituted by 1,2 or 3RgSubstitution;
R 1selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C3-6Cycloalkyl radical, said C1-3Alkyl radical, C1-3Alkoxy and C3-6Cycloalkyl is optionally substituted by 1,2 or 3RaSubstitution;
R 2selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RbSubstitution;
R 4selected from H, F, Cl、Br、I、OH、NH 2、CN、C 1-5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and a 5-6 membered heteroaromatic ring, said C1- 5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and 5-6 membered heteroaromatic rings optionally substituted with 1,2 or 3RdSubstitution;
R aand RbEach independently selected from F, Cl, Br, I, OH and NH2
R dSelected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C1-4An alkylamino group;
T 1、T 2、T 3and T5Each independently selected from CH and N;
T 4selected from the group consisting of CR5And N;
L 2selected from NH and O;
L 3is selected from-CH (R)6)-、-CH(R 6)CH 2-、-CHCH 2CH 2-、-CH(R 6)-O-、-CHCH 2-O-、-CH 2-N(R 6)-、-CH 2CH 2-N(R 6)-;
R 5Is selected from H;
R 6is selected from H and CH3
Or R5、R 6Are connected together to form a structural unit
Figure PCTCN2019101121-APPB-000004
Is selected from
Figure PCTCN2019101121-APPB-000005
R gSelected from H, F, Cl, Br, I, OH, NH2And CN;
the 5-to 6-membered heteroaryl group comprises 1,2,3 or 4 heteroatoms or groups of heteroatoms independently selected from-NH-, -O-, -S-and N, respectively.
The invention also provides a compound shown in the formula (II), an isomer thereof or a pharmaceutically acceptable salt thereof,
Figure PCTCN2019101121-APPB-000006
wherein the content of the first and second substances,
R 1selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RaSubstitution;
R 2selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RbSubstitution;
R 3selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RcSubstitution;
with "-" carbon atoms as chiral carbon atoms, in the form of (R) or (S) single enantiomers or enriched in one enantiomer;
R 4selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and a 5-6 membered heteroaromatic ring, said C1- 5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and 5-6 membered heteroaromatic ringsIs selected from 1,2 or 3RdSubstitution;
R a、R band RcEach independently selected from F, Cl, Br, I, OH and NH2
R dSelected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C1-4An alkylamino group;
T 1、T 2、T 3、T 4and T5Each independently selected from CH and N;
a ring is selected from
Figure PCTCN2019101121-APPB-000007
The above-mentioned
Figure PCTCN2019101121-APPB-000008
Optionally substituted by 1,2 or 3RgSubstitution;
L 1and L2Each independently selected from NH and O;
R gselected from H, F, Cl, Br, I, OH, NH2And CN;
the 5-to 6-membered heteroaryl group comprises 1,2,3 or 4 heteroatoms or groups of heteroatoms independently selected from-NH-, -O-, -S-and N, respectively.
The invention also provides a compound shown in the formula (I) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2019101121-APPB-000009
wherein the content of the first and second substances,
R 1selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RaSubstitution;
R 2selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RbSubstitution;
R 3selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RcSubstitution;
with "-" carbon atoms as chiral carbon atoms, in the form of (R) or (S) single enantiomers or enriched in one enantiomer;
R 4selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and a 5-6 membered heteroaromatic ring, said C1- 5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and 5-6 membered heteroaromatic rings optionally substituted with 1,2 or 3RdSubstitution;
R a、R band RcEach independently selected from F, Cl, Br, I, OH and NH2
R dSelected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C1-4An alkylamino group;
T 1、T 2、T 3、T 4and T5Each independently selected from CH and N;
a ring is selected from
Figure PCTCN2019101121-APPB-000010
The above-mentioned
Figure PCTCN2019101121-APPB-000011
Optionally substituted by 1,2 or 3RgSubstitution;
R gselected from H, F, Cl, Br, I, OH, NH2And CN;
the 5-to 6-membered heteroaryl group comprises 1,2,3 or 4 heteroatoms or groups of heteroatoms independently selected from-NH-, -O-, -S-and N, respectively.
In some embodiments of the invention, R is1Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3
Figure PCTCN2019101121-APPB-000012
The CH3、CH 2CH 3
Figure PCTCN2019101121-APPB-000013
Optionally substituted by 1,2 or 3RaAnd (4) substitution.
In some embodiments of the invention, R is1Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000014
the CH3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000015
optionally substituted by 1,2 or 3RaAnd the other variables are as defined herein.
In some embodiments of the invention, R is1Selected from H, F, Cl、Br、I、OH、NH 2、CN、CH 3、CH 2F、CHF 2、CF 3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000016
other variables are as defined herein.
In some embodiments of the invention, R is1Is selected from CH3And
Figure PCTCN2019101121-APPB-000017
other variables are as defined herein.
In some embodiments of the invention, R is1Is selected from CH3The other variables are as defined herein.
In some embodiments of the invention, R is2Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000018
the CH3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000019
optionally substituted by 1,2 or 3RbAnd the other variables are as defined herein.
In some embodiments of the invention, R is2Is selected from CH3The other variables are as defined herein.
In some embodiments of the invention, R is3Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000020
the CH3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000021
optionally substituted by 1,2 or 3RcAnd the other variables are as defined herein.
In some embodiments of the invention, R is3Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2F、CHF 2、CF 3、CH 2CH 3And
Figure PCTCN2019101121-APPB-000022
other variables are as defined herein.
In some embodiments of the invention, R is3Is selected from CH3The other variables are as defined herein.
In some embodiments of the invention, R isdSelected from H, F, Cl, Br, I, OH, NH2、CN、CH 3And
Figure PCTCN2019101121-APPB-000023
other variables are as defined herein.
In some embodiments of the invention, R is4Selected from: H. f, Cl, Br, I, OH, NH2、CN、CH 3Pyrazolyl, isoxazolyl, imidazolyl, triazolyl, oxazolyl,
Figure PCTCN2019101121-APPB-000024
The CH3Pyrazolyl, isoxazolylImidazolyl, triazolyl, oxazolyl,
Figure PCTCN2019101121-APPB-000025
Optionally substituted by 1,2 or 3RdAnd the other variables are as defined herein.
In some embodiments of the invention, R is4Selected from:
Figure PCTCN2019101121-APPB-000026
Figure PCTCN2019101121-APPB-000027
other variables are as defined herein.
In some embodiments of the invention, the structural unit
Figure PCTCN2019101121-APPB-000028
Is selected from
Figure PCTCN2019101121-APPB-000029
Figure PCTCN2019101121-APPB-000030
Other variables are as defined herein.
In some embodiments of the invention, the A ring is selected from
Figure PCTCN2019101121-APPB-000031
Other variables are as defined herein.
In some embodiments of the invention, the A ring is selected from
Figure PCTCN2019101121-APPB-000032
OthersThe variables are as defined herein.
In some embodiments of the invention, the structural unit
Figure PCTCN2019101121-APPB-000033
Is selected from
Figure PCTCN2019101121-APPB-000034
Figure PCTCN2019101121-APPB-000035
Other variables are as defined herein.
In some embodiments of the invention, the structural unit
Figure PCTCN2019101121-APPB-000036
Is selected from
Figure PCTCN2019101121-APPB-000037
Figure PCTCN2019101121-APPB-000038
Other variables are as defined herein.
In some embodiments of the invention, the structural unit
Figure PCTCN2019101121-APPB-000039
Is selected from
Figure PCTCN2019101121-APPB-000040
Other variables are as defined herein.
In some embodiments of the invention, the compound, isomer thereof or pharmaceutically acceptable salt thereof is selected from
Figure PCTCN2019101121-APPB-000041
Figure PCTCN2019101121-APPB-000042
Wherein R is1、R 2、R 3、R 4、L 2、L 3、T 1、T 2、T 3、T 4And T5As defined herein.
Further aspects of the invention are derived from any combination of the above variables.
The present invention provides a compound represented by the following formula, an isomer thereof, or a pharmaceutically acceptable salt thereof, selected from:
Figure PCTCN2019101121-APPB-000043
Figure PCTCN2019101121-APPB-000044
Figure PCTCN2019101121-APPB-000045
in some embodiments of the invention, the compound, isomer thereof, or pharmaceutically acceptable salt thereof, is selected from the group consisting of:
Figure PCTCN2019101121-APPB-000046
Figure PCTCN2019101121-APPB-000047
Figure PCTCN2019101121-APPB-000048
Figure PCTCN2019101121-APPB-000049
the invention also provides a pharmaceutical composition which contains the compound, the isomer or the pharmaceutically acceptable salt thereof with effective treatment amount as an active ingredient and a pharmaceutically acceptable carrier.
The invention also provides application of the compound, the isomer or the pharmaceutically acceptable salt thereof in preparing RET kinase inhibitors.
The invention also provides application of the composition in preparing RET kinase inhibitors.
Definitions and explanations
As used herein, the following terms and phrases are intended to have the following meanings, unless otherwise indicated. A particular term or phrase, unless specifically defined, should not be considered as indefinite or unclear, but rather construed according to ordinary meaning. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
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 salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amines or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of acid in neat 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 salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are salts of amino acids such as arginine and the like, and salts of organic acids such as glucuronic acid and the like. Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains an acid or base, by conventional chemical methods. In general, such salts are prepared by the following method: prepared by reacting 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 the two.
The compounds of the present 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, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
Unless otherwise indicated, the terms "enantiomer" or "optical isomer" refer to stereoisomers that are mirror images of each other.
Unless otherwise indicated, the term "cis-trans isomer" or "geometric isomer" results from the inability of a double bond or a single bond to rotate freely within a ring-forming carbon atom.
Unless otherwise indicated, the term "diastereomer" refers to a stereoisomer in which the molecules have two or more chiral centers and a non-mirror image relationship between the molecules.
Unless otherwise indicated, "(D)" or "(+)" means dextrorotation, "(L)" or "(-) -means levorotation," (DL) "or" (±) "means racemization.
Using solid wedge keys, unless otherwise indicated
Figure PCTCN2019101121-APPB-000050
And wedge dotted bond
Figure PCTCN2019101121-APPB-000051
Showing the absolute configuration of a solid centre, by means of straight solid keys
Figure PCTCN2019101121-APPB-000052
And straight dotted line bond
Figure PCTCN2019101121-APPB-000053
Showing the relative configuration of the centres of solids, by wavy lines
Figure PCTCN2019101121-APPB-000054
Representing solid-line keys of wedge shape
Figure PCTCN2019101121-APPB-000055
Or wedge dotted bond
Figure PCTCN2019101121-APPB-000056
Or by wavy lines
Figure PCTCN2019101121-APPB-000057
Indicating straight solid-line keys
Figure PCTCN2019101121-APPB-000058
And straight dotted line bond
Figure PCTCN2019101121-APPB-000059
The compounds of the invention may be present specifically. Unless otherwise indicated, the term "tautomer" or "tautomeric form" means that at room temperature, the isomers of different functional groups are in dynamic equilibrium and can be rapidly interconverted. If tautomers are possible (e.g., in solution), then the chemical equilibrium of the tautomers can be reached. For example, proton tautomers (prototropic tautomers), also known as proton transfer tautomers (prototropic tautomers), include interconversions by proton transfer, such as keto-enol isomerization and imine-enamine isomerization. Valence isomers (valencetatomer) include interconversion by recombination of some of the bonding electrons. A specific example of where keto-enol tautomerism is the interconversion between two tautomers of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one.
Unless otherwise indicated, the terms "enriched in one isomer", "isomer enriched", "enantiomer enriched" or "enantiomeric enrichment" refer to a content of one isomer or enantiomer of 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 "isomeric excess" or "enantiomeric excess" refers to the difference between the relative percentages of two isomers or enantiomers. For example, if the content of one isomer or enantiomer is 90%, and the content of the other isomer or enantiomer is 10%, the isomer or enantiomer excess (ee value) is 80%.
Optically active (R) -and (S) -isomers as well as D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one of the enantiomers of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is 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), diastereomeric salts are formed with an appropriate optically active acid or base, followed by diastereomeric resolution by conventional methods known in the art, and the pure enantiomers are recovered. Furthermore, separation of enantiomers and diastereomers is typically accomplished by using chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (e.g., carbamate formation from amines).
The compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be labelled with radioactive isotopes, such as tritium (A), (B), (C3H) Iodine-125 (125I) Or C-14(14C) In that respect For example, deuterium can be used to replace hydrogen to form deuterium-substituted drug, which has stronger bond than common hydrogen and carbon, and has reduced adverse side effect, increased stability and enhanced therapeutic effect compared with non-deuterated drugAnd the biological half-life period of the medicine is prolonged. 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.
"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 replaced with a substituent, and may include variations of deuterium and hydrogen, so long as the valency 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 aromatic groups. The term "optionally substituted" means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemical realizability.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 0-2R, the group may optionally be substituted with up to two R, and there are separate options for R in each case. Furthermore, combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
When the number of one linking group is 0, e.g. - (CRR)0-, represents that the linking group is a single bond.
When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly connected, for example, where L represents a single bond in A-L-Z means that the structure is actually A-Z.
When a substituent is absent, it indicates that the substituent is absent, e.g., when X is absent in A-X, it indicates that the structure is actually A. When the listed linking groups do not indicate their direction of attachment, the direction of attachment is arbitrary, for example,
Figure PCTCN2019101121-APPB-000060
wherein the linking group L is-M-W-, in which case-M-W-can be formed by connecting the ring A and the ring B in the same direction as the reading sequence from left to right
Figure PCTCN2019101121-APPB-000061
The ring A and the ring B may be connected in the reverse direction of the reading sequence from left to right
Figure PCTCN2019101121-APPB-000062
Combinations of the linking groups, substituents, and/or variants thereof are permissible only if such combinations result in stable compounds.
Unless otherwise specified, the term "C1-5Alkyl "is intended to mean a straight or branched saturated hydrocarbon group consisting of 1 to 5 carbon atoms. Said C is1-5The alkyl group comprising C1-4、C 1-3、C 1-2、C 2-5、C 2-4And C5Alkyl, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C1-5Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, s-butyl and t-butyl), pentyl (including n-pentyl, isopentyl and neopentyl), and the like.
Unless otherwise specified, the term "C1-3Alkyl "is intended to mean a straight or branched saturated hydrocarbon group consisting of 1 to 3 carbon atoms. Said C is1-3The alkyl group comprising C1-2And C2-3Alkyl, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C1- 3Examples 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" is2-8Alkenyl "is intended to mean a straight-chain or branched hydrocarbon group consisting of 2 to 8 carbon atoms containing at least one carbon-carbon double bond, which may be located anywhere in the group. Said C is2-8Alkenyl radicals comprising C2-6、C 2-4、C 2-3、C 4、C 3And C2Alkenyl and the like; it may be monovalent, divalent or polyvalent. C2-8Examples of alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl, butadienyl, piperylene, hexadienyl, and the like.
Unless otherwise specified, the term "C1-5Alkoxy "denotes those alkyl groups containing 1 to 5 carbon atoms which are attached to the rest of the molecule through an oxygen atom. Said C is1-5Alkoxy radicals comprising C1-5、C 1-4、C 1-3、C 1-2、C 2-5、C 2-4、C 5、C 4、C 3And C2Alkoxy, and the like. C1- 5Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), butoxy (including n-butoxy, isobutoxy, s-butoxy and t-butoxy), pentyloxy (including n-pentyloxy, isopentyloxy and neopentyloxy), and the like.
Unless otherwise specified, the term "C1-3Alkoxy "denotes those alkyl groups containing 1 to 3 carbon atoms which are attached to the rest of the molecule through an oxygen atom. Said C is1-3Alkoxy radicals comprising C1-2、C 2-3、C 3And C2Alkoxy, and the like. C1-3Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), and the like.
Unless otherwise specified, the term "C1-5Alkylamino "refers to those alkyl groups containing from 1 to 5 carbon atoms that are attached to the rest of the molecule through an amino group. Said C is 1-6Alkylamino radicals comprising C1-5、C 1-4、C 1-3、C 1-2、C 2-5、C 2-4、C 5、C 4、C 3And C2Alkylamino, and the like. C1-5Examples of alkylamino include, but are not limited to, -NHCH3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 3)CH 2CH 3、-N(CH 2CH 3)(CH 2CH 3)、-NHCH 2CH 2CH 3、-NHCH 2(CH 3) 2、-NHCH 2CH 2CH 2CH 3And the like.
Unless otherwise specified, the term "C1-4Alkylamino "refers to those alkyl groups containing from 1 to 4 carbon atoms that are attached to the rest of the molecule through an amino group. Said C is1-4Alkylamino radicals comprising C1-3、C 1-2、C 2-4、C 4、C 3And C2Alkylamino, and the like. C1-4Examples of alkylamino include, but are not limited to, -NHCH3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 3)CH 2CH 3、-N(CH 2CH 3)(CH 2CH 3)、-NHCH 2CH 2CH 3、-NHCH 2(CH 3) 2、-NHCH 2CH 2CH 2CH 3And the like.
Unless otherwise specified, "C" is3-6Cycloalkyl "denotes a saturated cyclic hydrocarbon group consisting of 3 to 6 carbon atoms, being a monocyclic and bicyclic ring system, said C3-6Cycloalkyl radicals including C3-5、C 4-5And C5-6Cycloalkyl groups and the like; it may be monovalent, divalent or polyvalent. C3-6Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
Unless otherwise specified, the terms "5-6 membered heteroaromatic ring" and "5-6 membered heteroaryl" are used interchangeably herein, and the term "5-6 membered heteroaryl" denotes a monocyclic group consisting of 5 to 6 ring atoms with a conjugated pi-electron system, of which 1,2,3 or 4 ring atoms are heteroatoms independently selected from O, S and N, the remainder being carbon atoms. Wherein the nitrogen atoms are optionally quaternized and the nitrogen and sulfur heteroatoms are optionally oxidized (i.e., NO and S (O))pAnd p is 1 or 2). The 5-6 membered heteroaryl group may be attached to the rest of the molecule through a heteroatom or a carbon atom. The 5-6 membered heteroaryl group includes 5-and 6-membered heteroaryl groups. Examples of such 5-6 membered heteroaryl groups include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, and the like), pyrazolyl (including 2-pyrazolyl, 3-pyrazolyl, and the like), imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, and the like), oxazolyl (including 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, and the like), triazolyl (1H-1,2, 3-triazolyl, 2H-1,2, 3-triazolyl, 1H-1,2, 4-triazolyl, and 4H-1,2, 4-triazolyl, and the like), tetrazolyl, isoxazolyl (3-isoxazolyl, 4-isoxazolyl, and 5-isoxazolyl, and the like), Thiazolyl (including 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, and the like), furyl (including 2-furyl, 3-furyl, and the like), thienyl (including 2-thienyl, 3-thienyl, and the like), pyridyl (including 2-pyridyl, 3-pyridyl, 4-pyridyl, and the like), pyrazinyl or pyrimidinyl (including 2-pyrimidinyl, 4-pyrimidinyl, and the like).
Unless otherwise specified, Cn-n+mOr Cn-C n+mIncluding any one particular case of n to n + m carbons, e.g. C1-12Comprising C1、C 2、C 3、C 4、C 5、C 6、C 7、C 8、C 9、C 10、C 11And C12Also, any one of n to n + m is includedRanges, e.g. C1-12Comprising C1- 3、C 1-6、C 1-9、C 3-6、C 3-9、C 3-12、C 6-9、C 6-12And C9-12Etc.; similarly, n to n + m means the number of atoms on the ring is n to n + m, for example, the 3-12 membered ring includes a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, a 9-membered ring, a 10-membered ring, a 11-membered ring, and a 12-membered ring, and any range of n to n + m is also included, for example, the 3-12 membered ring includes a 3-6-membered ring, a 3-9-membered ring, a 5-6-membered ring, a 5-7-membered ring, a 6-8-membered ring, and a 6-.
Unless otherwise specified, the term "halogen" or "halogen" by itself or as part of another substituent means a fluorine, chlorine, bromine or iodine atom. Furthermore, the term "haloalkyl" is intended to include monohaloalkyl and polyhaloalkyl. For example, the term "halo (C)1-C 4) Alkyl "is intended to include, but not be limited to, trifluoromethyl, 2, 2, 2-trifluoroethyl, 4-chlorobutyl, and 3-bromopropyl, and the like. Unless otherwise specified, examples of haloalkyl include, but are not limited to: trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
The term "leaving group" refers to a functional group or atom that can be substituted by another functional group or atom through a substitution reaction (e.g., an affinity substitution reaction). For example, representative leaving groups include triflate; chlorine, bromine, iodine; sulfonate groups such as methanesulfonate, toluenesulfonate, p-bromobenzenesulfonate, p-toluenesulfonate and the like; acyloxy groups such as acetoxy, trifluoroacetyloxy, and the like.
The term "protecting group" includes, but is not limited to, "amino protecting group," hydroxyl protecting group, "or" thiol protecting group. The term "amino protecting group" refers to a protecting group suitable for use in preventing side reactions at the amino nitrogen position. Representative amino protecting groups include, but are not limited to: a formyl group; acyl, for example alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl such as tert-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 hydroxy protecting groups include, but are not limited to: alkyl groups such as methyl, ethyl and tert-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.
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 listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention.
The compounds of the present invention may have a variety of uses or indications, including but not limited to the specific uses or indications enumerated herein.
The solvent used in the present invention can be commercially available. The invention employs the following abbreviations: aq represents water; HATU represents O- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate; EDC stands for N- (3-dimethylaminopropyl) -N' -ethylcarbodiimide hydrochloride; m-CPBA represents 3-chloroperoxybenzoic acid; eq represents equivalent, equivalent; m represents mol/L; CDI represents carbonyldiimidazole; DCM represents dichloromethane; PE represents petroleum ether; DIAD represents diisopropyl azodicarboxylate; DMF represents N, N-dimethylformamide; DMSO represents dimethyl sulfoxide; EtOAc for ethyl acetate; EtOH stands for ethanol; MeOH represents methanol; CBz represents benzyloxycarbonyl, an amine protecting group; BOC represents tert-butoxycarbonyl as an amine protecting group; HOAc represents acetic acid; n acetonitrile BH3Represents sodium cyanoborohydride; r.t. represents room temperature; O/N stands for overnight; THF represents tetrahydrofuran; boc2O represents di-tert-butyl dicarbonateAn ester; trifluoroacetic acid represents trifluoroacetic acid; DIPEA stands for diisopropylethylamine; SOCl2Represents thionyl chloride; CS2Represents carbon disulfide; TsOH represents p-toluenesulfonic acid; NFSI represents N-fluoro-N- (phenylsulfonyl) benzenesulfonamide; NCS represents 1-chloropyrrolidine-2, 5-dione; n-Bu4NF represents tetrabutyl ammonium fluoride; iPrOH represents 2-propanol; mp represents melting point; LDA stands for lithium diisopropylamide.
The compounds are used according to the conventional naming principle in the field
Figure PCTCN2019101121-APPB-000063
The software names, and the commercial compounds are under the supplier catalog name.
Technical effects
The compound has excellent inhibitor activity on RET and mutant RET V804M thereof, and has excellent treatment effect on patients with RET abnormal tumors.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. Having described the invention in detail and having disclosed specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof.
Example 1
Figure PCTCN2019101121-APPB-000064
Step one
A catalytic amount of DMF (2.32g,31.74mmol) was added to a mixture of 6-chloronicotinic acid (50g,317.35mmol) containing thionyl chloride (150mL) and refluxed at 85 ℃ for 3 hours under nitrogen. The solution was spun off and dissolved in DCM (1200mL), methoxy-N-methyl-amino hydrochloride (46.43g,476.03mmol) was added, cooled to 0 deg.C, triethylamine (96.34g,952.05mmol) was added dropwise, and stirring was carried out at 25 deg.C for 16 hours. Adding saturated sodium bicarbonate solution into the reaction solution, and layering to obtain an organic phase. The organic phase was dried over sodium sulfate and concentrated under reduced pressure to give 1 a.
LCMS(ESI)m/z:200.9[M+1] +
Step two
Methylmagnesium bromide (3M,119.04mL) was added dropwise to a solution of 1a (59.71g,297.60mmol) in THF (500mL) at 0 deg.C and stirred for 2h at 25 deg.C. The reaction mixture was quenched by addition of saturated ammonium chloride (500mL), extracted with EtOAc (500 mL. times.2), the combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give the crude product. The crude product was isolated by flash column chromatography on silica gel (PE: EtOAC ═ 3:1) to give 1 b.
Step three
To a solution of 4-fluoro-pyrazole (4.98g,57.85mmol), 1b (9g,57.85mmol) in DMF (80mL) was added potassium carbonate (19.99g,144.62mmol), and the mixture was stirred at 100 ℃ for 16 hours. Water (400mL) was added to the reaction solution, EtOAc (400 mL. times.2) was added thereto for extraction, and the organic phases were combined. The organic phase was washed successively with water (600mL) and saturated aqueous sodium carbonate (600 mL). Finally, the organic phase is dried by anhydrous sodium sulfate and is decompressed and concentrated to obtain a crude product. The crude product was isolated by flash column chromatography on silica gel (PE: EtOAC ═ 3:1) to give 1 c.
LCMS(ESI)m/z:205.9[M+1] +
Step four
To a solution of 1c (1g,4.87mmol) in THF (10mL) was added (R) -tert-butylsulfinamide (590.68mg,4.87mmol), followed by the addition of tetraethyltitanium oxide (2.62g,9.75mmol,2.38mL, 85% purity), stirring at 75 deg.C for 20 hours, cooling to-78 deg.C, and dropwise addition of a solution of lithium tri-sec-butylborohydride (1M,14.62mL) and stirring at-78 deg.C for 30 minutes. The reaction was quenched by addition of MeOH (5mL) to the reaction solution at-78 deg.C, followed by addition of water (20mL) and extraction with EtOAc (20 mL. times.2). The organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure to give crude product, which was separated by flash silica gel column (DCM: MeOH ═ 10:1) to give 1 d.
LCMS(ESI)m/z:310.9[M+1] +,333.1[M+23] +
Step five
To 1d (8g,25.77mmol) were added MeOH (67.30g,2.10mol,85mL) and hydrogen chloride in dioxane (4M,85mL,13.19eq) and stirred at 30 ℃ for 2 h. The reaction solution is decompressed, concentrated and dried to obtain a crude product, then water is added for dilution, and the pH value is adjusted to 8 by using a saturated sodium carbonate solution. Adding dichloromethane for extraction to obtain an organic phase. The organic phase is decompressed and concentrated to obtain a crude product. The crude product was isolated via flash silica gel column (DCM: MeOH ═ 10:1) to give 1 e. Chiral resolution by HPLC (column: DAICEL CHIRALPAK AD-H (250 mm. times.50 mm,10 μm); mobile phase: [ 0.1% ammonia EtOH ]; B%: 35% -35%, retention time: 5.386 min, main peak 2 was collected) to give 1e (99.85% ee%).
1H NMR(400MHz,DMSO-d 6)δ8.66(d,J=4.02Hz,1H),8.42(d,J=2.01Hz,1H),7.99(dd,J=2.01,8.53Hz,1H),7.87-7.91(m,1H),7.84(d,J=8.53Hz,1H),4.08(q,J=6.53Hz,1H),1.72-2.34(m,2H),1.29(d,J=6.53Hz,3H).
Step six
2, 4-dichloro-6-methyl-pyrimidine (10g,61.35mmol), 5-methyl-3-amino-1H-pyrazole (6.55g,67.48mmol), diisopropylethylamine (11.89g,92.02mmol,16.03mL) was added to DMSO (30mL), and the reaction was stirred at 60 ℃ for 16 hours. Cooling the reaction liquid to 20-30 ℃, pouring the reaction liquid into 200mL of ice water, stirring for 1 hour at 15-20 ℃, filtering, and performing reduced pressure spin drying on a filter cake at 40-50 ℃ to obtain 1 f.
LCMS(ESI)m/z:224.0[M+1] +,225.9[M+3] +
1H NMR(400MHz,DMSO-d 6)δppm 12.08(s,1H)10.14(s,1H)6.50-7.68(m,1H)5.55-6.45(m,1H)2.25(s,3H)2.20(s,3H).
Step seven
1f (30mg, 134.13. mu. mol), tert-butoxycarbonylpiperidine (44.83mg, 240.69. mu. mol), diisopropylethylamine (69.34mg, 536.52. mu. mol, 93.45. mu.L) was added to N-methylpyrrolidone (2 mL). The reaction was stirred at 90 ℃ for 16 hours, the reaction was cooled to room temperature, 5mL of water was added, followed by extraction with EtOAc (5 mL. times.2), and the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 1 g.
LCMS(ESI)m/z:374.7[M+1] +,318.2[M-55] +
Step eight
1g (200mg, 535.54. mu. mol) was added to EtOAc (5mL), and a solution of hydrogen chloride in ethyl acetate (4M, 267.77. mu.L) was added dropwise to the reaction mixture, followed by stirring at 20-30 ℃ for 16 hours. And (3) directly carrying out decompression spin-drying on the reaction liquid at 40-50 ℃ for 1 h.
LCMS(ESI)m/z:274.0[M+1] +
1H NMR(400MHz,DMSO-d6)δppm 11.18(m,1H)9.81(s,2H)6.18-6.43(m,1H)4.21-4.33(m,4H)3.25(m,4H)2.42(s,3H)2.28-2.31(m,1H)2.26(s,3H).
Step nine
Triphosgene (23.95mg, 80.70. mu. mol) was added to DCM (4mL), diisopropylethylamine (62.58mg,
484.19 μmol,84.33 μ L) was added to the reaction solution, 1e (26.63mg,129.12 μmol) was added to the reaction solution with stirring at a reaction temperature of 0 to 5 ℃, the reaction was stirred for 10 minutes at 0 to 5 ℃, then 1h (50mg,161.40 μmol) and diisopropylethylamine (62.58mg,484.19 μmol,84.33 μ L) were added to the reaction solution, and the system was stirred for 1 hour at 0 to 5 ℃. The reaction solution is directly decompressed and concentrated to obtain a crude product. The crude product was separated and purified by preparative chromatography (HCl system) to give the hydrochloride salt of example 1, the hydrochloride salt of example 1 was added to sodium bicarbonate solution, extracted with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 1.
LCMS(ESI)m/z:506.1[M+1] +
1H NMR(400MHz,MeOH-d 4)δppm 8.52(d,J=4.27Hz,1H),8.44(s,1H),7.98-8.06(m,1H),7.89-7.97(m,1H),7.72(d,J=4.02Hz,1H),6.19-6.65(m,2H),5.00-5.08(m,1H),3.91(m,4H),3.69(m,4H),2.46(s,3H),2.41(s,3H),1.57(d,J=7.03Hz,3H).
Examples 2 and 3
Figure PCTCN2019101121-APPB-000065
Step one
Compound 2a (500mg,3.21mmol), dimethylamine hydrochloride (1.81g,16.07mmol) was added to ethanol (5mL) and stirred at 80 ℃ under reflux for 2 hours. And cooling the reaction liquid to 25 ℃, then spin-drying, adding tert-butyl methyl ether for dissolving, washing with 10% citric acid, adjusting the pH to 10-11 with 1mol/L sodium hydroxide, extracting for 3 times with 20mL of tert-butyl methyl ether, collecting the organic phase, drying with anhydrous sodium sulfate, filtering, and spin-drying to obtain 2 b.
LCMS(ESI)m/z:164.8[M+1] +
1H NMR(400MHz,CDCl 3)δppm 8.67-8.86(m,1H),7.90-8.11(m,1H),6.36-6.66(m,1H),3.18(s,6H),2.49(s,3H).
Step two
Add 2b (220mg,1.34mmol) to MeOH (10mL) followed by ammonium acetate (1.03g,13.40mmol) and stir at 65 deg.C for 1h, after complete dissolution add sodium cyanoborohydride (252.59mg,4.02mmol) and stir at 65 deg.C overnight. The reaction solution after the reaction of the raw materials is directly spin-dried to obtain a crude product. The crude product was purified by preparative chromatography plate (DCM/MeOH-10/1, Rf-0.2.3) to give 2 c.
LCMS(ESI)m/z:165.8[M+1] +
1H NMR(400MHz,CDCl 3)δppm 8.11(d,J=2.00Hz,1H),7.59(dd,J=8.80,2.64Hz,1H),6.55(d,J=9.20Hz,1H),4.27-4.29(m,1H),2.93-3.23(m,16H),1.57(d,J=6.80Hz,3H).
Step three
2c (57mg, 344.96. mu. mol) DIEA (133.75mg,1.03mmol, 180.25. mu.L, 3eq) and triphosgene (51.18mg, 172.48. mu. mol) were added to DCM (2mL) at 0 ℃ and stirred for 10min, and a mixture of compound 1h (113.15mg, 413.95. mu. mol), DIEA (133.75mg,1.03mmol) and DCM (2mL) at 0 ℃ was added to the above reaction system and reacted at 0 ℃ for 20 min. Directly spin-drying the reaction solution to obtain a crude product. The crude product is subjected to preparative chromatography (column: Xtimate C18150X 25mm X5 μm; mobile phase: water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 10% -30%, 7min), separated and purified, and then subjected to SFC resolution (column: DAICEL CHIRALPAK AS-H (250mm X30 mm,5 μm); mobile phase: 0.1% ammonia EtOH; B%: 45% -45%)
Example 2 (retention time: 4.957min) was obtained.
LCMS(ESI)m/z:465.4[M+1] +
1H NMR(400MHz,MeOH-d 4)δppm 8.03(d,J=2.80Hz,1H),7.55(dd,J=8.80,2.51Hz,1H),6.65(d,J=8.80Hz,1H),6.11-6.12(m,2H),3.74-3.78(m,4H),3.48-3.52(m,4H),3.01(s,6H),2.28(s,3H),2.21(s,3H).
Example 3 (retention time: 5.737min) was obtained.
LCMS(ESI)m/z:465.4[M+1] +
1H NMR(400MHz,MeOH-d 4)δppm 8.03(d,J=2.80Hz,1H),7.56(dd,J=8.80,2.51Hz,1H),6.64(d,J=8.80Hz,1H),6.11-6.12(m,2H),3.75-3.79(m,4H),3.48-3.51(m,4H),3.04(s,6H),2.28(s,3H),2.21(s,3H).
Example 4
Figure PCTCN2019101121-APPB-000066
Step one
2, 6-dichloro, 4-methylpyridine (5g,30.86mmol), N-Boc piperazine (6.32g,33.95mmol) and cesium carbonate (30.17g,92.58mmol) were added together to DMF (100mL) and the reaction was stirred in an oil bath heated to 120 ℃ for 16 h. Directly filtering the reaction solution, collecting the filtrate, and directly spin-drying to obtain a crude product. The crude product was purified by flash silica gel column (PE/EtOAc ═ 5/1, Rf ═ 0.5) to give 4 b.
1H NMR(400MHz,CDCl3)δ6.49(s,1H),6.30(s,1H),3.51(s,8H),2.24(s,3H),1.48(s,9H).
Step two
4b (373.76mg,3.85mmol) was dissolved in 1, 4-dioxane (10mL) and Pd was then added2(dba) 3(146.84mg, 160.36. mu. mol), 5-methyl-3-amino-1H-pyrazole (1g,3.21mmol), 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl (229.33mg, 481.07. mu. mol) and cesium carbonate (3.13g,9.62mmol) were added together to the reaction solution, heated to 140 ℃ by a microwave synthesizer and reacted for 1 hour with stirring. Directly filtering the reaction solution, collecting the filtrate, and spin-drying to obtain a crude product. Crude product passing through quick siliconPurification on a gel column (PE/EtOAc ═ 1/1, Rf ═ 0.2) afforded product 4 c.
LCMS(ESI)m/z:373.3[M+H] +
Step three
4c (100mg, 268.48. mu. mol) was dissolved in EtOAc (5mL), followed by dropwise addition of HCl/EtOAc (4M,10mL) and finally the reaction was stirred at 20-30 ℃ for 2 h. As the reaction proceeds, the solids in the reaction solution gradually increase. The reaction solution was stirred for further 16 hours. LCMS detection showed disappearance of starting material and the main peak was the product peak. Directly concentrating the reaction solution to dryness to obtain 4 d.
LCMS(ESI)m/z:273.2[M+1] +
Step four
Compound 4d (40mg, 146.87. mu. mol) and N, N-diisopropylethylamine (56.95mg, 440.61. mu. mol, 76.75. mu.L) were added to methylene chloride (5mL) at 0 ℃ followed by the addition of compound 1e (36.35mg, 176.25. mu. mol), N, N-diisopropylethylamine (56.95mg, 440.61. mu. mol) and triphosgene (21.79mg, 73.44. mu. mol) and the reaction was stirred at 25-30 ℃ for 10 minutes. Directly spin-drying the reaction solution at 20-30 ℃ to obtain a crude product. The crude product was sent to a preparative chromatography column (column: Xtimate C18150X 25mm X5 μm; mobile phase: [ water (0.225% trifluoroacetic acid) -acetonitrile ]; B%: 25% -45%, 7 min). The trifluoroacetate salt of example 4 is obtained, the trifluoroacetate salt of example 4 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 4.
LCMS(ESI)m/z:505.3[M+1] +
1H NMR(400MHz,MeOH-d 4)δ8.47(br s,2H),7.98~7.90(m,1H),7.84(s,1H),7.69(s,1H),6.34(s,1H),6.23(s,1H),5.83(s,1H),5.02~4.99(m,1H),3.66-3.74(m,8H),2.37(s,3H),2.33(s,3H),1.57(d,J=6.8Hz,3H).
Examples 6 and 7
Figure PCTCN2019101121-APPB-000067
Step one
4-pyrazole-1-benzoic acid (7g,37.20mmol) and methoxymethylamide hydrochloride (5.44g,55.80mmol) were dissolved in DMF (110mL), HATU (14.14g,37.20mmol) and DIPEA (24.04g,185.99mmol,32.40mL) were added, and the mixture was stirred at 30 ℃ for 16 hours. LCMS indicated product formation. The reaction mixture was extracted with ethyl acetate (100 ml. times.3) and the combined organic phases were washed with water (100 ml. times.2), saturated sodium chloride solution (100 ml. times.2), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 6 a.
1H NMR(400MHz,CDCl 3-d 6)δ7.95(d,J=4Hz,1H),7.74-7.81(m,2H),7.58-7.73(m,3H),6.36-6.49(m,1H),3.51(s,3H),3.32(s,3H).
Step two
To a solution of compound 6a (9.4g,40.65mmol) in THF (90mL) was added dropwise methylmagnesium bromide (3M,27.10mL) at 0 deg.C, slowly warmed to 30 deg.C and stirred for 1 hour. The reaction solution was quenched by addition of a saturated ammonium chloride solution, followed by extraction with ethyl acetate (150 ml. times.3), and the organic layers were combined, washed with a saturated sodium chloride solution (200 ml. times.3), dried over anhydrous sodium sulfate, and finally the solvent was removed by rotary removal under reduced pressure to give a crude product. And (3) performing silica gel column chromatography (PE: EA is 3:1, and Rf is 0.57), finding that a product is easy to separate out, adding a small amount of ethyl acetate into a cross product obtained by column chromatography to dissolve the cross product, adding a large amount of petroleum ether, washing out white solid, and filtering to obtain a product 6 b.
Step three
MeOH (50mL) was added to 6b (4.75g,25.51mmol) to dissolve and add ammonium acetate (19.66g,255.09mmol), stir at 60 ℃ for 1h then add sodium cyanoborohydride (4.81g,76.53mmol), and keep stirring at 60 ℃ for 3 h. TLC monitoring indicated the formation of product. The reaction was dried by spinning to give crude product, which was separated by silica gel column chromatography (DCM: MeOH: 10:1, Rf: 0.22) to give product 6 c.
1H NMR(400MHz,DMSO-d 6)δ8.50(d,J=2.4Hz,1H),7.87(d,J=8.8Hz,2H),7.75(d,J=1.6Hz,1H),7.56(d,J=8.8Hz,2H),6.49-6.62(m,1H),4.36(q,J=6.8Hz,1H),1,45(d,J=6.8Hz,3H).
Step four
Compound 1h (72.99mg, 267.04. mu. mol) was dissolved in DCM (1mL) at 0 ℃ and DIEA (103.54mg, 801.11. mu. mol) was added to it, to which was added compound 6c, DIEA (103.54mg, 801.11. mu. mol, 139.54. mu.L), triphosgene (39.62mg, 133.52. mu. mol), and the mixture stirred for 10 min. Stirring at 0 deg.C for 10 min. LCMS indicated product formation. And (4) spin-drying the reaction liquid to obtain a crude product. The crude product was separated using a preparative column (column: XTTIMATE C18150X 25mm X5 μm; mobile phase: water (0.075% trifluoroacetic acid) -acetonitrile; B%: 15% -35%, 8min) to give 6 d.
LCMS(ESI)m/z:487.2[M+1] +
Step five
Subjecting compound 6d (28mg,57.55 μmol) to chiral resolution under SFC resolution conditions of column DAICEL CHIRALPAK AD (250 mm. times.30 mm,10 μm); mobile phase of [ 0.1% ammonia EtOH ]; b%: 55% -55% ]
Peak 1 was collected (retention time: 2.111min), giving example 6.
LCMS(ESI)m/z:487.2[M+1] +
1H NMR(400MHz,CDCl 3-d 6)δ7.88(d,J=2.4Hz,1H),7.69(s,1H),7.63(d,J=8.4Hz,2H),7.42(d,J=8.8Hz,2H),6.43(d,J=1.6Hz,1H),5.95-6.13(m.2H),5.01-5.10(m,1H),4.85(s,1H),3.74-3.8(m,4H),3.43-3.45(m,4H),2.27(s,3H),2.22(s,4H),1.51(d,J=7.2Hz,3H).
Peak 2 was collected (retention time: 3.756min) to give example 7.
LCMS(ESI)m/z:487.2[M+1] +
1H NMR(400MHz,CDCl 3-d 6)δ7.89(d,J=2.4Hz,1H),7.71(d,J=1.2Hz,1H),7.64(d,J=8.4Hz,2H),7.43(d,J=8.4Hz,2H),6.45(t,J=2Hz,1H),6.11(s,1H),6.01(s,1H),5.03-5.11(m,1H),4.76(d,J=7.2Hz,1H),3.77-3.86(m,4H),3.43-3.48(m,4H),2.29(s,3H),2.23(s,3H),1.53(d,J=7.2Hz,3H).
Examples 8 and 9
Figure PCTCN2019101121-APPB-000068
Step one
Compound 8a (500mg,3.03mmol) and ammonium acetate (2.33g,30.27mmol) were added to methanol (7mL), the reaction was stirred for 1 hour at 65 deg.C, then sodium cyanoborohydride (570.64mg,9.08mmol) was added portionwise to the reaction, and the reaction was stirred for an additional 12 hours at 65 deg.C. The reaction solution was directly spin dried at 40-50 ℃ under reduced pressure to give crude product, which was purified by thin layer chromatography plate (PE/EtOAc ═ 1/1) to give 8 b.
1H NMR(400MHz,CDCl 3)δppm 8.19(d,J=2.51Hz,1H),7.79(dd,J=8.78,2.51Hz,1H),6.80(s,1H),4.25(q,J=7.03Hz,2H),3.48(s,3H),1.37(t,J=7.03Hz,3H).
Step two
Compound 1h (100mg, 365.85. mu. mol) and DIEA (141.85mg,1.10mmol, 191.17. mu.L) were added to a reaction solution containing DCM (2mL) and stirred for 10 min. Adding NN-diisopropylethylamine (141.85mg,1.10mmol,191.17 mu L), 8B (60.81mg,365.85 mu mol) and triphosgene (54.28mg,182.92 mu mol) into a stirred DCM (2mL) solution at 0-5 ℃, stirring for 20min at 0-5 ℃, directly concentrating the reaction solution under reduced pressure to obtain a crude product, and purifying by using a preparative chromatographic column (chromatographic column: XYTIMATE C18150X 25mm X5 mu m; mobile phase of [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 17% -27%, 9min) to obtain 8C.
LCMS(ESI)m/z 466.3[M+1] +
Step three
Compound 8c (28mg, 60.14. mu. mol) was resolved by SFC (column: DAICEL CHIRALPAK IC (250 mm. times.30 mm,10 μm); mobile phase: [ 0.1% aqueous ammonia EtOH ]; B%: 50%).
Peak 1 (retention time 5.211min) was collected to give example 8.
LCMS(ESI)m/z:466.2[M+1] +
1H NMR(400MHz,MeOH-d 4)δppm 8.09(d,J=2.51Hz,1H)7.70(dd,J=8.78,2.51Hz,1H)6.76(d,J=8.53Hz,1H)6.13(s,2H)4.07-4.18(m,1H)3.77-3.80(m,4H)3.49-3.53(m,4H)3.49-3.52(m,1H)2.29(s,3H)2.22(s,3H)1.50(d,J=4.0Hz,3H)1.37-1.40(m,3H).
Peak 2 was collected (retention time 6.498min) to give example 9.
LCMS(ESI)m/z:466.2[M+1] +
1H NMR(400MHz,MeOH-d 4)δppm 8.09(d,J=2.51Hz,1H)7.70(dd,J=8.78,2.51Hz,1H)6.76(d,J=8.53Hz,1H)6.13(s,2H)3.78-3.80(m,4H)3.49-3.52(m,4H)2.29(s,3H)2.22(s,3H)1.50(d,J=4.0Hz,2H)1.48-1.51(m,1H)1.37-1.40(m,3H).
Examples 10 and 11
Figure PCTCN2019101121-APPB-000069
Figure PCTCN2019101121-APPB-000070
Step one
Compound 4d (40mg, 146.87. mu. mol) was dissolved in DCM (1mL), DIEA (56.95mg, 440.61. mu. mol, 76.75. mu.L) was added, and stirring was carried out at 0 ℃ for 10 min. To the above reaction solution was added dropwise a solution of compound 6c (27.50mg, 146.87. mu. mol), DIEA (56.95mg, 440.61. mu. mol), triphosgene (21.79mg, 73.44. mu. mol) in DCM (1mL) obtained after stirring at 0 ℃ for 10 minutes. Stirring at 0 deg.C for 10 min. The reaction solution was spin-dried to obtain a crude product, which was purified by preparative chromatography (column: Xtimate C18150X 25mm X5 μm; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 32% -46%, 7min) to obtain product 10 a.
LCMS(ESI)m/z:486.4[M+1] +
Step two
Compound 10a was subjected to chiral resolution. The SFC resolution conditions are that a chromatographic column is YMC CHIRAL Amylose-C (250mm multiplied by 30mm,10 mu m; mobile phase: 0.1% ammonia isopropanol; B%: 50% -50% for min.
Peak 1 was collected (retention time 1.642min) yielding example 10.
LCMS(ESI)m/z:486.4[M+1] +
1H NMR(400MHz,CDCl 3)δ7.90(d,J=2.4Hz,1H),7.71(d,J=1.6Hz,1H),7.66(d,J=8.8Hz,2H),7.43(d,J=8.8Hz,2H),6.52(s,1H),6.45(t,J=1.6Hz,1H),6.18(s,1H),5.95(s,1H),5.83(s,1H),5.02-5.16(m,1H),4.70(d,J=6.8Hz,1H),3.47-3.54(m,8H),2.28(s,3H),2.21(s,3H),1.36-1.45(m,3H).
Peak 2 was collected (retention time 2.516min) to give example 11.
LCMS(ESI)m/z:486.4[M+1] +
1H NMR(400MHz,CDCl 3)δ7.88(d,J=2.8Hz,1H),7.70(d,J=1.2Hz,1H),7.63(d,J=8.4Hz,2H),7.42(d,J=8.8Hz,2H),6.75(s,1H),6.43-6.46(m,1H),6.11(s,1H),5.94(s,1H),5.82(s,1H),5.02-5.14(m,1H),3.49-3.52(m,8H),2.26(s,4H),2.20(s,3H),1.36-1.38(m,3H).
Example 12
Figure PCTCN2019101121-APPB-000071
Step one
Compound 12a (200mg,1.23mmol) and 3-methyl-3-pyrazolin-5-one (144.44mg,1.47mmol) were added to DMSO (2mL), DIEA (237.86mg,1.84mmol, 320.57. mu.L) was added dropwise to the reaction solution, and the reaction was stirred at 60 ℃ for 16 hr. And cooling the reaction liquid to 20-30 ℃, then dropwise adding the reaction liquid into 20mL of ice water, precipitating a light yellow solid, and carrying out vacuum filtration to obtain a compound 12 b.
LCMS(ESI)m/z 224.6[M+1] +
Step two
Compound 12b (100mg, 445.14. mu. mol) and N-BOC piperazine (165.82mg, 890.29. mu. mol) were added to NMP (2mL), DIEA (230.13mg,1.78mmol) was added dropwise to the reaction solution, and the reaction was stirred at 100 ℃ for 1 hr. The reaction solution is cooled to room temperature, then 10mL of water is added, then ethyl acetate (10mL multiplied by 2) is used for extraction, organic phases are combined, the organic phases are dried by anhydrous sodium sulfate and are dried by decompression and spin-drying at 40-50 ℃ to obtain a crude product, and the crude product is purified by column chromatography (PE/EA is 3/1-1/1) to obtain the compound 12 c.
LCMS(ESI)m/z 375.2[M+1] +
Step three
Compound 12c (45mg, 120.18. mu. mol) was added to DCM (4mL), and TFA (2.74g,24.04mmol,1.78mL) was added to the reaction mixture, followed by reaction at 20-30 ℃ for 20 min. And (3) directly carrying out reduced pressure spin drying on the reaction liquid at 40-50 ℃ to obtain the TFA salt of the compound 12 d.
LCMS(ESI)m/z 274.8[M+1]+
Step four
The TFA salt of Compound 12d (50mg, 128.75. mu. mol) was added to DCM (5mL), DIEA (49.92mg, 386.26. mu. mol, 67.28. mu.L) was then added to the reaction mixture, the reaction temperature was lowered to 0 ℃ to determine reaction mixture 1, 1e (23.90mg, 115.88. mu. mol) was added to DCM (5mL), triphosgene (19.10mg, 64.38. mu. mol) was added to the reaction mixture, DIEA (49.92mg, 386.26. mu. mol, 67.28. mu.L) was added dropwise to the reaction mixture, the reaction temperature was lowered to 0 ℃ to determine reaction mixture 2, and reaction mixtures 1 and 2 were each stirred at 0 ℃ for 10min, reaction mixture 1 was slowly added dropwise to reaction mixture 2 at 0 ℃ and the reaction mixture was stirred at 0 ℃ for 10 min. 10mL of water is added into the reaction solution, then DCM (5mL multiplied by 2) is used for extraction, organic phases are combined, the organic phases are dried by anhydrous sodium sulfate and are dried by decompression and spin-drying at 40-45 ℃ to obtain a crude product, and the crude product is purified by a thin-layer chromatography plate to obtain the example 12.
LCMS(ESI)m/z 507.3[M+1] +
1H NMR(400MHz,DMSO-d 6)δ12.19(s,1H)8.58-8.76(m,1H)8.41(d,J=2.01Hz,1H)7.93-7.97(m,1H)7.91(d,J=4.27Hz,1H)7.85-7.89(m,1H)6.99(d,J=7.53Hz,1H)6.01(s,1H)5.83(s,1H)4.92(quin,J=7.03Hz,1H)3.59-3.67(m,4H)3.37-3.42(m,4H)2.23(s,6H)1.43(d,J=7.03Hz,3H).
Example 13
Figure PCTCN2019101121-APPB-000072
Step one
Compound 13a TFA salt (0.06g,154.90 μmol) was dissolved in anhydrous methanol (1mL), sodium borohydride (121.25mg,3.21mmol) was added at 0 deg.C, and stirred at 25 deg.C for 2 hr. The reaction mixture was added to water (10mL), extracted with ethyl acetate (10 mL. times.3), the organic phases combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by plate chromatography (petroleum ether: ethyl acetate 1:1) to give example 13b as a white oil.
LCMS(ESI)m/z:190.1[M+1] +
Step two
Compound 13b (0.06g, 154.90. mu. mol, TFA salt) and DIEA (60.06mg, 464.70. mu. mol, 80.94. mu.L) were added to dried dichloromethane (1mL), stirred for 10mins to form solution 1, and triphosgene (137.90mg, 464.70. mu. mol) was added to a solution of compound 1h and DIEA (60.06mg, 464.70. mu. mol, 80.94. mu.L, 3eq) in dichloromethane (1mL) at 0 ℃ to react for 10mins at 0 ℃, and then solution 1 was added dropwise to the reaction solution to react for 20mins at 0 ℃. Water (2mL) was added to the reaction mixture to quench the reaction and concentrated to give the crude product. The crude product was purified by preparative chromatography (column: Boston Green ODS 150X 30mm5 μm; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 29% -45%, 9min) to give the trifluoroacetate salt of example 13, which was added to a sodium bicarbonate solution, extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and concentrated to give example 13.
LCMS(ESI)m/z:475.2[M+1] +
1H NMR(400MHz,MeOH-d 4)δ=8.61(d,J=2.4Hz,1H),8.49(s,1H),8.07-7.99(m,1H),7.99-7.91(m,1H),7.78(s,1H),6.56(br s,1H),6.44-6.14(m,1H),5.93(q,J=6.6Hz,1H),3.96-3.49(m,9H),2.40(s,3H),2.34(s,3H),1.67(d,J=6.4Hz,3H).
Example 14
Figure PCTCN2019101121-APPB-000073
Step one
2-bromo-5-hydroxypyridine (1g,5.75mmol) was dissolved in acetone (10mL), methyl chloroacetate (748.46mg,6.90mmol,608.50uL) and potassium carbonate (794.31mg,5.75mmol) were added, and the mixture was stirred at 60 ℃ for 16 hours. Filtering to remove insoluble solid, and spin-drying the filtrate to obtain crude product. The crude product was purified by flash column chromatography on silica gel (PE: EA ═ 3:1) to give compound 14a (1.1g,4.47 mmol).
LCMS(ESI)m/z:245.4[M+1] +,247.4[M+3] +
Step two
Compound 14a (50mg,203.20umol) was dissolved in toluene (1mL), and 4-fluoro-1-hydro-pyrazole (14.57mg,169.34umol), cuprous iodide (3.23mg,16.93umol), potassium carbonate (46.81mg,338.67umol), N1, N2-dimethylcyclohexanediamine (4.82mg,33.87umol) were added and stirred at 110 ℃ for 16 hours. Insoluble substances are removed by filtration, and the filtrate is dried by spinning to obtain a crude product. The crude product was purified by preparative chromatography (PE: EA ═ 1:1) to afford compound 14 b.
1H NMR(400MHz,CDCl 3)δppm8.31(d,J=4.4Hz,1H),8.07(d,J=2.8Hz,1H),7.88(d,J=8.8Hz,1H),7.55(d,J=4.4Hz,1H),7.37(dd,J=8.8,2.8Hz,1H),4.71(s,2H),3.83(s,3H).
Step three
Compound 14b (10mg,39.81umol) was dissolved in a solution of tetrahydrofuran (0.3 mL)/water (0.1mL), and lithium hydroxide monohydrate (5.01mg,119.42umol) was added thereto, followed by stirring at 16 ℃ for 5 hours. 4M hydrochloric acid was added to adjust pH to 2-3, extraction was performed with ethyl acetate (2X 10mL), and the solvent was spin-dried to give crude compound 14 c.
LCMS(ESI)m/z:237.8[M+1] +
Step four
Compound 14c (10mg,42.16umol) was dissolved in N, N dimethylformamide (0.5mL), and compound 1h (16.33mg,42.16umol, TFA), HATU (24.05mg,63.24umol), N-diisopropylethylamine (16.35mg,126.48umol,22.03uL) was added and stirred at 16 ℃ for 3 hours. The reaction solution was directly spin-dried to obtain a crude product, which was then separated by preparative chromatography (column: Boston Green ODS 150X 30mm5 μm; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 23% -53%, 8min) to obtain the trifluoroacetate of example 14. The trifluoroacetate salt of example 14 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 14.
LCMS(ESI)m/z:493.1[M+1] +
1H NMR(400MHz,METHANOL-d4)δppm 8.43(d,J=4.02Hz,1H),8.19(d,J=3.04Hz,2H),7.89(d,J=9.20Hz,1H),7.66(d,J=4.02Hz,1H),7.62(d,J=3.04Hz,1H),7.60(d,J=2.76Hz,1H),5.06(s,2H),3.99(s,2H),3.91(s,2H),3.83(s,4H),2.42(s,3H),34(s,3H).
Examples 15 and 16
Figure PCTCN2019101121-APPB-000074
Figure PCTCN2019101121-APPB-000075
Step one
Compound 1c (6.5g,31.68mmol) was added to methanol (50mL), sodium borohydride (3.60g,95.03mmol) was added at 0 deg.C, and stirred at 0 deg.C for 1 hr. The reaction mixture was added with water (100mL), extracted with ethyl acetate (100mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give compound 15 a.
1H NMR(400MHz,DMSO-d6)δ8.68(d,J=4.5Hz,1H),8.42(d,J=2.3Hz,1H),7.98-7.95(m,1H),7.91(d,J=4.5Hz,1H),7.89-7.84(m,1H),5.41(d,J=4.5Hz,1H),4.90-4.79(m,1H),1.38(d,J=6.5Hz,3H).
Step two
15a (6.5g) was separated by SFC (column: Phenomenex-Amylose-1(250 mm. times.30 mm,5 μm); mobile phase: [ 0.1% aqueous ammonia ethanol ]; B%: 25% -25%, min). 15b1 (retention time: 1.882min) and 15b2 (retention time: 2.010min) were obtained.
Step three
15b1(882.61mg,4.26mmol), N, N-disuccinimidyl carbonate (1.49g,5.81mmol) and 4-N, N-dimethylaminopyridine (236.54mg,1.94mmol), triethylamine (391.85mg,3.87mmol,538.99uL) were added to DMF (1mL) and the mixture was stirred at 60 ℃ for 0.5hr to give an intermediate product. A mixed solution of triethylamine (391.85mg,3.87mmol,538.99uL), 4-N, N-dimethylaminopyridine (236.54mg,1.94mmol) and compound 1h (1.5g,3.87mmol, trifluoroacetate) in DMF (1mL) was added at 25 ℃ and reacted for 0.5hr at 25 ℃. The reaction was directly sent to a preparative column (column: Welch Xitinate C18150 x 25mm x 5 um; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 24% -44%, 12min) and purified to give the trifluoroacetate salt of example 15. The trifluoroacetate salt of example 15 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 15.
LCMS(ESI)m/z:507.0[M+1] +
1H NMR(400MHz,METHANOL-d4)δ=8.52(d,J=4.4Hz,1H),8.48(s,1H),8.07-7.93(m,2H),7.72(d,J=4.2Hz,1H),6.23(br s,1H),5.92(q,J=6.8Hz,1H),4.01-3.60(m,8H),2.40(s,3H),2.34(s,3H),1.66(d,J=6.8Hz,3H).
The trifluoroacetate salt of example 16 was obtained in the same manner as in 15b 2. The trifluoroacetate salt of example 16 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 16.
LCMS(ESI)m/z:507.2[M+1] +
1H NMR(400MHz,METHANOL-d4)δ=8.53(d,J=4.4Hz,1H),8.48(s,1H),8.07-7.95(m,2H),7.72(d,J=4.2Hz,1H),6.23(br s,1H),5.91(q,J=6.8Hz,1H),3.98-3.65(m,8H),2.40(s,3H),2.34(s,3H),1.66(d,J=6.8Hz,3H).
Examples 17 and 18
Figure PCTCN2019101121-APPB-000076
Step one
2, 4-dichloro-6-methylpyrimidine (2g,12.27mmol) and 3-cyclopropyl-5-amino-1H-pyrazole (1.66g,13.50 mmol) were dissolved in dimethylsulfoxide (30mL), N-diisopropylethylamine (2.38g,18.40mmol,3.21mL) was added thereto, and the mixture was stirred at 50 ℃ for 4 hours, then the reaction mixture was added to 500mL of water, stirred for 30 minutes, and then a solid was precipitated and filtered to obtain Compound 17 a.
LCMS(ESI)m/z:250.3[M+1] +
1H NMR(400MHz,DMSO-d6)δppm 12.15(s,1H),10.14(s,1H),7.55-6.85(m,1H),6.76(s,1H),6.22-5.49(m,1H),2.67(d,J=1.76Hz,1H),2.26(s,3H),1.97-1.79(m,1H),1.00-0.83(m,2H),0.77-0.60(m,2H)
Step two
Compound 17a (825mg,3.30mmol) and N-Boc piperazine (923.05mg,4.96mmol) were dissolved in dimethyl sulfoxide (15mL), N-diisopropylethylamine (1.28g,9.91mmol,1.73mL) was added, and the mixture was stirred at 140 ℃ for 5 hours. The reaction mixture was poured into 250ml of water, and a solid precipitated, followed by stirring for 30 minutes and filtration to obtain Compound 17 b. .
LCMS(ESI)m/z:400.2[M+1] +
1H NMR(400MHz,DMSO-d6)δppm 11.91(s,1H),9.26(s,1H),6.30-5.92(m,2H),3.72-3.62(m,4H),3.38(s,4H),2.12(s,3H),1.95-1.78(m,1H),1.46-1.42(m,9H),0.99-0.81(m,2H),0.76-0.59(m,2H).
Step three
To compound 17b (1.3g,3.25mmol) was added a mixed solution of trifluoroacetic acid (7.42g,65.08mmol,4.82 mL)/dichloromethane (5mL), and the mixture was stirred at 16 ℃ for 8 hours. The solvent was spun dry to give compound 17c (3g, crude, trifluoroacetate).
Step four
15b1(50mg,241.31umol) was dissolved in N, N dimethylformamide (1mL), N-disuccinimidyl carbonate (84.29mg,329.06umol) was added thereto, and the mixture was stirred at 60 ℃ for 30 minutes, and a solution of compound 17c (181.37mg,219.37umol, trifluoroacetate), triethylamine (22.20mg,219.37umol,30.53uL) and 4-N, N-dimethylaminopyridine (13.40mg,109.69umol) in N, N-dimethylformamide (1mL) was added thereto at 25 ℃ and stirred at 25 ℃ for 30 minutes. LCMS showed product formation. The reaction mixture was directly subjected to preparative column chromatography (column: Boston Green ODS 150: 30 mm. times.5. mu.m; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 25% -55%, 10min) to give the trifluoroacetate salt of example 17. The trifluoroacetate salt of example 17 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 17.
LCMS(ESI)m/z:533.1[M+1] +
1H NMR(400MHz,MeOH-d4)δppm 8.51(d,J=4.60Hz,1H),8.46(d,J=1.52Hz,1H),8.02-7.92(m,2H),7.70(d,J=4.02Hz,1H),6.20(s,1H),5.90(q,J=6.64Hz,1H),3.97-3.50(m,10H),2.38(s,3H),1.97-1.88(m,1H),1.64(d,J=6.64Hz,3H),1.07-0.97(m,2H),0.81-0.70(m,2H).
Step five
Compound 17c (1.00g,1.21mmol, TFA) was dissolved in dichloromethane (1mL) at 0 deg.C, N-diisopropylethylamine (470.04mg,3.64mmol,633.48uL) was added and stirred for 10 minutes to prepare solution 1, while Compound 1e (200mg,969.84umol) was dissolved in dichloromethane (1mL), triphosgene (179.87mg,606.15umol), N-diisopropylethylamine (470.04mg,3.64mmol,633.48uL) was added and stirred for 10 minutes to prepare solution 2, and then solution 2 was added to solution 1 at 0 deg.C and stirred for 10 minutes. The reaction mixture was quenched by adding 0.5ml of water, the reaction mixture was spin-dried, dissolved in methanol, filtered to remove insoluble materials, the filtrate was spin-dried to give a crude product, and the crude product was separated by preparative HPLC (column: YMC-Triart Prep C18150: 40 mm. multidot.7 um; mobile phase: [ water (0.1% trifluoroacetic acid) -acetonitrile ]; B%: 25% -45%, 10min) to give the trifluoroacetate of example 18. The trifluoroacetate salt of example 18 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 18.
LCMS(ESI)m/z:532.3[M+1] +
1H NMR(400MHz,METHANOL-d4)δppm 8.49(d,J=4.52Hz,1H),7.94-7.87(m,2H),8.41(s,1H),7.68(d,J=4.28Hz,1H),6.26(s,1H),5.01(q,J=7.02Hz,1H),3.91-3.81(m,4H),3.69-3.60(m,4H),2.38(s,3H),1.97-1.90(m,1H),1.54(d,J=7.02Hz,3H),1.05-0.96(m,2H),0.77-0.71(m,2H).
Example 19
Figure PCTCN2019101121-APPB-000077
Step one
To a solution of compound 19a (1.9g,8.88mmol), ethyl bromoacetate (14.08g,84.32mmol,9.33mL) in DMF (20mL) was added cesium carbonate (7.23g,22.19mmol), and the mixture was stirred at 110 ℃ for 20 hours. LCMS indicated the starting material had reacted to completion and product was formed. The reaction solution is filtered by diatomite to obtain filtrate, and then the filtrate is decompressed and concentrated by an oil pump to obtain a crude product. The crude product was isolated by flash column chromatography on silica gel (PE: EA ═ 3:1, Rf ═ 0.61) to afford compound 19 b.
LCMS(ESI)m/z:299.7[M+1] +,301.7[M+3] +
Step two
To a solution of compound 19b (2g,6.66mmol) in THF (10mL)/MeOH (10mL)/H2O (10mL) was added lithium hydroxide monohydrate (2.80g,66.63mmol) and stirred at 20 ℃ for 16H. The reaction mixture was concentrated to remove tetrahydrofuran and methanol, water (20mL) was added, extraction was performed with ethyl acetate (20mL × 2) to remove impurities, the pH of the aqueous phase was adjusted to 3 with 4N HCl, extraction was performed with ethyl acetate (20mL × 2), drying was performed with anhydrous sodium sulfate, and concentration was performed under reduced pressure to obtain compound 19 c.
LCMS(ESI)m/z:271.8[M+1] +,273.8[M+3] +
1H NMR(400MHz,DMSO-d6)δ12.68(br s,1H),6.73-6.93(m,2H),6.50(d,J=8.53Hz,1H),4.14-4.25(m,2H),4.06(s,2H),3.37-3.43(m,2H).
Step three
To a solution of compound 19c (0.1g,367.52umol), compound 1h (341.67mg,882.05umol, trifluoroacetate) in DMF (3mL) was added HATU (209.61mg,551.28umol), N, N-diisopropylethylamine (142.50mg,1.10mmol,192.05 uL). Stirred at 20 ℃ for 16 hours. To the reaction mixture was added water (15mL), and ethyl acetate (10mL × 2) was added for extraction, and the organic phases were combined and dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product. The crude product was isolated on a thin layer chromatography plate (PE: EA ═ 0:1, Rf ═ 0.44) to afford compound 19 d.
LCMS(ESI)m/z:527.0[M+1] +,529.0[M+1] +
Step four
To a solution of compound 19d (100mg,189.60umol), 4-fluoro-1 hydro-pyrazole (16.32mg,189.60umol) in DMF (4mL) was added cuprous iodide (3.61mg,18.96umol), cesium carbonate (123.55mg,379.21umol), (1S,2S) -N1, N2-dimethylcyclohexanediamine (5.39mg,37.92 umol). Stirring is carried out for 16 hours at 110 ℃ under the protection of nitrogen. The reaction solution was filtered to collect the filtrate. The filtrate was sent to a preparative chromatography column (column: Gemini 150 x 25mm 5 μm; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 17% -47%, 11.5min) to separate the white solid product, the trifluoroacetate salt of example 19. The trifluoroacetate salt of example 19 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 19.
LCMS(ESI)m/z:533.1[M+1] +
1H NMR(400MHz,DMSO-d6)δ12.32(s,1H),11.01(s,1H),8.46(d,J=4.52Hz,1H),7.68(d,J=4.27Hz,1H),7.06-7.09(m,1H),7.10(s,1H),6.65(d,J=8.78Hz,1H),4.36(s,2H),4.21-4.29(m,2H),3.89(br s,2H),3.79(br s,2H),3.70(br s,2H),3.62-3.66(m,2H),3.42(br d,J=4.27Hz,2H),2.33(s,3H),2.25(s,3H).
Examples 20, 21 and 22
Figure PCTCN2019101121-APPB-000078
Figure PCTCN2019101121-APPB-000079
Step one
Compound 20a (10g,50.19mmol) was dissolved in tetrahydrofuran (50mL), lithium bis (trimethylsilyl) amide (8.40g,50.19mmol,1M) was added dropwise to the reaction mixture at-78 ℃ under stirring for 0.5 hour, and then N-phenylbis (trifluoromethanesulfonyl) imide (17.93g,50.19mmol) was dissolved in THF (50mL) and added dropwise to the reaction mixture. The reaction solution was warmed to 0 ℃ and stirred for reaction for 3 hours. And adding 3mL of saturated ammonium chloride aqueous solution into the reaction solution to quench the reaction, then directly adding anhydrous sodium sulfate, drying, filtering, collecting the filtrate, and evaporating to dryness by rotary evaporation to obtain a crude product. The crude product was purified by silica gel column (PE/EA ═ 10/1, Rf ═ 0.8) to give a mixture of compounds 20b1 and 20b2 in a molar ratio of about 3: 1.
1H NMR(400MHz,CHLOROFORM-d)δ7.69(s,1H),5.93(s,0.35H),4.05~4.03m,0.7H),3.57~3.43(m,2.7H),2.45~2.42(m,2H),1.96~1.89(m,2.7H),1.49(s,9H),1.47(s,3H).
Step two
A mixture of compound 20b1(6g,18.11mmol), compound 20b2(2g,6.04mmol), bis pinacol boronate (7.36g,28.98mmol), Pd (dppf) Cl2(1.06g,1.45mmol) and potassium acetate (7.11g,72.44mmol) were added together to dioxane (160mL) and then heated to 80 ℃ by an oil bath under nitrogen and the reaction stirred for 16 h. And filtering the reaction solution, collecting filtrate, and spin-drying to obtain a crude product. The crude product was passed through a flash column (PE/EA: 10/1, Rf 1: 0.76)&Rf2 ═ 0.72) to give a mixture of compounds 20c1 and 20c2 in a molar ratio close to 3: 1.
1H NMR(400MHz,CHLOROFORM-d)δ7.79(m,1H),6.62(s,1H),3.96(s,2H),3.51(s,3H),2.19~2.16(m,3H),1.80~1.72(m,3H),1.45(s,13H),1.32(s,5H),1.23(s,16H).
Step three
A mixture of compound 20c1(1g,3.23mmol), compound 20c2(300.00mg,970.22umol), compound 1f (940.34mg,4.20mmol), Pd (PPh)3) 4(373.72mg,323.41umol) and K2CO 3(1.79g,12.94mmol) were added together to dioxane (12mL) and water (4mL), and the reaction was heated to 125 ℃ by a microwave synthesizer under nitrogen and stirred for 0.5 h. To the reaction solution, 5mL of water and 5mL of ethyl acetate were added for extraction, followed by liquid separation, extraction of the aqueous phase with 5mL of ethyl acetate again, and the organic phases were combined, dried over anhydrous sodium sulfate, and rotary-evaporated to dryness to obtain a crude product, which was purified by flash silica gel column (dichloromethane/methanol-10/1, Rf-0.43) to obtain a mixture of compounds 20d1 and 20d 2.
LCMS(ESI)m/z:371.3[M+1] +
Step four
A mixture of compound 20d1(100mg,269.94umol), compound 20d2(100.00mg,269.94umol), ammonium formate (170.21mg,2.70mmol) and Pd/C (50mg, 10% content) were added together to ethanol (5mL), then heated to 70 ℃ under an oil bath and the reaction stirred for 4 hours. The reaction solution was directly filtered, the filtrate was collected, and the crude product was obtained by rotary evaporation under reduced pressure and purified by preparative chromatography plate (dichloromethane/methanol: 20/1, Rf: 0.4) to obtain compound 20 e.
LCMS(ESI)m/z:373.3[M+1] +
Step five
Compound 20e (60mg,161.09umol) and hydrogen chloride/ethyl acetate (4M,2mL) were added in one portion to EtOAc (2mL) and the reaction was stirred at 20-30 ℃ for 16 h. The reaction solution was directly evaporated to dryness by rotary evaporation. Compound 20f (hydrochloride) was obtained and used directly in the next step.
LCMS(ESI)m/z:273.2[M+1] +
Step six
Compound 20f (25mg,80.96umol) and N, N-diisopropylethylamine (31.39mg,242.88umol) were added to DCM (2mL), followed by compound 1e (16.70mg,80.96umol), N, N-diisopropylethylamine (31.39mg,242.88umol,42.31uL) and triphosgene (12.01mg,40.48umol) being added together and stirred for 10 minutes and then added to the stirred reaction solution at 0 ℃ and the reaction solution stirred for 10 minutes at 25-30 ℃. The reaction mixture was quenched with 1mL of water, extracted with 5mL of water and 5mL of dichloromethane, separated, the aqueous phase extracted with 5mL of dichloromethane, the organic phases combined, dried over anhydrous sodium sulfate, evaporated to dryness to give a crude product, which was purified by preparative separation (column: Boston Green ODS 150 x 305 u; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 33% -53%, 7min) to give the trifluoroacetate salt of example 20. The trifluoroacetate salt of example 20 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 20.
LCMS(ESI)m/z:505.3[M+1] +
1H NMR(400MHz,METHANOL-d4)δ8.50~8.49(m,1H),8.39(d,J=8.4Hz,1H),7.90(d,J=8.4Hz,2H),7.70(t,J=3.6Hz,1H),7.56(s,1H),6.61(s,1H),5.02~4.97(m,1H),4.30~4.27(m,1H),4.05~3.97(m,1H),3.27~3.10(m,2H),2.96~2.87(m,1H),2.48(d,J=6.8Hz,3H),2.33(d,J=4Hz,3H),2.17(s,1H),2.04~1.91(m,2H),1.63(m,1H),1.54(d,J=6.8Hz,3H).
Step seven
The trifluoroacetate salt from example 20(60mg,118.92umol) was subjected to SFC separation (column: DAICEL CHIRALPAK AS-H (250mm 30mm,5 um); mobile phase: [ 0.1% aqueous ammonia ethanol ]; B%: 50% -50%, min). Example 21 (retention time: 3.391min) and example 22 retention times were finally obtained: 4.092min)
Example 21:
LCMS(ESI)m/z:505.2[M+1] +
1H NMR(400MHz,METHANOL-d4)δ8.48(d,J=4.4Hz,1H),8.38(s,1H),7.87~7.85(m,2H),7.68(d,J=4Hz,1H),6.74(s,1H),6.19(s,1H),5.03~4.98(m,1H),4.23(d,J=13.2Hz,1H),4.11(d,J=13.2Hz,1H),3.25~3.19(m,1H),2.88~2.81(m,2H),2.33(s,3H),2.29(s,3H),2.12(s,1H),1.89~1.81(m,2H),1.61~1.60(m,1H),1.52(d,J=7.2Hz,3H)
example 22:
LCMS(ESI)m/z:505.2[M+1] +
1H NMR(400MHz,METHANOL-d4)δ8.36(d,J=4Hz,1H),8.28(d,J=1.6Hz,1H),7.81~7.75(m,2H),7.84(s,1H),7.55(d,J=4Hz,1H),6.62(s,1H),6.08(s,1H),4.90~4.85(m,1H),4.11(d,J=13.2Hz,1H),4.00(d,J=13.2Hz,1H),3.11~3.05(m,1H),2.74~2.70(m,2H),2.22(s,3H),2.17(s,3H),1.99(s,1H),1.77~1.66(m,2H),1.47(m,1H),1.39(d,J=7.2Hz,3H)
example 23
Figure PCTCN2019101121-APPB-000080
Figure PCTCN2019101121-APPB-000081
Step one
Compound 1e (200mg,969.84umol)) And Boc2O (222.25mg,1.02mmol) was added together to THF (4mL) and H2O (4mL), and then the reaction was stirred at 10 ℃ for 16 hours. The reaction solution was directly rotary evaporated to dryness to give crude compound 23a, which was used directly in the next step.
LCMS(ESI)m/z:307.3[M+1] +
Step two
Compound 23a (250mg,816.10umol) was dissolved in THF (5mL), then sodium hydrogen (65.28mg,1.63mmol, 60%) was added to the reaction solution, stirred for ten minutes, iodomethane (231.67mg,1.63mmol) was added to the stirred reaction solution, and finally the reaction solution was stirred at 20 ℃ for 16 hours. Adding 5mL of water into the reaction solution to quench the reaction, then adding 20mL of ethyl acetate for extraction, separating the solution, extracting the aqueous phase with 20mL of ethyl acetate again, combining the organic phases, drying the organic phases with anhydrous sodium sulfate, and carrying out rotary evaporation and evaporation to obtain the compound 23 b.
LCMS(ESI)m/z:320.9[M+1] +
Step three
Compound 23b (200mg,624.30umol) was dissolved in ethyl acetate (5mL), and then hydrogen chloride/ethyl acetate (4M,4.00mL) was added to the reaction solution, followed by stirring at 20 ℃ for 16 hours. The reaction solution was directly filtered, and the filter cake was compound 23 c.
LCMS(ESI)m/z:220.9[M+1] +
Step four
Compound 1h (140mg,453.35umol, HCl) and N, N-diisopropylethylamine (175.77mg,1.36mmol) were added to dichloromethane (5mL), followed by compound 23c (99.85mg,453.35umol), N, N-diisopropylethylamine (175.77mg,1.36mmol,236.89uL) and triphosgene (67.27mg,226.68umol) being added together and stirred for 10 minutes and then added to the stirred reaction solution at 0 ℃ and the reaction solution stirred for 10 minutes at 15-20 ℃. Directly rotary-evaporating the reaction solution to dryness to obtain a crude product. The crude product was sent to preparative separation (column: Boston Green ODS 150 x 30mm x 5 um; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 30% -60%, 9min) to give the trifluoroacetic acid salt of example 23. The trifluoroacetate salt of example 23 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 23.
LCMS(ESI)m/z:519.1[M+1] +
1H NMR(400MHz,METHANOL-d 4)δ8.51(br d,J=3.8Hz,1H),8.44(br s,1H),7.95(br s,2H),7.71(br s,1H),6.37(s,1H),6.24(s,1H),5.83(s,1H),5.33(br d,J=6.8Hz,1H),3.70(br s,4H),3.55(br s,4H),2.79(s,3H),2.39(s,3H),2.34(s,3H),1.67(br d,J=6.8Hz,3H)
Examples 24 and 25
Figure PCTCN2019101121-APPB-000082
Step one
Potassium permanganate (45.60g,288.54mmol) was added to a mixed solution of compound 24a (7g,48.09mmol) in water (60mL) and pyridine (60mL), and the mixture was stirred at 100 ℃ for 3 hours. The reaction solution is directly filtered, washed by 100mL of methanol, and the filtrate is decompressed and concentrated to obtain the compound 24 b.
1H NMR(400MHz,DMSO-d 6)δ8.60(s,1H),8.02(dd,J=1.64,9.16Hz,1H).
Step two
Compound 24b (4g,22.79mmol), N, O-dimethylhydroxylamine hydrochloride (1.39g,22.79mmol) in DMF (100mL) was added to HATU (13.00g,34.18mmol), N, N-diisopropylethylamine (5.89g,45.57mmol,7.94mL) and stirred at 30 ℃ for 2 h. The reaction mixture was added with 100mL of water, and ethyl acetate (100mL × 2) was added for extraction, and the organic phases were combined and dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product. The crude product was isolated (PE: EA ═ 4:3, Rf ═ 0.42) to give compound 24 c.
LCMS(ESI)m/z:218.6[M+1] +
1H NMR(400MHz,CHLOROFORM-d)δ8.56(d,J=1.76Hz,1H),7.83(dd,J=1.88,8.42Hz,1H),3.52-3.57(m,3H),3.30-3.39(m,3H).
Step three
Methylmagnesium bromide (3M,5.49mL) was added dropwise to a solution of compound 24c (3g,13.72mmol) in THF (30mL) at 0 deg.C, and stirred for 2 hours at 25 deg.C. To the reaction mixture was added a saturated ammonium chloride solution (50mL), followed by extraction with ethyl acetate (50mL × 2), and the organic phases were combined and dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product. The crude product was isolated on a flash silica gel column (PE: EA ═ 3:1, Rf ═ 0.43) to afford compound 24 d.
LCMS(ESI)m/z:173.8[M+1] +
1H NMR(400MHz,CHLOROFORM-d)δ8.75(d,J=1.52Hz,1H),7.99(dd,J=1.64,8.16Hz,1H),2.65(s,3H).
Step four
To a solution of compound 24d (1.3g,7.49mmol), 1H-pyrazole (509.88mg,7.49mmol) in DMF (20mL) was added K2CO 3(3.11g,22.47mmol), and stirred at 100 ℃ for 16 hours. The reaction solution is filtered by diatomite, and the filtrate is decompressed and concentrated by an oil pump to obtain a crude product. The crude product was isolated on preparative chromatography plates (PE: EA ═ 3:1, Rf ═ 0.38) to give compound 24 e.
LCMS(ESI)m/z:205.8[M+1] +
1H NMR(400MHz,CHLOROFORM-d)δ8.84(s,1H),8.46(d,J=2.76Hz,1H),8.15(dd,J=1.76,11.54Hz,1H),7.89(d,J=1.00Hz,1H),6.52-6.61(m,1H),2.66(s,3H).
Step five
To a solution of compound 24e (0.60g,2.92mmol) in methanol (10mL) was added ammonium acetate (2.25g,29.24mmol), and after stirring at 65 ℃ for 1 hour, NaBH was added3CN (551.26mg,8.77mmol, stirring at 65 ℃ for 3 hours, directly concentrating the reaction solution to obtain a crude product, separating the crude product by preparative chromatography plate (DCM: MeOH: 10:1, Rf: 0.20) to obtain a product 24f, and further separating by SFC (column: DAICEL CHIRALPAK AD-H (250mm 30mm,5 μm); mobile phase: [ 0.1% ammonia water methanol: ] to obtain a crude product](ii) a 35% -35% of B%, min) to obtain 24g of 1 (retention time: 4.177min) and 24g2 (retention time: 4.980 min).
24g1:
LCMS(ESI)m/z:206.8[M+1] +
1H NMR(400MHz,CHLOROFORM-d)δ8.28(s,2H),7.83(d,J=1.00Hz,1H),7.70(dd,J=1.63,11.92Hz, 1H),6.46-6.55(m,1H),4.28(q,J=6.70Hz,1H),1.43(d,J=6.54Hz,3H).
24g2:
LCMS(ESI)m/z:206.8[M+1] +
1H NMR(400MHz,CHLOROFORM-d)δ8.28(s,2H),7.82(d,J=1.26Hz,1H),7.69(dd,J=1.64,11.92Hz,1H),6.47-6.51(m,1H),4.27(q,J=6.62Hz,1H),1.42(d,J=6.54Hz,3H).
Step six
To a solution of compound 24g1(59.63mg,289.14umol) at 0 ℃ in DCM (2mL) was added N, N-diisopropylamine (140.13mg,1.08mmol,188.86uL), triphosgene (53.63mg,180.71umol), and stirred at 0 ℃ for 10 minutes, and the reaction solution was added dropwise to a single vial containing compound 1h (140mg,361.42umol, trifluoroacetate), N, N-diisopropylamine (140.13mg,1.08mmol,188.86uL), and DCM (2mL), and stirring was continued at 0 ℃ for 10 minutes. 3mL of water was added to the reaction mixture, which was extracted with dichloromethane (3 mL. times.3), and the combined organic phases were concentrated under reduced pressure to give a crude product. The crude product was sent to preparative chromatography (column: Boston Green ODS 150 x 30mm5 μm; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 21% -41%, 7min) to give the trifluoroacetate salt of example 24. The trifluoroacetate salt of example 24 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 24.
LCMS(ESI)m/z:506.2[M+1] +
1H NMR(400MHz,DMSO-d 6) δ 12.39(br s,1H),11.72(br s,1H),11.05(br s,1H),8.31 to 8.43(m,1H),7.90 to 7.99(m,1H),7.80 to 7.87(m,1H),7.09(d, J ═ 7.54Hz,1H),6.58(t, J ═ 2.02Hz,1H),6.09 to 6.45(m,2H),4.98(q, J ═ 7.10Hz,1H),3.78(br s,4H),3.54(br s,4H),2.33(s,3H),2.25(s,3H),1.46(d, J ═ 7.03Hz,3H), the trifluoroacetate of example 25 was prepared in the same manner as compound 24g 2. The trifluoroacetate salt of example 25 is added to sodium bicarbonate solution, extracted with ethyl acetate and the organic phase is dried over anhydrous sodium sulfate and concentrated under reduced pressure to give example 25.
LCMS(ESI)m/z:506.2[M+1] +
1H NMR(400MHz,DMSO-d 6)δ12.09-12.66(m,1H),11.45-12.01(m,1H),11.04(br s,1H),8.30-8.43(m,1H),7.95(dd,J=1.52,11.80Hz,1H),7.83(d,J=1.52Hz,1H),7.08(d,J=7.54Hz,1H),6.58(t,J=2.14Hz,1H),6.37(br s,1H),6.18(br s,1H),4.98(q,J=7.04Hz,1H),3.78(br s,5H),3.53(br s,3H),2.32(s,3H),2.24(s,3H),1.46(d,J=7.04Hz,3H).
Examples 26 and 27
Figure PCTCN2019101121-APPB-000083
Figure PCTCN2019101121-APPB-000084
Step one
Compound 1f (3.56g,15.90mmol) was dissolved in 1-methylpyrrolidone (15mL), ethyl 3-carboxylate piperidine (5g,31.80mmol), N-diisopropylethylamine (6.17g,47.71mmol,8.31mL) were added, and stirring was carried out at 90 ℃ for 16 hours. The reaction mixture was extracted with ethyl acetate (40mL × 3) and the organic phases were combined, washed with water (100mL × 5), saturated sodium chloride solution (100mL × 1), dried over anhydrous sodium sulfate, filtered and the solution was spin-dried to give the crude product. The crude product was purified by flash silica gel column (PE: EA ═ 1:1) to afford compound 26 a.
LCMS(ESI)m/z:345.2[M+1] +
1H NMR(400MHz,DMSO-d6)δppm 9.23(s,1H),11.85(s,1H),6.15(s,1H),4.67(d,J=10.79Hz,1H),4.43(d,J=12.80Hz,1H),4.13-4.08(m,2H),3.17(d,J=5.26Hz,1H),3.06(t,J=11.54Hz,1H),2.99-2.89(m,1H),2.47-2.41(m,1H),2.19(s,3H),2.11(s,3H),2.03–1.96(m,1H),1.74-1.57(m,2H),1.45-1.36(m,1H),1.19(t,J=7.16Hz,4H).
Step two
Compound 26a (100mg,290.35umol) was dissolved in a tetrahydrofuran (1 mL)/water (0.3mL) solution, and lithium hydroxide monohydrate (36.55mg,871.05umol) was added thereto, followed by stirring at 16 ℃ for 2 hours. The reaction solution was adjusted to pH 2-3 with 4N hydrochloric acid, extracted with ethyl acetate (10mL × 2), and the solvent was dried to give compound 26 b. .
LCMS(ESI)m/z:316.9[M+1] +
Step three
Compound 26b (90mg,284.49umol) was dissolved in N, N dimethylformamide (1mL), and compound 1e (58.67mg,284.49umol), HATU (162.26mg,426.73umol), N-diisopropylethylamine (110.30mg,853.46umol,148.66uL) was added thereto, followed by stirring at 16 ℃ for 3 hours, and the solvent was spin-dried to obtain the crude product. The crude product was separated and purified by preparative chromatography (column: Xtimate C18150 × 25mm × 5 um; mobile phase: [ water (0.1% trifluoroacetic acid) -acetonitrile ]; B%: 30% -40%, 11min) and the product was resolved by SFC (column: DAICEL CHIRALCEL OJ-H (250mm × 30mm,5 um); mobile phase: [ 0.1% aqueous ammonia ethanol ]; B%: 30% -30%, min) to give example 26 (retention time: 2.947min), example 27 (retention time: 3.504 min).
Example 26:
LCMS(ESI)m/z:505.7[M+1] +
1H NMR(400MHz,METHANOL-d4)δppm 8.44(dd,J=4.52,0.80Hz,1H),8.30(d,J=2.00Hz,1H),7.84-7.79(m,1H),7.78-7.73(m,1H),7.66(d,J=4.28Hz,1H),6.11(s,1H),6.02(s,1H),5.07(q,J=6.80Hz,1H),4.29(dd,J=13.60,3.20Hz,1H),4.21-4.11(m,1H),3.57-3.51(m,2H),3.40-3.33(m,1H),2.58-2.45(m,1H),2.17(d,J=9.06Hz,6H),2.00–1.89(m,2H),1.71-1.63(m,1H),1.45(d,J=7.04Hz,3H).
example 27:
LCMS(ESI)m/z:505.6[M+1] +
1H NMR(400MHz,METHANOL-d4)δppm 8.46(d,J=4.02Hz,1H),8.35(s,1H),7.94-781(m,2H),7.67(d,J=4.28Hz,1H),5.05(q,J=7.04Hz,1H),4.38(d,J=12.00Hz,1H),4.21(d,J=13.80Hz,1H)3.54-3.49(m,2H),3.35(s,1H),2.65-2.54(m,1H),2.35-2.27(m,6H),2.08-1.98(m,1H),1.87-1.78(m,2H),1.70-1.58(m,1H),1.50(d,J=7.20Hz,3H).
examples 28 and 29
Figure PCTCN2019101121-APPB-000085
Step one
Compound 15b1(335.49mg,1.62mmol), N, N-disuccinimidyl carbonate (622.15mg,2.43mmol) and 4-N, N-dimethylaminopyridine (98.90mg,809.56umol) together with triethylamine (163.84mg,1.62mmol,225.36uL) were added to DMF (5mL), and then the reaction was stirred at 60 ℃ for 1 hour, then a solution of compound 20f (500mg,1.62mmol, HCl) and triethylamine (163.84mg,1.62mmol) in DMF (5mL) was added to the reaction solution at 25 ℃ and finally the reaction solution was stirred at 25 ℃ for 0.5 hour. The reaction solution is directly concentrated under reduced pressure to obtain a crude product. Purifying with preparative chromatographic column (chromatographic column: Phenomenex Gemini-NX 150: 30 mM: 5 um; mobile phase: [ water (0.04% ammonia water +10mM ammonium bicarbonate) -acetonitrile ]; B%: 39% -69%, 8 min). And then sent to preparative chromatography column (column: Boston Green ODS 150: 30 mm: 5 um; mobile phase: [ water (0.075% trifluoroacetic acid) -acetonitrile ]; B%: 25% -55%, 7min) to obtain Compound 28 a.
LCMS(ESI)m/z:506.2[M+1] +
Step two
28a (130mg,257.15umol) was sent to SFC for separation. SFC conditions were (column: DAICEL CHIRALPAK AD (250 mm. about.30 mm,10um), mobile phase: [ 0.1% NH3H2O, IPA ]; B%: 60% -60%, min). Finally, example 28 (retention time: 1.499min) and example 29 (retention time: 2.252min) were obtained.
Example 28
LCMS(ESI)m/z:506.1[M+1] +
1H NMR(400MHz,METHANOL-d4)δ8.46-8.21(m,2H),7.90-7.71(m,2H),7.57(br s,1H),6.73-5.99(m,1H),5.73(q,J=6.5Hz,1H),5.19-5.00(m,1H),4.24-4.02(m,2H),2.90-2.55(m,2H),2.21(br s,3H),2.16(s,3H),1.99(br d,J=11.8Hz,1H),1.75(br s,2H),1.55-1.37(m,5H).
Example 29
LCMS(ESI)m/z:506.6[M+1] +
1H NMR(400MHz,METHANOL-d4)δ=8.49(d,J=4.4Hz,1H),8.43(br s,1H),7.97-7.89(m,2H),7.68(d,J=4.3Hz,1H),6.78-6.16(m,1H),5.85(br s,1H),5.32-5.19(m,1H),4.36-4.29(m,2H),3.04-2.72(m,2H),2.30-2.25(m,1H),2.28(s,2H),2.05-2.03(m,4H),1.84(br d,J=11.0Hz,2H),1.61-1.53(m,5H).
Biological test data:
experimental example 1: in vitro enzymatic Activity assay for Compounds of the invention
Purpose of experiment
By Z' -LYTETMKinase assay to detect enzyme activity as IC of compound50The values are used as indicators to evaluate the inhibitory effect of compounds on RET and RET (V804M) kinase.
Experimental methods
Testing of RET and RET (V804M) kinase compounds were diluted 3-fold at 10 concentrations from 3 μ M to 0.152 nM; the content of DMSO in the assay reaction was 1%.
Reagent:
basic reaction buffer, 20mM hydroxyethylpiperazine ethanethiosulfonic acid (Hepes) (pH 7.5) buffer, 10mM MgCl2Ethylene glycol bisaminoethylether tetraacetic acid (EGTA) at 1mM, polyoxyethylene dodecanol ether (Brij35) at 0.02% by weight, bovine serum albumin () at 0.02mg/ml, Na at 0.1mM3VO 42mM Dithiothreitol (DTT), 1% DMSO.
A compound:
the test compound was dissolved in 100% DMSO and diluted to 10mM for use. Solution dilution was performed using an Integra Viaflo Assist. Reaction process of general enzyme:
and (3) testing conditions are as follows: the concentration of RET enzyme is 3 mu M, the concentration of peptide substrate CHKtide is 1000 mu M, and the concentration of ATP is 20 mu M; the concentration of RET (V804M) enzyme was 80. mu.M, the concentration of substrate peptide was 1000. mu.M, and the concentration of ATP was 30. mu.M.
The reaction process is as follows: kinase/polypeptide solutions were prepared according to the test conditions. The compound solutions of different concentrations were added and incubated for 20 minutes at room temperature, the corresponding concentration of 33P-ATP was added and the reaction was incubated for 120 minutes at room temperature. Radioactivity was detected by the filter-binding method.
Reaction detection:
the reaction was stopped by adding 0.5% phosphoric acid to the reaction mixture, and the Envision apparatus read the plate.
Data analysis
Converting the data into phosphorylation rate and inhibition rate, and obtaining compound IC by parameter curve fitting (GraphPad Software)50And (4) data.
The results are shown in Table 1:
table 1 example 1 kinase activity IC50Test results
RET enzyme IC 50 (nM) RET V804M IC 50(nM)
Hydrochloride salt of example 1 1.1 7.86
Example 2 293 609
Example 3 62.1 204
Trifluoroacetic acid salt of example 4 0.5 2.58
Example 6 60.7 69.6
Example 7 4.2 11.7
Example 8 50.4 134
Example 9 201 250
Example 10 23.9 31.7
Example 11 4.76 11.6
Example 12 68.2 83.3
Trifluoroacetic acid salt of example 13 6.73 9.16
Trifluoroacetic acid salt of example 14 4.57 8.24
Trifluoroacetic acid salt of example 15 4.00 9.00
Trifluoroacetic acid salt of example 16 7.3 11.0
Trifluoroacetic acid salt of example 17 4.3 2.74
Trifluoroacetic acid salt of example 18 1.06 2.36
Trifluoroacetic acid salt of example 19 44.9 35.6
Trifluoroacetic acid salt of example 20 4.31 4.19
Example 21 96.4 91.4
Example 22 0.9 1.62
Trifluoroacetic acid salt of example 23 1.35 12.8
Trifluoroacetic acid salt of example 24 22.6 24.7
Trifluoroacetic acid salt of example 25 5.67 14.4
Example 26 N/A N/A
Example 27 N/A N/A
Example 28 231 131
Example 29 2.40 4.77
N/A represents not detected
And (4) experimental conclusion: the compound has excellent inhibitor activity on RET and mutant RET V804M thereof, and has excellent treatment effect on patients with RET abnormal tumors.

Claims (19)

  1. A compound represented by the formula (III), an isomer thereof or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2019101121-APPB-100001
    wherein the content of the first and second substances,
    a ring is selected from
    Figure PCTCN2019101121-APPB-100002
    The above-mentioned
    Figure PCTCN2019101121-APPB-100003
    Optionally substituted by 1,2 or 3RgSubstitution;
    R 1selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C3-6Cycloalkyl radical, said C1-3Alkyl radical, C1-3Alkoxy and C3-6Cycloalkyl is optionally substituted by 1,2 or 3RaSubstitution;
    R 2selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl and C1-3Alkoxy radical, said C1-3Alkyl and C1-3Alkoxy is optionally substituted by 1,2 or 3RbSubstitution;
    R 4selected from H, F, Cl, Br, I, OH, NH2、CN、C 1-5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and a 5-6 membered heteroaromatic ring, said C1- 5Alkyl radical, C1-5Alkoxy radical, C1-5Alkylamino and 5-6 membered heteroaromatic rings optionally substituted with 1,2 or 3RdSubstitution;
    R aand RbEach independently selected from F, Cl, Br, I, OH and NH2
    R dSelected from H, F, Cl, Br, I, OH, NH2、CN、C 1-3Alkyl radical, C1-3Alkoxy and C1-4An alkylamino group;
    T 1、T 2、T 3and T5Each independently selected from CH and N;
    T 4selected from the group consisting of CR5And N;
    L 2selected from NH and O;
    L 3is selected from-CH (R)6)-、-CH(R 6)CH 2-、-CHCH 2CH 2-、-CH(R 6)-O-、-CHCH 2-O-、-CH 2-N(R 6)-、-CH 2CH 2-N(R 6)-;
    R 5Is selected from H;
    R 6is selected from H and CH3
    Or R5、R 6Are connected together to form a structural unit
    Figure PCTCN2019101121-APPB-100004
    Is selected from
    Figure PCTCN2019101121-APPB-100005
    R gSelected from H, F, Cl, Br, I, OH, NH2And CN;
    the 5-to 6-membered heteroaryl group comprises 1,2,3 or 4 heteroatoms or groups of heteroatoms independently selected from-NH-, -O-, -S-and N, respectively.
  2. A compound, isomer thereof or pharmaceutically acceptable salt thereof according to claim 1, wherein R1Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3
    Figure PCTCN2019101121-APPB-100006
    The CH3、CH 2CH 3
    Figure PCTCN2019101121-APPB-100007
    Optionally substituted by 1,2 or 3RaAnd (4) substitution.
  3. A compound, isomer thereof or pharmaceutically acceptable salt thereof according to claim 2, wherein R1Is selected from CH3And
    Figure PCTCN2019101121-APPB-100008
  4. a compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein R2Selected from H, F, Cl, Br, I, OH, NH2、CN、CH 3、CH 2CH 3And
    Figure PCTCN2019101121-APPB-100009
    the CH3、CH 2CH 3And
    Figure PCTCN2019101121-APPB-100010
    optionally substituted by 1,2 or 3RbAnd (4) substitution.
  5. A compound, isomer thereof or pharmaceutically acceptable salt thereof according to claim 4, wherein R2Is selected from CH3
  6. Any of claims 1 to 3The compound, an isomer thereof, or a pharmaceutically acceptable salt thereof, wherein RdSelected from H, F, Cl, Br, I, OH, NH2、CN、CH 3And
    Figure PCTCN2019101121-APPB-100011
  7. a compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein R4Selected from: H. f, Cl, Br, I, OH, NH2、CN、CH 3Pyrazolyl, isoxazolyl, imidazolyl, triazolyl, oxazolyl,
    Figure PCTCN2019101121-APPB-100012
    The CH3Pyrazolyl, isoxazolyl, imidazolyl, triazolyl, oxazolyl,
    Figure PCTCN2019101121-APPB-100013
    Optionally substituted by 1,2 or 3RdAnd (4) substitution.
  8. A compound, isomer, or pharmaceutically acceptable salt according to claim 7, wherein R4Selected from:
    Figure PCTCN2019101121-APPB-100014
    Figure PCTCN2019101121-APPB-100015
  9. the compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein the structural unit
    Figure PCTCN2019101121-APPB-100016
    Is selected from
    Figure PCTCN2019101121-APPB-100017
  10. The compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein ring A is selected from the group consisting of
    Figure PCTCN2019101121-APPB-100018
    Figure PCTCN2019101121-APPB-100019
  11. The compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein the structural unit
    Figure PCTCN2019101121-APPB-100020
    Is selected from
    Figure PCTCN2019101121-APPB-100021
  12. The compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to claim 11, wherein the structural unit
    Figure PCTCN2019101121-APPB-100022
    Is selected from
    Figure PCTCN2019101121-APPB-100023
  13. The compound, an isomer thereof, or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 3, wherein the structural unit
    Figure PCTCN2019101121-APPB-100024
    Is selected from
    Figure PCTCN2019101121-APPB-100025
  14. The compound, isomer thereof or pharmaceutically acceptable salt thereof according to any one of claims 1 to 9 and 10 to 13, which is selected from
    Figure PCTCN2019101121-APPB-100026
    Wherein R is1、R 2、R 4、L 2、L 3、T 1、T 2、T 3、T 4And T5The composition as defined in any one of claims 1 to 9 and 11 to 13.
  15. A compound of the formula, an isomer thereof, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
    Figure PCTCN2019101121-APPB-100027
    Figure PCTCN2019101121-APPB-100028
    Figure PCTCN2019101121-APPB-100029
  16. the compound of formula, an isomer thereof, or a pharmaceutically acceptable salt thereof according to claim 15, which is selected from the group consisting of:
    Figure PCTCN2019101121-APPB-100030
    Figure PCTCN2019101121-APPB-100031
    Figure PCTCN2019101121-APPB-100032
    Figure PCTCN2019101121-APPB-100033
  17. a pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1 to 16, an isomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient and a pharmaceutically acceptable carrier.
  18. Use of a compound according to any one of claims 1 to 16, an isomer thereof or a pharmaceutically acceptable salt thereof for the preparation of a RET kinase inhibitor.
  19. Use of a composition according to claim 17 for the preparation of a RET kinase inhibitor.
CN201980053423.0A 2018-08-17 2019-08-16 Pyrazole derivatives as RET inhibitors Pending CN112566907A (en)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
CN2018109473371 2018-08-17
CN201810947337 2018-08-17
CN201811322012 2018-11-07
CN2018113220120 2018-11-07
CN201811501055 2018-12-07
CN2018115010555 2018-12-07
CN201910482535 2019-06-04
CN201910482535X 2019-06-04
CN201910487945 2019-06-05
CN2019104879453 2019-06-05
PCT/CN2019/101121 WO2020035065A1 (en) 2018-08-17 2019-08-16 Pyrazole derivative as ret inhibitor

Publications (1)

Publication Number Publication Date
CN112566907A true CN112566907A (en) 2021-03-26

Family

ID=69524686

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201980053423.0A Pending CN112566907A (en) 2018-08-17 2019-08-16 Pyrazole derivatives as RET inhibitors

Country Status (2)

Country Link
CN (1) CN112566907A (en)
WO (1) WO2020035065A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111484479A (en) * 2019-01-25 2020-08-04 四川科伦博泰生物医药股份有限公司 Nitrogen heterocyclic compound, pharmaceutical composition containing nitrogen heterocyclic compound, preparation method and application of nitrogen heterocyclic compound
CN111499613A (en) * 2019-01-31 2020-08-07 浙江海正药业股份有限公司 N-formamide derivative, preparation method and medical application thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR121914A1 (en) 2020-04-27 2022-07-20 Lilly Co Eli COMPOUNDS USEFUL TO INHIBIT RET KINASE
US20230226057A1 (en) * 2020-06-25 2023-07-20 Tolremo Therapeutics Ag Heterocyclic derivatives, pharmaceutical compositions and their use in the treatment, amelioration or prevention of fibrotic disease
IL302542A (en) 2020-11-06 2023-07-01 Lilly Co Eli Pyrazole derivatives as ret kinase inhibitors
CN112656796B (en) * 2020-12-29 2022-06-24 北京鑫开元医药科技有限公司 Preparation composition of N- (5-methyl-1H-pyrazol-3-yl) pyridine-2-amine medicine
CN112279836B (en) * 2020-12-29 2021-04-30 北京鑫开元医药科技有限公司 N- (5-methyl-1H-pyrazol-3-yl) pyridine-2-amine compound and preparation method thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017079140A1 (en) * 2015-11-02 2017-05-11 Blueprint Medicines Corporation Inhibitors of ret
WO2017161269A1 (en) * 2016-03-17 2017-09-21 Blueprint Medicines Corporation Inhibitors of ret receptor tyrosine kinases
WO2018017983A1 (en) * 2016-07-22 2018-01-25 Blueprint Medicines Corporation Compounds useful for treating disorders related to ret
WO2018022761A1 (en) * 2016-07-27 2018-02-01 Blueprint Medicines Corporation Substituted cyclopentane-amides for treating disorders related to ret

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103242341B (en) * 2013-04-19 2015-12-09 中国科学院广州生物医药与健康研究院 Thieno-2,4 substituted pyrimidines compounds and pharmaceutical composition thereof and application
WO2016127074A1 (en) * 2015-02-06 2016-08-11 Blueprint Medicines Corporation 2-(pyridin-3-yl)-pyrimidine derivatives as ret inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017079140A1 (en) * 2015-11-02 2017-05-11 Blueprint Medicines Corporation Inhibitors of ret
WO2017161269A1 (en) * 2016-03-17 2017-09-21 Blueprint Medicines Corporation Inhibitors of ret receptor tyrosine kinases
WO2018017983A1 (en) * 2016-07-22 2018-01-25 Blueprint Medicines Corporation Compounds useful for treating disorders related to ret
WO2018022761A1 (en) * 2016-07-27 2018-02-01 Blueprint Medicines Corporation Substituted cyclopentane-amides for treating disorders related to ret

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111484479A (en) * 2019-01-25 2020-08-04 四川科伦博泰生物医药股份有限公司 Nitrogen heterocyclic compound, pharmaceutical composition containing nitrogen heterocyclic compound, preparation method and application of nitrogen heterocyclic compound
CN111484479B (en) * 2019-01-25 2023-06-13 四川科伦博泰生物医药股份有限公司 Azacyclic compounds, pharmaceutical compositions containing the same, and methods of making and using the same
CN111499613A (en) * 2019-01-31 2020-08-07 浙江海正药业股份有限公司 N-formamide derivative, preparation method and medical application thereof

Also Published As

Publication number Publication date
WO2020035065A1 (en) 2020-02-20

Similar Documents

Publication Publication Date Title
CN112566907A (en) Pyrazole derivatives as RET inhibitors
CN111372925B (en) Uracil compounds as c-MET/AXL inhibitors
CA2314115C (en) Process for the optical resolution of the racemic 6-(4-amino-phenyl)-5- methyl-pyridazin-3(2h)one
EP3333157B1 (en) Vinyl compounds as fgfr and vegfr inhibitors
BR112020001124A2 (en) compound or pharmaceutically acceptable salt thereof, pharmaceutical composition, use of the compound or pharmaceutically acceptable salt thereof and method for treating cancer
WO2019154365A1 (en) Atr inhibitor and application thereof
WO2020048546A1 (en) Tricyclic substituted piperidine dione compound
CN111683928B (en) Pyrazine-2 (1H) -ones as FGFR inhibitors
CN111344290A (en) Macrocyclic compound as Wee1 inhibitor and application thereof
CN110382479A (en) The fluoro- 3- hydroxyl propyl- 2- base ester derivative of carbamic acid 1,1,1- tri- as MAGL inhibitor
CN102256944A (en) Sphingosine-1-phosphate receptor antagonists
JP2019519606A (en) Thiophene compounds, their synthesis method and application in medicine
WO2020038457A1 (en) [1,2,4]triazolo[1,5-a]pyridine compound as jak inhibitor and application thereof
CN114105950B (en) Pyrazole compound and application thereof
CN111201225B (en) Coumarin-like ring compound as MEK inhibitor and application thereof
CN112601746B (en) Pyrazolopyrimidine derivatives and their use as PI3K inhibitors
CN114502561A (en) LSD1 inhibitors
TWI706950B (en) Diaza-benzofluoranthene compound
KR20230154953A (en) Glutarimide-based compounds substituted with furan condensed rings
WO2021129817A1 (en) Pyrimidine-based compound having inhibitory effect of ketohexokinase (khk)
CN107709323A (en) Oxipurinol class compound and its application
CN116425770A (en) Tetrafused ring compounds as Cdc7 inhibitors
WO2020094084A1 (en) Tricyclic derivative as ret inhibitor
CN111315750B (en) Pyridopyrimidines as mTORC1/2 dikinase inhibitors
CN106661009B (en) Anti- enteric virus71 thiadiazolidine derivative

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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210326

WD01 Invention patent application deemed withdrawn after publication