CN119768404A - Heteroaryl-substituted pyridopyrrolone derivatives, pharmaceutical compositions and applications thereof - Google Patents
Heteroaryl-substituted pyridopyrrolone derivatives, pharmaceutical compositions and applications thereofInfo
- Publication number
- CN119768404A CN119768404A CN202380062550.3A CN202380062550A CN119768404A CN 119768404 A CN119768404 A CN 119768404A CN 202380062550 A CN202380062550 A CN 202380062550A CN 119768404 A CN119768404 A CN 119768404A
- Authority
- CN
- China
- Prior art keywords
- alkyl
- ring
- halogenated
- membered
- pharmaceutically acceptable
- 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
Links
Abstract
The application relates to a heteroaryl substituted pyridopyrrolidone derivative, a pharmaceutical composition and application thereof. The heteroaryl substituted pyridopyrrolidone derivative has a structure shown in a formula (I). The compound has higher selective inhibition activity on P13K gamma, improves in vivo drug substitution activity, and has practical value.
Description
The present application claims priority from chinese patent office, application number CN202211062585.0 entitled "heteroaryl substituted pyridopyrrolidone derivatives, pharmaceutical compositions and uses thereof," filed on month 01 of 2022, the entire contents of which are incorporated herein by reference.
The application relates to the technical field of medicines, in particular to a heteroaryl substituted pyridopyrrolidone derivative, a pharmaceutical composition and application thereof.
Phosphoinositide 3-kinases (PI 3 ks) are a class of phosphatidylkinases with phosphoinositide cyclic phosphorylation and are divided into three classes, type I, type II and type III, with type I PI3 ks being the most widely studied. The types are further divided into two subgroups (IA and IB). Class IA PI3 ks consist of three closely related kinases pi3kα, pi3kβ and pi3kδ, existing as heterodimers consisting of catalytic subunits (p110α, p110β, or p110δ) and one of several regulatory subunits. Pi3kα and pi3kβ are widely expressed and play a role in cell growth, division and survival (Thomas M, et al, curr. Opin. Pharmacol.,2008; 8:267-274). Roles of these two kinases in many biological functions embryonic lethality observed in mice lacking pi3kα or pi3kβ is enhanced. Clinical evaluation of pi3kα and pi3kβ is limited to the oncology field due to their role in homeostasis, and some compounds are also at different stages of clinical development. PI3kγ single class 1B isomer, mainly responsive to G protein coupled receptors (G-protein coupled receptors, GPCRs), and consists of a p110γ catalytic subunit and one of two different regulatory subunits. PI3K gamma subtypes are expressed in immune cells and have limited expression in normal or malignant epithelial and connective tissue cells. The results of studies in PI3K gamma knockout mice indicate that PI3K gamma is important for cell activation and migration of some chemokines (Sasaki T., et al, science,2000;287:1040-1046; hirsch E., et al, science,2000; 287:1049-1053). PI3K gamma signaling is particularly important for bone marrow cell function and Camps et al describe that treatment with selective PI3K gamma inhibitor AS-60485023 can inhibit progression of joint inflammation and injury in two distinct mouse models of rheumatoid arthritis (Camps M, et al, nat. Med.2005,11, 936-943). Based on studies of cellular levels and efficacy observed in various disease models, PI3K gamma inhibitors can potentially be used to treat various diseases, e.g., inflammation, metabolism, cancer (Cushing,T.D.,et al.,J.Med.Chem.2012,55,8559-8581;Ruckle,T.,et al.,Nat.Rev.Drug Discovery 2006,5,903-918;Stark,A.K.,et al.,Curr.Opin.Pharmacol.2015,23,82-91).
Although selective PI3K gamma inhibitors such as WO2017153527 and WO2020210379 have been reported in various documents, the selectivity, pharmacokinetic parameters and other properties of these PI3K gamma inhibitors still need to be further improved, and thus there is a need in the art to develop PI3K gamma inhibitors with more excellent selectivity and pharmacokinetic properties suitable for later development to be better used in clinical demands.
Disclosure of Invention
The application aims to provide heteroaryl substituted pyridopyrrolidone derivatives with good selectivity and good pharmacokinetic properties.
In a first aspect, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof:
Wherein,
Ring a is a 5 or 6 membered heteroaryl ring;
(R 3)n represents that the hydrogen on the ring A is substituted by n R 3, n is 0, 1,2,3 or 4; each R 3 is the same or different and is each independently deuterium, halogen, cyano, hydroxy, carboxyl, C 1-3 alkyl, C 1-3 alkoxy, C 2-4 alkenyl, C 2-4 alkynyl, halogenated C 1-3 alkyl, halogenated C 1-3 alkoxy, NR a1Rb1、-N(Ra3)-C(O)C1-3 alkyl, -N (R a3) -C (O) -deuterated C 1-3 alkyl, -N (R a3)-C(O)OC1-3 alkyl), -SO 2C1-3 alkyl, -SO 2C3-6 cycloalkyl, -C (O) NR a1Rb1、-C(O)OC1-3 alkyl, -OC (O) C 1-3 alkyl, Wherein the 3-to 6-membered heterocycloalkyl, phenyl, 5-to 6-membered heteroaryl are each independently unsubstituted or substituted with 1,2, or 3 substituents each independently selected from substituent group Q;
Z is N or CR Z, T is N or CR T, U is N or CR U, W is N or CR W, Y is N or CR Y;
R Z、RT、RU is each independently hydrogen, halogen, NR a1Rb1 or C 1-8 alkyl, wherein the C 1-8 alkyl is preferably C 1-6 alkyl, more preferably C 1-3 alkyl;
R W、RY is each independently hydrogen, halogen, C 1-8 alkyl, halogenated C 1-8 alkyl, halogenated C 1-8 alkoxy, NR a1Rb1 or C 3-6 cycloalkyl, wherein the C 1-8 alkyl is preferably C 1-6 alkyl, more preferably C 1-3 alkyl;
Or R W、RY is linked to form a 9 or 10 membered heteroaryl ring, a 9 or 10 membered phenylheterocycloalkylring or a 9 or 10 membered heteroarylheterocycloalkylring with the adjacent 6 membered ring, each of said 9 or 10 membered heteroaryl ring, 9 or 10 membered phenylheterocycloalkylring, 9 or 10 membered heteroarylheterocycloalkylring being independently unsubstituted or substituted with 1, 2 or 3 substituents each independently selected from substituent group Q;
R 1、R2 is each independently C 1-8 alkyl, halogenated C 1-8 alkyl or C 3-6 cycloalkyl, wherein the C 3-6 cycloalkyl is unsubstituted or substituted with 1 or 2 substituents each independently selected from halogen and C 1-3 alkyl, the C 1-8 alkyl is preferably C 1-6 alkyl, more preferably C 1-3 alkyl, the halogenated C 1-8 alkyl is preferably halogenated C 1-6 alkyl, more preferably halogenated C 1-3 alkyl;
The substituent group Q is selected from halogen, cyano, hydroxy, carboxy, C 1-3 alkyl, C 1-3 alkoxy, C 2-4 alkenyl, C 2-4 alkynyl, halogenated C 1-3 alkyl, halogenated C 1-3 alkoxy, NR a1Rb1、-SO2C1-3 alkyl, -S (O) C 1-3 alkyl, -C (O) NR a1Rb1、-C(O)OC1-3 alkyl, -OC (O) C 1-3 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyloxy, 3-to 6-membered heterocycloalkyl, phenyl or 5-to 6-membered heteroaryl;
R a1、Rb1 is each independently hydrogen, C 1-3 alkyl or acetyl, or R a1、Rb1 together with the attached nitrogen atom forms a 4-to 6-membered saturated mono-heterocycle which is unsubstituted or substituted with 1,2 or 3 substituents each independently selected from deuterium, halogen, cyano, hydroxy, carboxy, C 1-3 alkyl, C 1-3 alkoxy, C 2-4 alkenyl, C 2-4 alkynyl, halo C 1-3 alkyl, halo C 1-3 alkoxy, -SO 2C1-3 alkyl, -S (O) C 1-3 alkyl, -C (O) NH 2、-C(O)NH(C1-3 alkyl), -C (O) N (C 1-3 alkyl) 2、-C(O)OC1-3 alkyl, -OC (O) C 1-3 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyloxy or 3-to 6-membered heterocycloalkyl;
R a3 is hydrogen or C 1-8 alkyl, wherein the C 1-8 alkyl is preferably C 1-6 alkyl, more preferably C 1-3 alkyl.
In one embodiment of the application, one of T, W, Y, U is N.
In one embodiment of the application, T is CR T, U is CR U, W is N or CR W, Y is N or CR Y, and W, Y is not both N.
In one embodiment of the application, two of T, W, Y, U are N.
In one embodiment of the application, T is N, U is CR U, W is CR W, and Y is N.
In one embodiment of the present application,Is selected from the structures shown in the formulas (A), (B) and (C):
In one embodiment of the application, the 9-or 10-membered heteroaryl ring is selected from the group consisting of indolyl, isoindolyl, indazolyl, benzotriazole, benzothienyl, isobenzothienyl, benzofuranyl, benzisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, indenazinyl, purinyl, pyrido [3,2-d ] pyrimidinyl, pyrido [2,3-d ] pyrimidinyl, pyrido [3,4-d ] pyrimidinyl, pyrido [4,3-d ] pyrimidinyl, 1, 8-naphthyridinyl, 1, 7-naphthyridinyl, 1, 6-naphthyridinyl, 1, 5-naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl.
In one embodiment of the present application, the heterocycloalkyl groups in the 9-or 10-membered phenylheterocycloalkylring, 9-or 10-membered heteroarylheterocycloalkylring are each independently selected from tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyrrolyl, oxazolidinyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, dioxane, thiomorpholinyl, thiomorpholin-1, 1-dioxide, tetrahydropyranyl, azetidin-2-one, oxetan-2-one, dihydrofuran-2 (3H) -one, pyrrolidin-2-one, pyrrolidine-2, 5-dione, dihydrofuran-2, 5-dione, piperidin-2-one, tetrahydro-2H-pyran-2-one, piperazin-2-one, morpholin-3-one.
In one embodiment of the application, the heteroaryl in the 9-or 10-membered heteroarylheterocycloalkylring is selected from pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl.
In one embodiment of the application, in formula (A), R T is hydrogen, R U is hydrogen, R W is hydrogen, halogen, C 1-6 alkyl, halogenated C 1-6 alkyl or halogenated C 1-6 alkoxy.
In one embodiment of the application, in formula (A), R T is hydrogen, R U is hydrogen, R W is hydrogen, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, trifluoromethyl or trifluoromethoxy.
In one embodiment of the application, in formula (B), R T is hydrogen, R U is hydrogen, R Y is hydrogen, halogen, C 1-6 alkyl, halogenated C 1-6 alkyl, halogenated C 1-6 alkoxy or C 3-6 cycloalkyl.
In one embodiment of the application, in formula (B), R T is hydrogen, R U is hydrogen, R Y is fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, trifluoromethoxy, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In one embodiment of the application, the structureSelected from one of the following structures:
In one embodiment of the application, ring A is selected from the group consisting of a thiophene ring, a furan ring, a thiazole ring, an isothiazole ring, an imidazole ring, an oxazole ring, a pyrrole ring, a pyrazole ring, a1, 2, 3-triazole ring, a1, 2, 4-triazole ring, a1, 2, 5-triazole ring, a1, 3, 4-triazole ring, a tetrazole ring, an isoxazole ring, a1, 2, 3-oxadiazole ring, a1, 2, 4-oxadiazole ring, a1, 2, 5-oxadiazole ring, a1, 3, 4-oxadiazole ring, a thiadiazole ring, a pyridine ring, a pyridazine ring, a pyrimidine ring, a pyrazine ring, a triazine ring, or a tetrazine ring.
Preferably, ring a is a thiazole ring.
Preferably, n is 2.
In one embodiment of the application, each R 3, which are the same or different, is independently C 1-3 alkyl, -NH-C (O) C 1-3 alkyl, -NH-C (O) -deuterated C 1-3 alkyl or-NH-C (O) OC 1-3 alkyl.
In one embodiment of the application, in formula (I),Is of the structure shown in formula (a):
R 01、R02 is each independently deuterium, halogen, cyano, hydroxy, carboxy, C 1-3 alkyl, C 1-3 alkoxy, C 2-4 alkenyl, C 2-4 alkynyl, halogenated C 1-3 alkyl, halogenated C 1-3 alkoxy, NR a1Rb1、-N(Ra3)-C(O)C1-3 alkyl, -N (R a3) -C (O) -deuterated C 1-3 alkyl, -N (R a3)-C(O)OC1-3 alkyl, -SO 2C1-3 alkyl, -SO 2C3-6 cycloalkyl, -C (O) NR a1Rb1、-C(O)OC1-3 alkyl, -OC (O) C 1-3 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyloxy, 3-to 6-membered heterocycloalkyl, phenyl, or 5-to 6-membered heteroaryl, wherein each of the 3-to 6-membered heterocycloalkyl, phenyl, 5-to 6-membered heteroaryl is independently unsubstituted or substituted with 1,2, or 3 substituents each independently selected from substituent group Q.
In one embodiment of the application, R 01 is halogen, cyano, hydroxy, carboxy, C 1-3 alkyl, C 1-3 alkoxy, halogenated C 1-3 alkyl or halogenated C 1-3 alkoxy; R 02 is-NH-C (O) C 1-3 alkyl-NH-C (O) -deuterated C 1-3 alkyl or-NH-C (O) OC 1-3 alkyl.
Preferably, R 01 is methyl, ethyl, n-propyl or isopropyl. Further preferably, R 01 is methyl.
Preferably, the method comprises the steps of, R 02 is-NH-C (O) C 1-3 alkyl-NH-C (O) -deuterated C 1-3 alkyl or-NH-C (O) OC 1-3 alkyl. Further preferably, R 02 is-NH-C (O) CH 3、-NH-C(O)-CD3 or-NH-C (O) OCH 3.
In one embodiment of the application, R 1 is a monochloromethyl, dichloromethyl, trichloromethyl, monochloroethyl, 1, 2-dichloroethyl, trichloroethyl, monobromoethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, difluoroethyl, trifluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, monofluoro-substituted cyclopropyl or monofluoro-substituted cyclobutyl, and R 2 is methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, monofluoro-substituted cyclopropyl or monofluoro-substituted cyclobutyl.
Preferably, R 1 is cyclopropyl, cyclobutyl or trifluoromethyl.
Preferably, R 2 is methyl, ethyl, cyclopropyl or cyclobutyl.
In one embodiment of the application, in formula (I),Selected from one of the following structures:
in one embodiment of the application, in formula (I), Selected from one of the following structures:
In one embodiment of the application, Z is N or CH.
In one embodiment of the application, the 5-or 6-membered heteroaryl (ring) groups described in the above formulae, R 3 and in the substituent group Q are each independently selected from thienyl, furyl, thiazolyl, isothiazolyl, imidazolyl, oxazolyl, pyrrolyl, pyrazolyl, triazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 5-triazolyl, 1,3, 4-triazolyl, tetrazolyl, isoxazolyl, oxadiazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl or tetrazinyl.
In one embodiment of the application, the 5-or 6-membered heteroaryl (ring) groups are each independently selected from the structures:
In one embodiment of the present application, the 3-to 6-membered heterocycloalkyl groups described in the above formulae, R 3 and substituent group Q are 4-to 6-membered heterocycloalkyl groups, each independently selected from azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, oxazolidinyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, dioxane, thiomorpholinyl, thiomorpholin-1, 1-dioxide, tetrahydropyranyl, pyrrolidin-2-one, dihydrofuran-2 (3H) -one, morpholin-3-one, piperazin-2-one and piperidin-2-one.
In one embodiment of the application, the compound of formula (I) is selected from any one of the following structures:
The second aspect of the present application provides a pharmaceutical composition comprising a compound according to the first aspect of the present application, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, and a pharmaceutically acceptable carrier.
In a third aspect, the present application provides the use of a compound according to the first aspect of the present application, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, and a pharmaceutical composition according to the second aspect of the present application, for the manufacture of a medicament for the treatment and/or prophylaxis of a disease associated with or mediated by pi3kγ activity.
In one embodiment of the application, the disease associated with or mediated by PI3K gamma activity is an inflammatory, metabolic or cancer disease.
It is understood that within the scope of the present application, the above-described technical features of the present application and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute new or preferred technical solutions. And are limited to a space, and are not described in detail herein.
The accompanying drawings, which are included to provide a further understanding of the application and are incorporated in and constitute a part of this specification, illustrate embodiments of the application and together with the description serve to explain the application and do not constitute a limitation on the application.
FIG. 1 is a graph showing tumor growth curves of a C57BL/6 mouse MC38 and macrophage mixed inoculation model;
FIG. 2 is a graph showing tumor weights at day 27 of C57BL/6 mice MC38 and macrophage mixed vaccination model dosing.
In order to make the objects, technical solutions and advantages of the present application more apparent, the present application will be further described in detail below by referring to the accompanying drawings and examples. It will be apparent that the described embodiments are only some, but not all, embodiments of the application. All other embodiments obtained by the person skilled in the art based on the present application fall within the scope of protection of the present application.
The present inventors have conducted extensive and intensive studies and have unexpectedly found that heteroaryl-substituted pyridopyrrolidone derivatives represented by the formula (I) have excellent PI3K gamma kinase and cell selective inhibitory activity, more excellent in vivo pharmacological parameters, and thus the series of compounds are expected to be developed as PI3K gamma selective inhibitors and are useful for the treatment and/or prevention of diseases associated with PI3K gamma activity. On the basis of this, the applicant has completed the present application.
Definition of terms
In order that the technical content of the present application can be more clearly understood, the following terms of the present application will be further described.
"Alkyl" refers to straight and branched chain saturated aliphatic hydrocarbon groups. "C 1-8 alkyl" refers to an alkyl group having 1 to 8 carbon atoms, preferably a C 1-6 alkyl group, more preferably a C 1-3 alkyl group; non-limiting examples of alkyl groups include: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof.
"Alkenyl" refers to a straight or branched unsaturated aliphatic hydrocarbon group having one or more carbon-carbon double bonds (c=c), and "C 2-8 alkenyl" refers to alkenyl groups having 2 to 8 carbon atoms, preferably C 2-6 alkenyl, more preferably C 2-4 alkenyl, similarly defined, non-limiting examples of alkenyl groups include ethenyl, propenyl, isopropenyl, n-butenyl, isobutenyl, pentenyl, hexenyl, and the like.
"Alkynyl" refers to straight and branched chain unsaturated aliphatic hydrocarbon groups having one or more carbon-carbon triple bonds, "C 2-8 alkynyl" refers to alkynyl groups having 2 to 8 carbon atoms, preferably C 2-6 alkynyl, more preferably C 2-4 alkynyl, similarly defined, non-limiting examples of alkynyl groups include ethynyl, propynyl, n-butynyl, isobutynyl, pentynyl, hexynyl, and the like.
"Cycloalkyl" and "cycloalkyl ring" are used interchangeably and refer to a saturated monocyclic, bicyclic, or polycyclic cyclic hydrocarbon group, which may be fused to an aryl or heteroaryl group. The cycloalkyl ring may be optionally substituted. In certain embodiments, the cycloalkyl ring contains one or more carbonyl groups, such as oxo groups. "C 3-8 cycloalkyl" refers to a monocyclic cycloalkyl group having 3 to 8 carbon atoms, non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclobutanone, cyclopentanone, cyclopentane-1, 3-dione, and the like. Preferred are C 3-6 cycloalkyl groups including cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. "C 8-10 cycloalkyl" refers to a fused bicyclic cyclic hydrocarbon group having 8 to 10 ring atoms, non-limiting examples of C 8-10 cycloalkyl include
"Heterocycloalkyl" and "heterocycloalkyl ring" are used interchangeably and refer to a cycloalkyl group containing at least one heteroatom selected from nitrogen, oxygen and sulfur, which group may be fused to an aryl or heteroaryl group. The heterocycloalkyl ring may be optionally substituted. In certain embodiments, the heterocycloalkyl ring contains one or more carbonyl or thiocarbonyl groups, for example, groups containing oxo and thio. "3-to 8-membered heterocycloalkyl" means a monocyclic cyclic hydrocarbon group having 3 to 8 ring atoms, wherein 1,2 or 3 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, preferably 4-to 8-membered heterocycloalkyl. More preferably a 3 to 6 membered heterocycloalkyl group having 3 to 6 ring atoms wherein 1 or 2 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur. Further preferred are 4 to 6 membered heterocycloalkyl groups having 4 to 6 ring atoms, wherein 1 or 2 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur. Non-limiting examples include aziridinyl, oxiranyl, azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, oxazolidinyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, dioxacyclyl, thiomorpholinyl, thiomorpholin-1, 1-dioxide, tetrahydropyranyl, azetidin-2-one, oxetan-2-one, dihydrofuran-2 (3H) -one, pyrrolidin-2-one, pyrrolidine-2, 5-dione, dihydrofuran-2, 5-dione, piperidin-2-one, tetrahydro-2H-pyran-2-one, piperazin-2-one, morpholin-3-one, and the like. "6-to 12-membered heterocycloalkyl" and "6-to 12-membered fused heterocycloalkyl" are used interchangeably and refer to a fused bicyclic cyclic hydrocarbon group having 6 to 12 ring atoms, wherein 1,2 or 3 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur. "8-to 10-membered heterocycloalkyl" and "8-to 10-membered fused heterocycloalkyl" are used interchangeably and refer to a fused bicyclic cyclic hydrocarbon group having 8 to 10 ring atoms, wherein 1,2 or 3 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur. Non-limiting examples include hexahydro-1H-furo [3,4-c ] pyrrole, octahydro-1H-cyclopenta [ c ] pyridine, hexahydro-1H-pyrrolo [2,1-c ] [1,4] oxazine, octahydropyrrolo [1,2-a ] pyrazine, hexahydropyrrolo [1,2-a ] pyrazin-4 (1H) -one, octahydrocyclopenta [ c ] pyrrole, and the like. In a fused bicyclic heterocycloalkyl group containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom as the valency permits. The bicyclic heterocycloalkyl system may include one or more heteroatoms in one or both rings.
"Heteroaryl" and "heteroaryl ring" are used interchangeably and refer to a group of a monocyclic, bicyclic, or polycyclic 4n+2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) having ring carbon atoms and ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur. Heteroaryl rings may be optionally substituted. "5-to 10-membered heteroaryl" refers to a monocyclic or bicyclic heteroaryl group having 5 to 10 ring atoms, wherein 1,2,3 or 4 ring atoms are heteroatoms. "5-to 6-membered heteroaryl" refers to a monocyclic heteroaryl group having 5 to 6 ring atoms, wherein 1,2,3 or 4 ring atoms are heteroatoms, non-limiting examples include thienyl, furyl, thiazolyl, isothiazolyl, imidazolyl, oxazolyl, pyrrolyl, pyrazolyl, triazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 5-triazolyl, 1,3, 4-triazolyl, tetrazolyl, isoxazolyl, oxadiazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl. "9 or 10 membered heteroaryl" refers to a bicyclic heteroaryl group having 9 or 10 ring atoms, wherein 1,2,3 or 4 ring atoms are heteroatoms, non-limiting examples of which include indolyl, isoindolyl, indazolyl, benzotriazole, benzothienyl, isobenzothienyl, benzofuranyl, benzisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, indenazinyl, purinyl, pyrido [3,2-d ] pyrimidinyl, pyrido [2,3-d ] pyrimidinyl, pyrido [3,4-d ] pyrimidinyl, 1, 8-naphthyridinyl, 1, 7-naphthyridinyl, 1, 6-naphthyridinyl, 1, 5-naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, quinazolinyl and quinazolinyl. "heteroatom" means nitrogen, oxygen or sulfur. In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as the valency permits. The heteroaryl bicyclic ring system may include one or more heteroatoms in one or both rings.
"Fused" refers to a structure in which two or more rings share one or more bonds.
"Phenylacetycloalkyl" refers to groups in which the benzene ring is fused to a heterocycloalkyl ring to form a bicyclic, tricyclic, or polycyclic ring system, wherein the heterocycloalkyl ring is as defined above. "9 or 10 membered phenylaminoheterocycloalkyl" means a bicyclic cyclic group having 9 or 10 ring atoms wherein 1,2,3 or 4 ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur. Non-limiting examples include indoline, benzo [ d ] [1,3] dioxazole, 1,2,3, 4-tetrahydroisoquinoline, 3, 4-dihydro-2H-benzo [ b ] [1,4] oxazine, indol-2-one, isoindol-1-one, 1, 4-dihydroisoquinolin-3 (2H) -one, and the like.
"Heteroaryl-heterocycloalkyl" refers to a group in which a heteroaryl ring is fused to a heterocycloalkyl ring to form a bicyclic, tricyclic, or polycyclic ring system, wherein the heterocycloalkyl ring is as defined above. "9 or 10 membered heteroarylheterocycloalkyl" refers to a bicyclic ring group having 9 or 10 ring atoms, where 1,2,3, or 4 ring atoms are heteroatoms selected from nitrogen, oxygen, and sulfur. Non-limiting examples include 2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridine, [1,3] dioxolo [4,5-b ] pyridine, 2, 3-dihydro-1H-pyrido [3,4-b ] [1,4] oxazine, 2,3,4, 6-tetrahydropyrrolo [3,4-b ] [1,4] oxazine, 2,4,5, 6-tetrahydropyrano [2,3-c ] pyrazole, 5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidine, and the like.
"Alkoxy" refers to an-O-alkyl group, wherein alkyl is as defined above. Preferably a C 1-8 alkoxy group, more preferably a C 1-6 alkoxy group, most preferably a C 1-3 alkoxy group. Non-limiting examples of alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, butoxy, t-butoxy, isobutoxy, pentoxy, and the like.
"Cycloalkyloxy" refers to-O-cycloalkyl, wherein cycloalkyl is as defined above. Preferably a C 3-8 cycloalkyloxy group, more preferably a C 3-6 cycloalkyloxy group. Non-limiting examples of cycloalkyloxy groups include cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
"A bond" means that two groups joined by it are joined by a covalent bond.
"Halogen" means fluorine, chlorine, bromine or iodine.
"Halo" means that one or more (e.g., 1,2,3, 4, or 5) hydrogens in the group are replaced with a halogen.
For example, "haloalkyl" refers to hydrogen on an alkyl group substituted with one or more (e.g., 1,2, 3, 4, or 5) halogens, where the alkyl group is defined above. Preferably a halogenated C 1-8 alkyl group, more preferably a halogenated C 1-6 alkyl group, and even more preferably a halogenated C 1-3 alkyl group. Examples of haloalkyl include, but are not limited to, monochloromethyl, dichloromethyl, trichloromethyl, monochloroethyl, 1, 2-dichloroethyl, trichloroethyl, monobromoethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, difluoroethyl, trifluoroethyl, and the like.
Also for example, "haloalkoxy" refers to hydrogen on an alkoxy group substituted with one or more (e.g., 1,2,3,4, or 5) halogens, where the definition of the alkoxy group is as described above. Preferably a halogenated C 1-8 alkoxy group, more preferably a halogenated C 1-6 alkoxy group, and even more preferably a halogenated C 1-3 alkoxy group. Haloalkoxy groups include, but are not limited to, trifluoromethoxy, trifluoroethoxy, monofluoromethoxy, monofluoroethoxy, difluoromethoxy, difluoroethoxy, and the like.
Also for example, "halocycloalkyl" refers to a cycloalkyl group in which the hydrogen on the cycloalkyl group is substituted with one or more (e.g., 1,2,3,4, or 5) halogens, wherein cycloalkyl is as defined above. Preferably a halogenated C 3-8 cycloalkyl group, more preferably a halogenated C 3-6 cycloalkyl group. Halogenated cycloalkyl groups include, but are not limited to, trifluorocyclopropyl, monofluorocyclopropyl, monofluorocyclohexyl, difluorocyclopropyl, difluorocyclohexyl, and the like.
"Deuterated alkyl" refers to an alkyl group in which the hydrogen is replaced by one or more (e.g., 1,2,3, 4, or 5) deuterium atoms, wherein alkyl is as defined above. Preferably deuterated C 1-8 alkyl, more preferably deuterated C 1-6 alkyl, more preferably deuterated C 1-3 alkyl. Examples of deuterated alkyl groups include, but are not limited to, mono-deuterated methyl, mono-deuterated ethyl, di-deuterated methyl, di-deuterated ethyl, tri-deuterated methyl, tri-deuterated ethyl, and the like.
"Amino" means NH 2, "cyano" means CN, "nitro" means NO 2, "benzyl" means-CH 2 -phenyl, "oxo" means =o, "carboxy" means-C (O) OH, "acetyl" means-C (O) CH 3, "hydroxymethyl" means-CH 2 OH, "hydroxyethyl" means-CH 2CH2 OH or-CHOHCH 3, "hydroxy" means-OH, "mercapto" means SH, "cyclopropyl" structure:
"substituted" means that one or more, preferably 1 to 5, hydrogen atoms in the group are each independently substituted with a corresponding number of substituents, more preferably 1 to 3 hydrogen atoms are each independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
Unless otherwise defined, the "substituents each independently selected from the group consisting of:. And:. Are defined as substituents, and when one or more hydrogens on a group are replaced with a substituent, the substituent types may be the same or different, and the substituents selected are each independent type.
Unless otherwise defined, the ". The application is identical or different, and each independently is an independent species, meaning that when more than one identical substituent group is present in the formula, the groups may be the same or different. For example, L is (CR 01R02)s, when s is 2, i.e., L is (CR 01R02)-(CR01R02), wherein two R 01 may be the same or different, and two R 02 may be the same or different, each independently of the other, e.g., L may be C (CH 3)(CN)-C(CH2CH3)(OH),C(CH3)(CN)-C(CH3) (OH) or C (CN) (CH 2CH3)-C(OH)(CH2CH3).
Any of the groups of the present application may be substituted or unsubstituted, unless otherwise defined. When the above groups are substituted, the substituents are preferably 1 to 5 or less and are independently selected from cyano, halogen (preferably fluorine or chlorine), C 1-8 alkyl (preferably C 1-6 alkyl, more preferably C 1-3 alkyl), C 1-8 alkoxy (preferably C 1-6 alkoxy, more preferably C 1-3 alkoxy), Halogenated C 1-8 alkyl (preferably halogenated C 1-6 alkyl, more preferably halogenated C 1-3 alkyl), C 3-8 cycloalkyl (preferably C 3-6 cycloalkyl), Halogenated C 1-8 alkoxy (preferably halogenated C 1-6 alkoxy, more preferably halogenated C 1-3 alkoxy), C 1-8 alkyl substituted amino, Halo C 1-8 alkyl substituted amino, acetyl, hydroxy, hydroxymethyl, hydroxyethyl, carboxyl, nitro, C 6-10 aryl (preferably phenyl), C 3-8 cycloalkyloxy (preferably C 3-6 cycloalkyloxy), C 2-8 alkenyl (preferably C 2-6 alkenyl, more preferably C 2-4 alkenyl), C 2-8 alkynyl (preferably C 2-6 alkynyl, more preferably C 2-4 alkynyl), -CONR a0Rb0、-C(O)OC1-10 alkyl (preferably-C (O) OC 1-6 alkyl, more preferably-C (O) OC 1-3 alkyl), -CHO, -OC (O) C 1-10 alkyl (preferably-OC (O) C 1-6 alkyl, more preferably-OC (O) C 1-3 alkyl), -SO 2C1-10 alkyl (preferably-SO 2C1-6 alkyl, more preferably-SO 2C1-3 alkyl), -SO 2C6-10 aryl (preferably-SO 2C6 aryl, such as-SO 2 -phenyl), -COC 6-10 aryl (preferably-COC 6 aryl, such as-CO-phenyl), 4 to 6 membered saturated or unsaturated mono-heterocycle, 4 to 6 membered saturated or unsaturated mono-ring, 5 to 6 membered mono-ring heteroaryl ring, 8 to 10 membered bi-ring heteroaryl ring, spiro ring, bridged ring or bridged ring, wherein R a0、Rb0 is each independently hydrogen or C 1-3 alkyl.
In the present application, when two or more "preferences" occur in one embodiment, any two "preferences" may be independent of each other.
In the present application, when the number of substituents is greater than 1, any two substituents may be the same or different. For example, two identical or different halogen substitutions may be made, one halogen and one hydroxy substitution.
The various substituents described above in the present application may themselves be substituted with the groups described in the present application.
When the 4-to 6-membered saturated mono-heterocycle of the present application is substituted, the positions of the substituents may be in their possible chemical positions, and representative substitution cases for exemplary mono-heterocycles are as follows:
Wherein "Sub" represents various substituents described herein; Indicating the connection position to other atoms.
Pharmaceutical composition
In general, the compounds of the application or pharmaceutically acceptable salts or stereoisomers thereof may be administered in a suitable dosage form with one or more pharmaceutically acceptable carriers. These dosage forms are suitable for oral, rectal, topical, intraoral, and other parenteral administration (e.g., subcutaneous, intramuscular, intravenous, etc.). For example, dosage forms suitable for oral administration include capsules, tablets, granules, syrups and the like. The compounds of the present application contained in these formulations may be solid powders or granules, solutions or suspensions in aqueous or non-aqueous liquids, water-in-oil or oil-in-water emulsions, and the like. The above-mentioned dosage forms can be prepared from the active compound and one or more carriers or adjuvants by means of customary pharmaceutical methods. The above-mentioned carriers are required to be compatible with the active compound or other excipients. The active compound may form a solution or suspension with the carrier.
By "pharmaceutically acceptable carrier" is meant a non-toxic, inert, solid, semi-solid substance or liquid filling machine, diluent, encapsulating material or co-formulation or any type of adjuvant compatible with the patient, preferably a mammal, more preferably a human, which is suitable for delivering the active agent to the target site without stopping the activity of the agent.
"Compounds of the present application" refers to compounds of formula (I) of the present application, or pharmaceutically acceptable salts thereof, or stereoisomers thereof, which have PI3K gamma selective inhibitory activity, and intermediate compounds for preparing compounds of formula (I), or pharmaceutically acceptable salts thereof, or stereoisomers thereof.
The compositions of the present application are formulated, quantified and administered in a manner consistent with medical practice specifications. The "therapeutically effective amount" of a compound to be administered will be determined by the particular condition being treated, the individual being treated, the cause of the condition, the target of the drug, and the mode of administration, among other factors.
By "therapeutically effective amount" is meant an amount of a compound of the application that will elicit a biological or medical response in an individual, e.g., reduce or inhibit enzyme or protein activity or ameliorate symptoms, alleviate a condition, slow or delay the progression of a disease or prevent a disease, etc.
The therapeutically effective amount of the compound of the present application or a pharmaceutically acceptable salt thereof or a stereoisomer thereof contained in the pharmaceutical composition of the present application is preferably 0.1mg/kg to 5g/kg (body weight).
By "patient" is meant an animal, preferably a mammal, more preferably a human. The term "mammal" refers to a warm-blooded vertebrate mammal, including, for example, cats, dogs, rabbits, bears, foxes, wolves, monkeys, deer, mice, pigs, and humans.
"Treating" refers to alleviating, slowing progression, attenuating, preventing, or maintaining an existing disease or condition (e.g., cancer). Treatment also includes curing, preventing the development of, or alleviating to some extent, one or more symptoms of the disease or disorder.
The "pharmaceutically acceptable salts" include pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts. Pharmaceutically acceptable acid addition salts refer to salts with inorganic or organic acids that retain the biological effectiveness of the free base without other side effects. These salts can be prepared by methods known in the art.
"Pharmaceutically acceptable base addition salts" include, but are not limited to, salts of inorganic bases such as sodium, potassium, calcium, and magnesium salts, and the like. Including but not limited to salts of organic bases such as ammonium salts, triethylamine salts, lysine salts, arginine salts, and the like. These salts can be prepared by methods known in the art.
When the compounds of formula (I) according to the application contain one or more chiral centers, they may exist in different optically active forms. When the compound of formula (I) contains a chiral center, the compound contains a pair of enantiomers. Both enantiomers of the compound as well as mixtures of the pair of enantiomers, such as racemic mixtures, are also within the scope of the present application. Enantiomers may be resolved by methods known in the art, such as crystallization and chiral chromatography. When the compound of formula (I) contains more than one chiral center, the compound comprises enantiomers and diastereomers. All enantiomers and diastereomers, as well as mixtures of enantiomers, mixtures of diastereomers, and mixtures of enantiomers and diastereomers of such compounds are also within the scope of the application. Enantiomers, diastereomers may be resolved by methods known in the art, such as crystallization and preparative chromatography.
Preparation method
The present application provides methods for the preparation of compounds of formula (I), which may be synthesized using standard synthetic techniques known to those skilled in the art or using methods known in the art in combination with the methods described herein. The solvents, temperatures and other reaction conditions set forth herein may vary according to the art. The reactions may be used sequentially to provide the compounds of the application, or they may be used to synthesize fragments that are subsequently added by the methods described herein and/or by methods known in the art.
The compounds described herein may be synthesized using methods analogous to those described below or exemplary methods described in the examples, or related publications used by those skilled in the art, by using appropriate alternative starting materials. The starting materials for the synthesis of the compounds described herein may be synthesized or may be obtained from commercial sources. The compounds described herein and other related compounds having different substituents can be synthesized using techniques and starting materials known to those skilled in the art. The general methods of preparing the compounds disclosed herein may come from reactions known in the art, and the reactions may be modified by reagents and conditions deemed appropriate by one skilled in the art to introduce various moieties into the molecules provided herein.
The application has the beneficial effects that:
A range of heteroaryl substituted pyridopyrrolidone derivatives are provided which have a higher selective inhibitory activity against PI3K gamma enzymes and cells and have more excellent in vivo pharmacokinetic parameters, an inhibitory activity against PI3K gamma kinase with an IC 50 value of less than 100nM, in some embodiments with an IC 50 value of less than 50nM, in some embodiments with an IC 50 value of less than 10nM, and are therefore useful as medicaments for the treatment and/or prophylaxis of diseases associated with or mediated by PI3K gamma activity.
The application will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present application and are not intended to limit the scope of the present application. The experimental procedure, which does not address the specific conditions in the examples below, is generally followed by conventional conditions such as those described in Sambrook et al, molecular cloning, A laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989), or by the manufacturer's recommendations. Percentages and parts are by weight unless otherwise indicated. Unless defined otherwise, terms used herein have the same meaning as those familiar to one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the present application.
Known starting materials may be synthesized using or following methods known in the art, or may be purchased from ABCR GmbH & Co.KG, acros Organics, ALDRICH CHEMICAL Company, shao Yuan chemical technology (Accela ChemBio Inc), darui chemical, and other companies.
Unless otherwise specified, the reactions in the examples were all carried out under nitrogen or argon atmosphere.
DCM, dichloromethane, DMF, dimethylformamide, THF, tetrahydrofuran, pd (dppf) Cl 2, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium dichloride, dppf 1,1' -bis (diphenylphosphino) ferrocene, pin 2B2, bisboronic acid pinacol ester, SEM-Cl 2- (trimethylsilyl) ethoxymethyl chloride, ACN, acetonitrile, TFA, trifluoroacetic acid, DCM, meOH, n-BuLi, n-butyllithium, KOAc, potassium acetate, 1,4-Dioxane, combiflash, EA, ethyl acetate, PE, petroleum ether, DMSO, BSA, bovine serum albumin, FA, formic acid, DDQ, 3-dichloro-5, 6-dicyanop-benzoquinone.
The percentage content referred to in the present application refers to mass percentage for both solid-liquid mixing and solid-solid mixing and volume percentage for liquid-liquid mixing unless otherwise specified.
The final product of the application can be obtained by preparation and purification, and the purification and separation conditions are obtained by testing the routine knowledge in the field according to the characteristics of the compound by a person skilled in the art.
As used herein, room temperature refers to about 20-30 ℃.
Yield = actual synthetic product mass/theoretical synthetic product mass x 100%.
Preparation of intermediate a
Step 12, 6-dichloro-4-methylnicotinic acid (40.00 g,194.15 mmol) and potassium carbonate (67.08 g,485.38 mmol) were dissolved in DMF (200 mL), methyl iodide (41.34 g,291.23 mmol) was added dropwise at room temperature, then reacted for 1 hour at 24℃the reaction solution was poured into water, extracted with ethyl acetate, the organic phase was washed twice with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give methyl 2, 6-dichloro-4-methylnicotinate (39.00 g, yield: 92.86%) as a brown crude product. MS (ESI) 220.0[ M+H ] +.
Step 2 the crude product methyl 2, 6-dichloro-4-methylnicotinate (39.00 g,177.23 mmol) and N-bromosuccinimide (47.32 g,265.85 mmol) and benzoyl peroxide (64.40 g,265.85 mmol) were dissolved in carbon tetrachloride CCl 4 (300 mL) and the reaction was then brought to 90℃and stirred at 90℃for 48 hours. Water was added, extracted with dichloromethane, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give a crude product. Separation using CombiFlash (0-30%, ethyl acetate/petroleum ether) successfully yielded a yellow oily mixture of methyl 2, 6-dichloro-4-methylnicotinate-4- (bromomethyl) -2, 6-dichloronicotinate (25.00 g, yield: 48.08%) and methyl 2, 6-dichloro-4- (dibromomethyl) nicotinate (25.00 g, yield: 37.88%). MS (ESI) 299.8[ M+H ] +, MS (ESI) 379.7[ M+H ] +.
Step 3, dissolving crude product methyl 2, 6-dichloro-4-methylnicotinate-4- (bromomethyl) -2, 6-dichloronicotinate (25.00 g,84.20 mmol) and methyl 2, 6-dichloro-4- (dibromomethyl) nicotinate (25.00 g,66.17 mmol) in ACN (300 mL), cooling to 5 ℃ under the protection of N 2, dropwise adding N, N-diisopropylethylamine (34.20 g,264.66 mmol) at the temperature, dropwise adding diethyl phosphite (18.14 g,132.33 mmol) at the temperature, reacting for 0.5 hours, adding saturated sodium bicarbonate aqueous solution, extracting with ethyl acetate, washing twice with saturated salt water, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, and separating by adopting Combiflash (0-20% ethyl acetate/petroleum ether). The product methyl 2, 6-dichloro-4-methylnicotinic acid-4- (bromomethyl) -2, 6-dichloronicotinic acid (28.00 g, yield: 70.78%) was obtained as a yellow oil. MS (ESI) 299.9[ M+H ] +.
Step 4 methyl 2, 6-dichloro-4-methylnicotinic acid-4- (bromomethyl) -2, 6-dichloronicotinic acid (28.00 g,93.66 mmol) and (S) -1-cyclopropylethylamine hydrochloride (11.39 g,93.66 mmol) were dissolved in acetonitrile (200 mL), and then boric acid (5.79 g,93.66 mmol) and potassium carbonate (25.89 g,187.32 mmol) were added thereto. The reaction was stirred at 24 ℃ for 12 hours. The suspension is filtered, and the filtrate is dried by spin to obtain a crude product. Separation using CombiFlash (0-50%, ethyl acetate/petroleum ether) afforded intermediate a (22.60 g, yield: 88.99%) as a white solid. MS (ESI) 271.0[ M+H ] +.
Preparation of intermediate b
N-BuLi (2.5M in THF,5.12mL,12.8mmol) was taken and added dropwise to a solution of diisopropylamine (1.29 g,12.8 mmol) in THF (20 mL) at 0deg.C, and the reaction was maintained at 0deg.C for 30 min. The above solution was slowly added dropwise to a solution of N- (4-methylthiazol-2-yl) acetamide (500 mg,3.2 mmol) in THF (5 mL) at-78℃and the reaction continued for 30 min. 2-isopropyl-4, 5-tetramethyl-1, 3, 2-dioxaborane (1.19 g,6.4 mmol) was slowly dropped into the reaction solution, and the reaction was kept at-78℃for two hours. Quench with saturated ammonium chloride solution, extract with ethyl acetate, wash the organic phase twice with saline, dry over sodium sulfate, filter and concentrate to give intermediate b (410 mg, yield: 45%) as a pale yellow solid. 1HNMR(400MHz,CDCl3 ) δ2.53 (s, 3H), 2.25 (d, j=2.4 hz, 3H), 1.25 (s, 12H).
Preparation of intermediate 1
Intermediate a (1.200 g,4.43 mmol) and intermediate b (3.75 g,13.28 mmol) were dissolved in N, N-dimethylformamide (20 mL) and bis (2-diphenylphosphinocyclopentan-2, 4-dien-1-yl) iron dichloropalladium (323.83 mg, 442.57. Mu. Mol), sodium carbonate (1.41 g,13.28 mmol) was added under argon and the mixture was stirred for 10 hours at 100 ℃. LC-MS detection finds the target product. The reaction solution was poured into water, extracted with methylene chloride (50 ml. Times.2), dried over anhydrous sodium sulfate, concentrated to dryness under reduced pressure, and purified by silica gel column separation (EA: PE=0 to 100%) to give intermediate 1 (380 mg, 972.15. Mu. Mol, yield: 21.97%) as a yellow solid. MS (ESI) 391[ M+H ] +.
Preparation of intermediate 2
6-Bromoindolin-2-one (500 mg,2.36mmol,1 eq), pin 2B2 (719 mg,2.83mmol,1.2 eq), KOAc (463 mg,4.72mmol,2 eq) were dissolved in1, 4-dioxane (20 mL), replaced with argon, pd (dppf) Cl 2 (176 mg,0.24mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred at 120℃for 6 hours under argon protection. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted three times with methylene chloride (30 mL. Times.3). The organic phase was washed once with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate and spun-dried to give intermediate 2 (1.1 g, crude) as a dark grey solid. Can be directly put into the next step.
Preparation of intermediate 3
6-Bromoisoindolin-1-one (500 mg,2.36mmol,1 eq), pin 2B2 (719 mg,2.83mmol,1.2 eq), KOAc (463 mg,4.72mmol,2 eq) were dissolved in1, 4-dioxane (20 mL), replaced with argon, pd (dppf) Cl 2 (176 mg,0.24mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred for 6 hours under argon protection. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted three times with methylene chloride (30 mL. Times.3). The organic phase was washed once with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate and spun-dried to give 1g of crude solid intermediate 3 (1 g, crude) as dark grey. Directly put into the next step.
Preparation of intermediate 4
4-Bromo-2-fluoropyridine (2 g,11.36mmol,1 eq), pin 2B2 (3.463 g,13.64mmol,1.2 eq), KOAc (2.23 g,22.72mmol,2 eq) were dissolved in 1, 4-dioxane (20 mL), replaced with argon, pd (dppf) Cl 2 (416 mg, 0.418 mmol,0.1 eq), replaced with argon, and stirred under argon for 5 hours at 120 ℃. The reaction was checked for success by LCMS. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted three times with methylene chloride (30 mL. Times.3). The organic phase was washed once with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate and spun-dried to give crude intermediate 4 (2.4 g, crude).
Preparation of intermediate 5
Step 1 6-Chloropyrazoles (1 g,6.51mmol,1 eq) were dissolved in DMF (10 mL), naH (188 mg,7.81mmol,1.2eq,60 wt%) was added under argon at-5℃for 10min, SEM-Cl (1.3 g,7.81mmol,1.2 eq) was added dropwise at-5℃and the ice bath was removed after addition and the reaction was resumed at room temperature (rt) for 3h 10 min. The reaction was checked for success by LCMS. The reaction mixture was poured into an appropriate amount of ice water, extracted 3 times with ethyl acetate (30 mL. Times.3), and washed 1 time with a saturated aqueous solution of sodium chloride (30 mL). The combined organic phases were dried over anhydrous sodium sulfate, dried by spin-drying, and purified by column to give the product 6-chloro-1- (2-trimethylsilylethoxy) methylpyrazolopyridine (1.3 g, purity: 88%, yield: 62%, as a pale yellow liquid).
Step 2 6-chloro-1- (2-trimethylsilylethoxy) methylpyrazolopyridine (1.3 g,4.58mmol,1 eq), pin 2B2 (1.4 g,5.50mmol,1.2 eq), KOAc (899 mg,9.16mmol,2 eq) were dissolved in DMF (20 mL), replaced with argon, pd (dppf) Cl 2 (335 mg,0.4mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred for 5 hours at 100℃under argon protection. The reaction was successfully detected by LCMS. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted three times with methylene chloride (30 mL. Times.3). The organic phase was washed once with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate and spun-dried to give intermediate 5 (1.9 g, purity: 77%, yield: 99%, as a dark gray solid).
Preparation of intermediate 6
Step 1 6-chloropyrrole (4.5 g,29.49mmol,1 eq) was dissolved in DMF (40 mL), replaced with argon, under argon protection, naH (60 wt%) (849 mg,35.39mmol,1.2 eq) was added at-5℃and reacted for 10min, SEM-Cl (5.9 g,35.39mmol,1.2 eq) was added dropwise at-5℃and the ice bath was removed and the reaction was resumed at Room Temperature (RT) for 3 h. The reaction was checked for success by LCMS. The reaction solution was poured into ice water, extracted 3 times with ethyl acetate, washed 1 time with sodium chloride, dried over anhydrous sodium sulfate, and purified by spin-drying through a column to give the product 6-chloro-1- (2-trimethylsilylethoxy) methylpyrrolidine (8 g, purity: 80.08%, yield: 76.80%, as a pale yellow liquid).
Step 2 6-chloro-1- (2-trimethylsilylethoxy) methylpyrrolidine (3 g,10.61mmol,1 eq), pin 2B2 (3.2 g,12.73mmol,1.2 eq), KOAc (2.1 g,21.22mmol,2 eq) were dissolved in1, 4-dioxane (30 mL), replaced with argon, pd (dppf) Cl 2 (776 mg,1.06mmol,0.1 eq), replaced with argon and stirred for 5 hours at 90℃under argon protection. The reaction was checked by LCMS and was successful. After the reaction solution was cooled, dichloromethane was extracted with water, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and spun-dried to obtain intermediate 6 (3.5 g, purity: 76.53%, yield: 86.42%, as a dark gray solid).
Preparation of intermediate 7
Step 1 6-bromo-1H-pyrazolopyridine (4.5 g,22.73mmol,1 eq) was dissolved in DMF (40 mL), replaced with argon, naH (60 wt%) (254 mg,27.27mmol,1.2 eq) was added under argon at-5℃for 10min, SEM-Cl (4.5 g,27.27mmol,1.2 eq) was added dropwise at-5℃and the ice bath was removed, and the reaction was resumed at room temperature for 3H. The reaction was checked for success by LCMS. Pouring the reaction solution into ice water, extracting with ethyl acetate for 3 times, washing with sodium chloride for 1 time, drying with anhydrous sodium sulfate, and purifying by spin drying and passing through a column to obtain the product 6-bromo-1- (2-trimethylsilylethoxy) methyl-1H-pyrazolo [4,3-b ] pyridine (4.3 g, purity: 97.50%, yield: 56.20%, as light yellow liquid).
Step 2 6-bromo-1- (2-trimethylsilylethoxy) methyl-1H-pyrazolo [4,3-b ] pyridine (4.3 g,13.098mmol,1 eq), pin 2B2 (3.991 g, 15.7198 mmol,1.2 eq), KOAc (2.571 g,26.196mmol,2 eq) were dissolved in1, 4-dioxane (100 mL), replaced with argon, pd (dppf) Cl 2 (479 mg,0.655mmol,0.05 eq) was added, and the mixture was stirred for reaction at 120℃for 12 hours under argon protection. The reaction was checked by LCMS and was successful. After the reaction solution was cooled, dichloromethane was extracted with water, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, and dried by spinning to obtain intermediate 7 (5.9 g, purity: 62.90%, yield: 98.68%).
Preparation of intermediate 8
Step 1 6-chloro-1H-pyrazolo [3,4-b ] pyrazine (2 g,13mmol,1 eq) was dissolved in DMF (50 mL), ice-salt bath (-5 ℃) NaH (0.67 g,16.9mmol,1.3eq,60 wt%) was added in portions, stirred for 30 min, SEM-Cl (2.6 g,15.6mmol,1.2 eq) was added dropwise, argon protected, the reaction was allowed to return to room temperature for 3H, quenched with ice water (30 mL), extracted three times with ethyl acetate (50 mL. Times.3), split, dried over anhydrous sodium sulfate, and filtered to dryness to give the product 6-chloro-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazoline [3,4-b ] pyrazine (2.3 g, crude) as a brown oily product. The product was used directly in the next reaction.
Step 2 6-chloro-1- ((2- (trimethylsilyl) ethoxy) methyl) -1 h-pyrazoline [3,4-b ] pyrazine (1.5 g,5.3mmol,1 eq) was dissolved in 1, 4-dioxane (50 mL), replaced three times with Pin2B2(1.74g,6.84mmol,1.3eq),Pd(dppf)Cl2)(384mg,0.53mmol,0.1eq),KOAc(1.56g,15.9mmol,3eq), argon, and the oil bath was warmed to 120℃for 12 hours. The reaction solution was cooled, diluted with water (30 mL), extracted three times with ethyl acetate (50 mL. Times.3), separated, dried over anhydrous sodium sulfate, and filtered to dryness to give intermediate 8 (1.8 g, crude product) as a brown oil. Directly used for the next reaction .1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.45(s,1H),5.78(s,2H),3.95–3.90(m,2H),1.03(dd,J=14.1,6.0Hz,3H),0.08(d,J=6.4Hz,12H),-0.00(s,9H).
Preparation of intermediate 9
5-Bromo-2-methylpyridine (1 g,5.81mmol,1 eq), pin 2B2 (1.771 g,6.98mmol,1.2 eq), KOAc (1.140 g,11.63mmol,2 eq) were dissolved in 1, 4-dioxane (20 mL), replaced with argon, pd (dppf) Cl 2 (425 mg, 0.81 mmol,0.05 eq), replaced with argon, and stirred for 12 hours under argon protection at 120 ℃. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to water (100 mL), and extracted with methylene chloride (200 mL) 3 times. The collected organic phase was dried over anhydrous sodium sulfate and spin-dried to give intermediate 9 (2 g, crude).
Preparation of intermediate 10
4-Bromo-2-methylpyridine (1.5 g,8.7mmol,1 eq) was dissolved in 1, 4-dioxane (20 mL), replaced three times with Pin2B2(2.88g,11.3mmol,1.3eq),Pd(dppf)Cl2(631mg,0.87mmol),KOAc(2.13g,21.7mmol), argon, and the oil bath was warmed to 120℃for 12 hours. The reaction solution was cooled, diluted with water (30 mL), extracted three times with ethyl acetate (50 ml×3), separated, dried over anhydrous sodium sulfate, and filtered to dryness to give intermediate 10 (3.2 g, crude product) as a brown oily product. The product was used directly in the next reaction. LCMS: MS m/z (ESI): 138.1[ M+H ] +.
Preparation of intermediates 11a and 11b
5-Bromo-2-cyclopropylpyridine (1 g,5.049mmol,1 eq), pin 2B2 (1.539 g,6.059mmol,1.2 eq), KOAc (991 mg,10.098mmol,2 eq) were dissolved in1, 4-dioxane (20 mL) and replaced with argon. Pd (dppf) Cl 2 (369 mg,0.505mmol,0.1 eq) was further added, replaced with argon and stirred for 12 hours at 120℃under argon protection. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to 200mL of water, extracted with methylene chloride (200 mL) and extracted 3 times. The collected organic phase was dried over anhydrous sodium sulfate, and dried by spin to obtain a mixture (2 g, crude) of intermediates 11a and 11 b.
Preparation of intermediate 12
5-Chloro-2-trifluoromethylpyridine (1 g,5.508mmol,1 eq), pin 2B2 (1.678 g,6.610mmol,1.2 eq), KOAc (1.081 g,11.016mmol,2 eq) were dissolved in1, 4-dioxane (20 mL) and replaced with argon. Pd (dppf) Cl 2 (403 mg,0.5508mmol,0.1 eq) was further added, replaced with argon and stirred for 5 hours at 120℃under argon protection. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to 200mL of water, extracted with methylene chloride (200 mL) and extracted 3 times. The collected organic phase was dried over anhydrous sodium sulfate, dried by spin, and purified by reverse phase column to give intermediate 12 (480 mg,2.51mmol, yield: 45.64%).
Preparation of intermediate 13
7-Bromo-1, 4-dihydroisoquinolin-3 (2H) -one (500 mg,2.2 mmol) was dissolved in 1, 4-dioxane (25 mL), and then subjected to reaction at 120℃for 12 hours under the protection of Pin2B2(620mg,2.4mmol),Pd(dppf)Cl2(80mg,0.11mmol),KOAc(755mg,7.7mmol),dppf(60mg,0.11mmol), argon. The reaction was allowed to stand at room temperature, filtered and concentrated under reduced pressure to give 700mg of product intermediate 13 as a brown oil (0.7 g, crude). Can be directly put into the next step. LCMS: MS m/z (ESI): 274.2[ M+H ] +.
Preparation of intermediate 14
4-Chloro-2-trifluoromethylpyridine (1 g,5.52mmol,1 eq) was placed in a 100mL clean three-necked flask, pin 2B2 (2.8 g,11.00mmol,2 eq) was added sequentially at room temperature, KOAc (1.6 g,16.56mmol,3 eq) and Pd (dppf) Cl 2 (0.6 g,0.83mmoL,0.15 eq) was dissolved by adding 1, 4-dioxane (20 mL), and stirred at 120℃for 12 hours. Cooled to room temperature, 20mL of water was added to the reaction solution, extraction was performed three times with ethyl acetate (30 ml×3), the organic phase was washed with saturated sodium chloride solution, the solution was separated, and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. Crude intermediate 14 (2.1 g, crude) was obtained as a black color. The product was used directly in the next reaction. LCMS: MS m/z (ESI): 274.0[ M+H ] +.
Preparation of intermediate 15
5-Chloro-2-fluoropyridine (1.0 g,7.63mmol,1 eq) was placed in a 100mL clean three-necked flask, pin 2B2 (3.88 g,15.3mmol,2 eq), KOAc (2.24 g,22.9mmol,3 eq) and Pd (dppf) Cl 2 (0.83 g,1.14mmoL,0.15 eq) were added sequentially at room temperature, dissolved by adding 1, 4-dioxane (20 mL), and stirred at 120℃for 12 hours. Cooled to room temperature, 20mL of water was added to the reaction solution, extraction was performed three times with ethyl acetate (30 ml×3), the organic phase was washed with saturated sodium chloride solution, the solution was separated, and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. Intermediate 15 (2.3 g, crude) was obtained as a black oil. The product was used directly in the next reaction. LCMS: MS m/z (ESI): 224.1[ M+H ] +.
Preparation of intermediate 16
Step 1, placing raw material 4-bromopyridine (2 g,12.6mmol,1 eq) into a 250mL single-port bottle, adding 100mL THF for dissolution, protecting by argon, cooling to-78 ℃, starting to dropwise add cyclopropylmagnesium bromide (40 mL,25.2mmol,2 eq), keeping the temperature at-78 ℃, stirring for 5 minutes after dropwise adding, starting to dropwise add phenyl chloroformate (2.4 mL,12.6mmol,1 eq), reacting for 10 minutes after dropwise adding, recovering room temperature, diluting the reaction solution with ethyl acetate (150 mL), respectively washing with water (30 mL), 0.1M (mol/L) hydrochloric acid (30 mL), respectively washing with brine (30 mL), and drying the organic phase by spin-drying to obtain 2.1g of crude product. The crude product was placed in a 250mL single-necked flask, 80mL of toluene was added for dissolution, DDQ (2.9 g,12.6mmol,1 eq) was added, and stirred at room temperature overnight. LCMS samples were taken to check complete reaction of the starting material, quenched with 1M NaOH (40 mL), ph=7, separated, aqueous ethyl acetate (50×3) extracted three times, combined organic phases, washed with brine, separated, dried over anhydrous sodium sulfate, spun-dried and purified over normal phase column (PE: ea=7:1) to give 2-cyclopropyl-4-bromopyridine (1.5 g, yield: 86%). LCMS: MS m/z (ESI): 198.0[ M+H ] +.
Step 2-raw material 2-cyclopropyl-4-bromopyridine (1.5 g,7.5mmol, 1 eq), pin 2B2 (2.3 g,9mmol, 1.2 eq), potassium acetate (1.5 g,15mmol,2 eq) were placed in a 250mL three-necked flask, 75mL1, 4-dioxane was added, argon was substituted, pd (dppf) Cl 2 (280 mg,0.39mmol,0.05 eq) was added, argon was substituted, stirring was raised to 120℃and reacted for 12 hours, LCMS sampling detection, LCMS showed complete reaction of raw material, and product LCMS did not show. The reaction was taken up in 50mL of water, 100mL of ethyl acetate, the layers separated, the aqueous phase extracted three times with EA, the organic phases combined, washed with brine, the layers separated and the organic phases dried to desolventize. The product was used directly for the next step.
EXAMPLE 1 preparation of Compound S1
Intermediate 1 (200 mg,0.51 mmol) and 3-pyridineboronic acid (76 mg,0.61 mmol) were dissolved in 1, 4-dioxane (20 mL) and water (4 mL), and then [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (42 mg,0.05 mmol) and potassium carbonate (142 mg,1.02 mmol) were added and the reaction stirred under nitrogen at 100℃for 2 hours. The reaction solution was cooled to room temperature, filtered, the filtrate was extracted three times with ethyl acetate and water, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give Compound S1 (38.03 mg, yield) :17.1%).LCMS:MS m/z(ESI):434.1[M+H]+.1H NMR(400MHz,DMSO-d6)δ12.23(s,1H),9.07(s,1H),8.66(d,J=4.8Hz,1H),8.32(d,J=7.9Hz,1H),7.94(s,1H),7.54(dd,J=7.9,4.8Hz,1H),4.70(s,2H),3.62–3.56(m,1H),2.66(s,3H),2.17(s,3H),1.29(d,J=6.8Hz,3H),1.17–1.11(m,1H),0.62–0.55(m,1H),0.44–0.35(m,2H),0.24(dd,J=8.7,3.5Hz,1H).
EXAMPLE 2 preparation of Compound S2
Intermediate 1 (120 mg,0.30 mmol) and 5-boronic acid pyrimidine (77 mg,0.36 mmol) were dissolved in 1, 4-dioxane (10 mL) and water (2 mL), and then [1,1' -bis (diphenylphosphine) ferrocene ] palladium dichloride dichloromethane complex (26 mg,0.03 mmol) and potassium carbonate (85 mg,0.61 mmol) were added and the reaction stirred under nitrogen at 100℃for 2 hours. The reaction solution was cooled to room temperature, filtered, the filtrate was extracted three times with ethyl acetate and water, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give Compound S2 (31.48 mg, yield) :23.4%).LCMS:MS m/z(ESI):435.2[M+H]+.1H NMR(400MHz,DMSO-d6)δ12.25(s,1H),9.28(d,J=1.8Hz,3H),7.99(s,1H),4.72(s,2H),3.63–3.57(m,1H),2.66(s,3H),2.17(s,3H),1.30(d,J=6.8Hz,3H),1.15(s,1H),0.58(s,1H),0.45–0.36(m,2H),0.25(d,J=5.4Hz,1H).
EXAMPLE 3 preparation of Compound S3
Intermediate 1 (100 mg,0.26mmol,1 eq), intermediate 2 (199mg, 0.77mmol,3 eq), K 2CO3 (71 mg,0.51mmol,2 eq) were dissolved in 1, 4-dioxane (20 mL) and water (4 mL), replaced with argon, pd (dppf) Cl 2 (19 mg,0.03mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred for 2 hours at 100℃under argon. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL), extracted with methylene chloride (30 mL. Times.3), and the combined organic phases were washed with saturated sodium chloride water and dried over anhydrous sodium sulfate. Purification by spin-drying gave compound S3 (18.11 mg,0.037mmol, yield) :14%).LCMS:MS m/z(ESI):488.1[M+H]+.1H NMR(400MHz,DMSO-d6)δ12.18(s,1H),10.48(s,1H),7.85(s,1H),7.51(d,J=7.7Hz,1H),7.41(s,1H),7.30(d,J=7.7Hz,1H),4.65(s,2H),3.59(dd,J=15.7,6.9Hz,1H),2.64(s,3H),2.16(s,3H),1.26(t,J=9.3Hz,3H),1.13(dd,J=8.3,4.4Hz,1H),0.57(dd,J=8.2,4.5Hz,1H),0.47–0.31(m,2H),0.28–0.16(m,1H).
EXAMPLE 4 preparation of Compound S4
Intermediate 1 (300 mg,0.77mmol,1 eq), intermediate 3 (597 mg,2.30mmol,3 eq), K 2CO3 (212 mg,1.53mmol,2 eq) were dissolved in 1, 4-dioxane (20 mL) and water (4 mL), replaced with argon, pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred at 100℃for 2 hours under argon protection. The reaction was successfully detected by LCMS. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were washed once with saturated sodium chloride solution (20 mL) and dried over anhydrous sodium sulfate. Purification by spin-drying gave compound S4 (3.85 mg,0.0079mmol, yield) :1.03%).LCMS:MS m/z(ESI):488.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ12.22(s,1H),8.63(s,1H),8.21(s,1H),8.14(dd,J=7.9,1.5Hz,1H),7.91(s,1H),7.68(d,J=7.9Hz,1H),4.69(s,2H),4.46(s,2H),3.64–3.49(m,1H),2.66(s,3H),2.16(s,3H),1.29(d,J=6.8Hz,3H),1.18–1.05(m,1H),0.59(d,J=8.5Hz,1H),0.47–0.33(m,2H),0.24(d,J=5.1Hz,1H).
EXAMPLE 5 preparation of Compound S5
Intermediate 1 (1.4 g,3.58mmol,1 eq), intermediate 4 (1.261 g,8.95mmol,2.5 eq), K 2CO3 (99mg, 7.16mmol,2 eq), dissolved in 1, 4-dioxane (30 mL) and water (6 mL), were replaced with argon, pd (dppf) Cl 2 (131 mg, 0.178 mmol,0.05 eq) was added and stirred for 2 hours under argon protection. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were washed once with saturated sodium chloride solution (20 mL) and dried over anhydrous sodium sulfate. Spin-drying, and purifying to give compound S5 (1175 mg,2.60mmol, yield) :73%).LCMS:MS m/z(ESI):452.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ12.23(s,1H),8.37(d,J=5.1Hz,1H),7.96(s,1H),7.89(d,J=4.6Hz,1H),7.74(s,1H),4.69(s,2H),3.65–3.54(m,1H),2.63(s,3H),2.16(s,3H),1.28(d,J=6.7Hz,3H),1.17–1.08(m,1H),0.58(d,J=3.9Hz,1H),0.40(dd,J=14.4,7.5Hz,2H),0.25(d,J=5.0Hz,1H).
EXAMPLE 6 preparation of Compound S6
Step 1 intermediate 1 (400 mg,1.02mmol,1 eq), intermediate 5 (900 mg,3.07mmol,3 eq), K 2CO3 (283 mg,2.05mmol,2 eq) were dissolved in1, 4-dioxane (20 mL) and water (4 mL), replaced with argon, pd (dppf) Cl 2 (75 mg,0.10mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred at 100℃for 2 hours under argon protection. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were washed once with saturated sodium chloride solution (20 mL) and dried over anhydrous sodium sulfate. Spin-drying yielded 1.2g of crude compound S6-1 (1.2 g, purity: 73%).
Step 2 Compound S6-1 (1.2 g,1.45mmol,1 eq) was dissolved in dichloromethane (50 mL), trifluoroacetic acid (9.4 g,82.7mmol,57 eq) was added and reacted at room temperature under argon atmosphere for 12 hours. K 2CO3 (1.0 g,7.25mmol,5 eq), methanol (50 mL), water (10 mL) were added and stirred for 12 hours. Dichloromethane (80 ml×3) was extracted. The combined organic phases were washed once with saturated sodium chloride solution (20 mL) and dried over anhydrous sodium sulfate. Spin-drying, and purification by passing to give compound S6 (16.31 mg,0.034mmol, yield) :2.37%).LCMS:MS m/z(ESI):474.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ13.78(s,1H),12.21(s,1H),8.33(d,J=8.2Hz,1H),8.23(s,1H),8.00(s,1H),7.51(d,J=8.2Hz,1H),4.71(s,2H),3.56–3.47(m,1H),2.65(s,3H),2.15(s,3H),1.27(d,J=6.8Hz,3H),1.13(d,J=8.6Hz,1H),0.62–0.14(m,4H).
EXAMPLE 7 preparation of Compound S7
Step 1-intermediate 1 (300 mg,0.77mmol,1 eq), intermediate 6 (673 mg,2.30mmol,3 eq), K 2CO3 (212 mg,1.53mmol,2 eq) were dissolved in1, 4-dioxane (20 mL), then distilled water of dropwort (4 mL) was added, replaced with argon, pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred for 2 hours at 100℃under argon protection. The reaction was checked by LCMS and was successful. After the reaction solution was cooled, dichloromethane was extracted with water, washed with saturated sodium chloride, and dried over anhydrous sodium sulfate. The crude product of the compound S7-1 (453 mg, purity: 76.59%, yield: 74.99%) was obtained by spin-drying purification.
Step 2 Compound S7-1 (453 mg, purity: 76.59%,0.58mmol,1 eq) was dissolved in dichloromethane (50 mL), trifluoroacetic acid (3.7 g,32.81mmol,57 eq) was added and reacted at room temperature under argon atmosphere for 10 hours. After the completion of the reaction, the pH was adjusted to neutral, the mixture was dried by spinning, and then K 2CO3 (398 mg,2.88mmol,5 eq), 50mL of methanol and 10mL of dropwort distilled water were added to carry out the reaction for 2 hours. After the reaction, dichloromethane and water are extracted, sodium chloride is washed with water, anhydrous sodium sulfate is dried, and the mixture is sent to be prepared and purified to obtain a compound S7 (26.90 mg,0.057mmol, yield) :32.76%).LCMS:MS m/z(ESI):473.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ12.20(s,1H),11.84(s,1H),8.04(d,J=8.1Hz,1H),7.93(s,1H),7.57(s,1H),7.42(d,J=8.1Hz,1H),6.53(s,1H),4.69(s,2H),3.52(s,1H),2.65(s,3H),2.15(s,3H),1.27(d,J=6.8Hz,3H),1.12(s,1H),0.60–0.19(m,4H).
EXAMPLE 8 preparation of Compound S8
Step 1-intermediate 1 (300 mg,0.77mmol,1 eq), intermediate 7 (432 mg,1.15mmol,1.5 eq), K 2CO3 (212 mg,1.53mmol,2 eq) were dissolved in1, 4-dioxane (20 mL), then distilled water of dropwort (4 mL) was added, replaced with argon, pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was added, replaced with argon, and the reaction was stirred for 2 hours at 100℃under argon protection. The reaction was checked by LCMS and was successful. After the reaction solution was cooled, dichloromethane was extracted with water, washed with saturated sodium chloride, and dried over anhydrous sodium sulfate. The crude product of compound S8-1 (500 mg, purity: 74.49%, yield: 80.37%) was obtained by spin-drying.
Step 2 Compound S8-1 (500 mg, purity: 74.49%, 0.611 mmol,1 eq) was dissolved in dichloromethane (50 mL), trifluoroacetic acid (4.0 g,35.16mmol,57 eq) was added and reacted at room temperature under argon atmosphere for 10 hours. After the completion of the reaction, the pH was adjusted to neutral, the mixture was dried by spinning, and then K 2CO3 (426 mg,3.08mmol,5 eq), 50mL of methanol and 10mL of dropwort distilled water were added to carry out the reaction for 2 hours. After the reaction, dichloromethane and water are extracted, sodium chloride is washed with water, anhydrous sodium sulfate is dried, and the mixture is sent to be prepared and purified to obtain a compound S8 (118.15 mg, 0.247 mmol, yield) :40.45%).LCMS:MS m/z(ESI):474.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ13.61(s,1H),12.27(s,1H),9.03(d,J=1.7Hz,1H),8.64(s,1H),8.38(s,1H),7.96(s,1H),4.72(s,2H),3.61(dd,J=9.0,6.8Hz,1H),2.67(s,3H),2.17(s,3H),1.30(d,J=6.8Hz,3H),1.16(dd,J=8.4,4.3Hz,1H),0.59(dt,J=8.5,5.1Hz,1H),0.46–0.36(m,2H),0.25(dt,J=8.5,4.2Hz,1H).
EXAMPLE 9 preparation of Compound S9
Step 1 intermediate 1 (500 mg,1.28mmol,1 eq) was dissolved in1, 4-dioxane (25 mL), water (5 mL), intermediate 8 (1.5 g,5.1mmol,4 eq) was added, pd (dppf) Cl 2 (94.2 mg,0.13mmol,0.1 eq), sodium carbonate (1.25 g,0.77mmol,3 eq), argon was added and the reaction was stirred at 100℃for 2 hours. LCMS monitored product formation. The reaction solution was cooled, diluted with water (30 mL), extracted three times with methylene chloride (30 mL. Times.3) and separated, and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude compound S9-1 (1 g). LCMS: MS m/z (ESI): 605.3[ M+H ] +.
Step 2 Compound S9-1 (1 g, crude) was dissolved in DCM (35 mL) and trifluoroacetic acid (5 mL) was added and reacted at room temperature for 10 hours. The reaction mixture was concentrated to dryness, 25mL of methanol and 5mL of water were added to dissolve the crude product, potassium carbonate (1.37 g,10mmol,5 eq) was added, stirred at room temperature for 2 hours, filtered, the filter cake was rinsed with a small amount of methanol, and the solvent was concentrated under reduced pressure. Dissolving the crude product with small amount of dimethylformamide, separating (0.1% NH 3.H2 O) for purification, and lyophilizing to obtain compound S9 (127.21 mg, yield) :16.2%).LCMS:MS m/z(ESI):475.3[M+H]+.1H NMR(400MHz,DMSO-d6)δ14.20(s,1H),12.22(s,1H),8.92(s,1H),8.52(s,1H),8.02(s,1H),4.71(s,2H),3.58–3.42(m,1H),2.62(s,3H),2.12(s,3H),1.24(d,J=6.8Hz,3H),1.09(dq,J=8.3,4.2,3.6Hz,1H),0.53(dq,J=8.4,4.3Hz,1H),0.45–0.27(m,2H),0.18(dt,J=9.5,4.7Hz,1H).
EXAMPLE 10 preparation of Compound S10
Intermediate 1 (250 mg,0.64mmol,1 eq), intermediate 9 (263 mg,1.92mmol,3 eq), K 2CO3 (177 mg,1.28mmol,2 eq), dissolved in 1, 4-dioxane (20 mL), was replaced with water (4 mL), pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was added and replaced with argon under argon protection, and stirred for 2 hours at 100 ℃. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to 100mL of water, extracted with methylene chloride (100 mL) and extracted 3 times. The collected organic phase was dried over anhydrous sodium sulfate and spun dry. Purified compound S10 (65.49 mg,0.146mmol, yield) :22.88%).LCMS:MS m/z(ESI):448.2[M+H]+.1H NMR(400MHz,DMSO-d6))δ12.24(s,1H),8.94(d,J=2.1Hz,1H),8.21(dd,J=8.0,2.3Hz,1H),7.90(s,1H),7.38(d,J=8.1Hz,1H),4.68(s,2H),3.63–3.54(m,1H),2.65(s,3H),2.55(s,3H),2.17(s,3H),1.29(d,J=6.8Hz,3H),1.17–1.09(m,1H),0.61–0.22(m,4H).
EXAMPLE 11 preparation of Compound S11
Intermediate 1 (350 mg,0.89 mmol) was dissolved in 1, 4-dioxane (15 mL), water (3 mL), intermediate 10 (370 mg,2.68mmol,3 eq), pd (dppf) Cl 2 (64.5 mg,0.089mmol,0.1 eq), sodium carbonate (284.1 mg,2.68mmol,3 eq), argon shield, and stirred at 100℃for 3 hours. LCMS monitored product formation. The reaction solution was cooled, diluted with water (30 mL), extracted three times with methylene chloride (30 mL. Times.3), separated, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the crude product was dissolved in a small amount of dimethylformamide to give a product (preparation column: waters-SunFire C18, elution phase: 0.04vol% aqueous FA solution/acetonitrile, elution gradient: beginning 54vol% acetonitrile-ending 62vol% acetonitrile, flow rate: 30 mL/min), and Compound S11 (112.18 mg, purity) was isolated and purified :99.3%).LCMS:MS m/z(ESI):448.2[M+H]+.1HNMR(400MHz,DMSO-d6)δ12.24(s,1H),8.57(d,J=5.1Hz,1H),7.96(s,1H),7.74(s,1H),7.66(d,J=4.9Hz,1H),4.69(s,2H),3.66–3.51(m,1H),2.65(s,3H),2.56(s,3H),2.16(s,3H),1.28(d,J=6.8Hz,3H),1.16–1.03(m,1H),0.57(t,J=4.1Hz,1H),0.49–0.32(m,2H),0.24(d,J=5.1Hz,1H).
EXAMPLE 12 preparation of Compound S12
Intermediate 1 (250 mg,0.640mmol,1 eq), a mixture comprising intermediates 11a and 11b (314 mg,1.279mmol,2 eq), K 2CO3 (177 mg,1.28mmol,2 eq), dissolved in 1, 4-dioxane (20 mL), and replaced with water (4 mL) with argon. Pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was further added, replaced with argon and stirred at 100℃for 2 hours under argon protection. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to 100mL of water, extracted with methylene chloride (100 mL) and extracted 3 times. The collected organic phase was dried over anhydrous sodium sulfate and spun dry. Purification of the product was carried out to give Compound S12 (33.38 mg,0.070mmol, yield) :11.02%).LCMS:MS m/z(ESI):474.2[M+H]+.1H NMR(400MHz,DMSO-d6)δ12.23(s,1H),8.90(d,J=1.7Hz,1H),8.18(dd,J=8.1,2.2Hz,1H),7.89(s,1H),7.41(d,J=8.1Hz,1H),4.67(s,2H),3.63–3.54(m,1H),2.65(s,3H),2.20–2.14(m,4H),1.29(d,J=6.8Hz,3H),1.14(s,1H),1.01(d,J=6.4Hz,4H),0.61–0.23(m,4H).
EXAMPLE 13 preparation of Compound S13
Intermediate 1 (300 mg,0.767mmol,1 eq), intermediate 12 (176 mg,0.920mmol,1.2 eq), K 2CO3 (212 mg, 1.284 mmol,2 eq), dissolved in 1, 4-dioxane (20 mL), and replaced with water (4 mL) and argon. Pd (dppf) Cl 2 (56 mg,0.077mmol,0.1 eq) was further added and replaced with argon, and stirred at 100℃for 2 hours under argon protection. The reaction was checked for success by LCMS. After the reaction solution was cooled, the reaction solution was transferred to 100mL of water, extracted with methylene chloride (100 mL) and extracted 3 times. The collected organic phase was dried over anhydrous sodium sulfate and spun dry. Purified compound S13 (60.10 mg,0.120mmol, yield) :15.61%).LCMS:MS m/z(ESI):502.1[M+H]+.1H NMR(400MHz,DMSO-d6))δ12.24(s,1H),9.21(d,J=1.6Hz,1H),8.57(dd,J=8.1,1.7Hz,1H),8.07(d,J=8.1Hz,1H),8.00(s,1H),4.72(s,2H),3.59(dq,J=13.5,6.8Hz,1H),2.66(s,3H),2.17(s,3H),1.30(d,J=6.8Hz,3H),1.14(dt,J=13.2,4.8Hz,1H),0.63–0.55(m,1H),0.46–0.35(m,2H),0.28–0.22(m,1H).
EXAMPLE 14 preparation of Compound S14
Intermediate 1 (100 mg,0.25 mmol), intermediate 13 (103 mg,0.37 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (18.3 mg,0.025 mmol), cesium carbonate (244 mg,0.75 mmol) was dissolved in1, 4-dioxane (8 mL) and water (2 mL), and reacted for 2 hours in an oil bath at 100 ℃. LCMS detection, reaction was successful. After the reaction mixture was cooled, it was diluted with water (20 mL), extracted with methylene chloride (30 mL. Times.3), and the combined organic phases were washed with saturated sodium chloride water and dried over anhydrous sodium sulfate. Purification by spin drying gave compound S14 (2.4 mg, purity) :100%).LCMS:MS m/z(ESI):502.3[M+H]+.1H NMR(400MHz,DMSO-d6)δ12.21(s,1H),8.06(s,1H),7.87(s,1H),7.80(s,1H),7.77(d,J=7.9Hz,1H),7.31(d,J=7.9Hz,1H),4.66(s,2H),4.40(s,2H),3.62–3.54(m,1H),3.52(s,2H),2.64(s,3H),2.16(s,3H),1.28(d,J=6.8Hz,3H),0.85(s,1H),0.57(s,1H),0.48–0.31(m,2H),0.23(s,1H).
EXAMPLE 15 preparation of Compound S15
Intermediate 1 (100 mg,0.26 mmol), pyridin-4-ylboronic acid (62.9 g,0.51 mmol), a [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (18.3 mg,0.025 mmol), cesium carbonate (250 mg,0.77 mmol) was dissolved in 1, 4-dioxane (6 mL) and water (1.5 mL) and reacted for 2 hours under argon protection in an oil bath at 100 ℃. The reaction solution was cooled to room temperature, filtered, extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, and dried by spin-drying, and then purified to give Compound S15 (62.7 mg, purity) :99%).LCMS:MS m/z(ESI):434.1[M+H]+.1H NMR(400MHz,DMSO-d6)δ11.89(s,1H),8.72(d,J=5.9Hz,2H),7.97(s,1H),7.93–7.87(m,2H),4.70(s,2H),3.59(dq,J=13.8,6.9Hz,1H),2.65(s,3H),2.17(s,3H),1.29(d,J=6.8Hz,3H),1.18–1.06(m,1H),0.70–0.51(m,1H),0.50–0.31(m,2H),0.25(dd,J=11.4,6.3Hz,1H).
EXAMPLE 16 preparation of Compound S16
Intermediate 1 (0.3 g,0.77mmol,1 eq) was placed in a 50mL clean single-neck flask, 1, 4-dioxane (12 mL), water (3 mL) was added to dissolve, intermediate 14 (0.4 g,1.46mmol,1.9 eq), sodium carbonate (0.246 g,2.31mmol,3 eq) and Pd (dppf) Cl 2 (0.056 g,0.077mmol,0.1 eq) were added at room temperature, and the reaction was stirred for 2 hours under argon at 100 ℃. The reaction solution was cooled to room temperature, 20mL of water was added to the reaction solution, extraction was performed three times with ethyl acetate (30 mL. Times.3), the organic phase was washed with saturated sodium chloride solution, separated, and the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated, and isolated to give Compound S16 (86.73 mg, yield) :22.5%).LCMS:MS m/z(ESI):502.1[M+H]+.1HNMR(400MHz,DMSO-d6)δ12.26(s,1H),8.95(t,J=6.1Hz,1H),8.50(s,1H),8.27(d,J=5.0Hz,1H),8.02(s,1H),4.73(s,2H),3.62(dq,J=13.7,6.8Hz,1H),2.66(s,3H),2.17(s,3H),1.30(d,J=6.8Hz,3H),1.20–1.07(m,1H),0.68–0.54(m,1H),0.48–0.35(m,2H),0.32–0.18(m,1H).
EXAMPLE 17 preparation of Compound S17
Intermediate 1 (0.30 g,0.77mmol,1 eq) was placed in a 100mL clean single-neck flask, 1, 4-dioxane (12 mL), water (3 mL) was added to dissolve, intermediate 15 (0.33 g,1.46mmol,1.9 eq), sodium carbonate (0.25 g,2.31mmol,3 eq) and Pd (dppf) Cl 2 (0.056 g,0.077mmol,0.1 eq) were added at room temperature and the reaction was stirred for 2 hours at 100 ℃. The reaction solution was cooled to room temperature, 20mL of water was added to the reaction solution, extraction was performed three times with ethyl acetate (30 mL. Times.3), the organic phase was washed with saturated sodium chloride solution, separated, and the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated, and isolated to give Compound S17 (9.2 mg, purity) :99.7%).LCMS:MS m/z(ESI):452.1[M+H]+.1H NMR(400MHz,DMSO-d6)δ11.98(s,1H),8.52(s,1H),8.27(t,J=7.3Hz,1H),7.68(s,1H),7.09(d,J=7.6Hz,1H),4.45(s,2H),3.39–3.32(m,1H),2.40(s,3H),1.93(s,3H),1.05(d,J=6.4Hz,3H),0.90(s,1H),0.34(s,1H),0.16(d,J=6.9Hz,2H),0.01(d,J=4.9Hz,1H).
EXAMPLE 18 preparation of Compound S18
Compound 1 (400 mg,1.02mmol,1 eq), intermediate 16 (1.5 g,6.12mmol,6 eq), na 2CO3 (325 mg,3.06mmol,3 eq) were placed in a 100mL three-port bottle, 40mL 1, 4-dioxane, 8mL water, argon replacement, pd (dppf) Cl 2 (73 mg,0.1mmol,0.1 eq) argon replacement, stirred and warmed to 100℃for 2 hours, sampling and detection, the starting material reaction was complete, the reaction solution was added with 30mL water, 50mL ethyl acetate was layered, the aqueous phase was extracted three times with 20mL ethyl acetate, the organic phase was washed with brine and the layered, and the organic phase was dried and desolventized with anhydrous sodium sulfate and sent to give Compound S18 (110 mg,0.211mmol, pale yellow solid, yield) :22.88%).LCMS:MS m/z(ESI):474.2[M+H]+.1HNMR(400MHz,DMSO-d6)δ12.24(s,1H),8.51(d,J=4.8Hz,1H),7.96(s,1H),7.75(s,1H),7.59(dd,J=5.1,1.6Hz,1H),4.69(s,2H),3.60(dd,J=9.1,6.8Hz,1H),2.65(s,3H),2.16(s,3H),2.00(dd,J=14.9,7.1Hz,1H),1.29(d,J=6.8Hz,4H),1.16–1.05(m,1H),1.00(t,J=2.1Hz,1H),0.98(d,J=4.5Hz,2H),0.64–0.52(m,1H),0.46–0.33(m,2H),0.25(dd,J=9.1,5.0Hz,1H).
Preparation of Compound S-34 of comparative example 1
Step 1 Compound 1 (100 mg,0.256 mmol) was dissolved in N, N-dimethylformamide (3 mL), zinc cyanide (150 mg,1.28 mmol), 1' -bis (diphenylphosphine) ferrocene (56.7 mg,0.1 mmol), activated zinc powder (16.6 mg,0.256 mmol) were added to each solution, argon gas was blown into the reaction solution for 30 seconds, and tris (dibenzylideneacetone) dipalladium (46.6 mg,0.05 mmol) was added thereto, and argon gas was blown into the reaction solution for 1 minute. Microwave 125 ℃ and reaction for one hour. The reaction solution was allowed to stand at room temperature, filtered, and the cake was washed twice with dichloromethane. The filter cake was further slurried with dimethyl sulfoxide (5 mL) for 30 minutes, and filtered and pumped dry with an oil pump to give compound S-34-a (50 mg, gray solid, yield) :51%).1H-NMR(400MHz,DMSO-d6)δ12.92–11.68(m,1H),8.20(s,1H),4.71(s,2H),3.71–3.49(m,1H),2.63(s,3H),2.18(s,3H),1.31(d,J=6.8Hz,3H),1.24(s,1H),0.59(t,J=7.0Hz,1H),0.48–0.35(m,2H),0.27(d,J=4.9Hz,1H).
Step 2 Compound S-34-a (30 mg,0.079 mmol) was dissolved in toluene (2 mL), argon was replaced three times, and the reaction was carried out in an oil bath at 120℃for 36 hours. The reaction solution was allowed to stand at room temperature, filtered, and the cake was washed twice with dichloromethane. Dissolving the filter cake with dimethyl sulfoxide (5 mL), and preparing under alkaline [ NH 4HCO3 ] to obtain compound S-34 (1.21 mg, white solid, purity) 99%).1H-NMR(400MHz,DMSO-d6)δ12.29(s,1H),8.10(s,1H),5.33(t,J=4.9Hz,1H),4.82(s,2H),3.68–3.53(m,1H),2.69(s,3H),2.19(s,3H),1.33(d,J=6.8Hz,3H),0.86(t,J=6.7Hz,1H),0.61(t,J=6.8Hz,1H),0.44(d,J=5.4Hz,2H),0.28(d,J=5.5Hz,1H).
Test example 1 inhibition of Akt S473 phosphorylation levels in Raw264.7 cells
The experiment was performed by ELISA.
1. Material and instrument sources
Raw264.7 cells were purchased from ATCC, mC5a protein from R & D, DMEM serum-free medium from Gibco, fetal Bovine Serum (BSA) from Gibco, RIPA buffer from CST, protease phosphatase inhibitor from CST, ELISA detection kit for phosphorylated Akt S473 from R & D, 96 well plates from Falcon, high binding 96 well plates from Costar, INFINITE M1000 fluorescent microplate reader from Tecan.
2. Preparation of test Compound solutions
Powders of test compounds in Table 1 were formulated into 10mM DMSO solutions each with 100% DMSO, then 4. Mu.L was added to 196. Mu.L of 100% DMSO, and 3-fold gradient dilutions were made with 100% DMSO for a total of 8 concentration points. mu.L of each concentration of the test compound solution was added to 190. Mu.L of the serum-free medium.
Raw264.7 cell treatment
Raw264.7 cells (PI 3K gamma) in logarithmic growth phase (culture conditions: DMEM+10% FBS (v/v)), were plated in 96-well plates (Falcon 353072) at a concentration of 2.2X10 6/mL, cultured at 37℃under 5% CO 2, after overnight cell attachment, 10. Mu.L of DMSO solutions of the above-prepared test compounds at different concentrations (control well and blank well with DMSO only) were added, respectively, cultured at 37℃under 5% CO 2, cultured at 2 hours, 5. Mu.L of DMEM serum-free medium solution of mC5a protein (R & D2150-C5) (blank well with medium only) were added), incubated at room temperature for 5 minutes, the medium was removed, 100. Mu.L of 1 Xcell lysate (prepared from PA buffer (CST 9806S) and protease phosphatase inhibitor (CST 5872S) were added, and shaking at rpm for 1 minute to form control group, blank group and blank group, respectively, and stored at-800℃for use.
4. Detection step
Capture antibody (Capture) solution was prepared according to the instructions of ELISA detection kit (R & D DYC887 BE) for phosphorylating Akt S473, and 100. Mu.L per well of Capture solution was added to high binding 96-well plates (Costar 42592), and incubated overnight at room temperature. Washing three times with 300. Mu.L of PBST, adding blocking solution (PBS solution containing 1% (w/v) BSA) (GENVIEW FA 016-100G), incubating for 2 hours at room temperature, washing three times with 300. Mu.L of PBST, respectively adding 90. Mu.L of cell samples of control group, blank group and experimental group, incubating for 2 hours at room temperature, washing three times with 300. Mu.L of PBST, adding 100. Mu.L of anti-pAkt S473 diluent, incubating for 2 hours at room temperature, washing three times with 300. Mu.L of PBST, adding 100. Mu.L of Streptavidin (strepitavidin-HRP) diluent marked by horseradish peroxidase, incubating for 40 minutes at room temperature, washing three times with 300. Mu.L of PBST, adding 100. Mu.L of ELISA substrate Tetramethylbenzidine (TMB), incubating for 20 minutes at room temperature, and adding 50. Mu.L of 2N (mol/L) H 2SO4 solution to terminate the reaction. OD values were read on INFINITE M.sup.1000 fluorescence microplate reader (Tecan) with absorption wavelengths of 450nm and 570nm.
5. Data analysis
The blank group was DMSO-added without mC5a protein, and the control group was DMSO-added with mC5a protein.
Inhibition% = [1- (Total mean (experimental group) -Total mean (blank group))/(Total mean (control group) -Total mean (blank group)) ] ×100%, wherein Inhibition%: percent Inhibition of compounds against Akt S473 phosphorylation level of raw264.7 cells; total mean.
6. IC 50 values were calculated using XLFIT 5.0.0 software from the calculated inhibition and the test results are shown in table 1.
TABLE 1 Compounds inhibit IC 50 values for RAW264.7 cell Activity
Test example 2 inhibition of T47D and PC3 cell Akt S473 phosphorylation levels
The experiment was performed using INCELL ELISA method.
1. Material and instrument sources
T47D cells were purchased from ATCC, PC3 cells were purchased from SIBS, RPMI 1640 and F12K medium were purchased from Gibco, phosphorylated Akt S473 (Ser 473-p-Akt) antibody was purchased from CST, 96 well plates were purchased from Falcon, INFINITE M fluorescent microplate reader was purchased from Tecan, and YZJ-0673 was compound S-37 in patent WO 2016095833.
2. Preparation of test Compound solutions
Powders of test compounds in Table 2 were formulated as 10mM DMSO solutions each with 100% DMSO, then 20. Mu.L was added to 80. Mu.L of 100% DMSO, and 3-fold gradient dilutions were made with 100% DMSO for a total of 8 concentration points. mu.L of each concentration of the test compound solution was added to 190. Mu.L of the serum-free medium.
T47d and PC3 cell treatment
Cells of logarithmic phase T47D (PI 3K alpha) (culture conditions: RPMI 1640+10% FBS (v/v)) and PC3 (PI 3K beta) (culture conditions: F12K+10% FBS (v/v)) were plated in 96-well plates (Falcon 353072) at a concentration of 90. Mu.L for 2.2X10 5/mL (PC 3) or 4.4X10 5/mL (T47D), cultured overnight, after the cells had adhered, 10mL of DMSO solutions of the test compounds prepared as described above were added (control wells were added with DMSO only, background wells were added with 10. Mu.M YZJ-0673), cultured at 37℃and 5% CO 2, after 2 hours, 100. Mu.L of 4% paraformaldehyde (Biosharp BL 539A) was added, and after 45 minutes incubation at room temperature, 100. Mu.1% Triton 0.1% Triton-100 (v/v) was removed, and the incubation was continued for 30 minutes, to form control groups and experimental groups, respectively.
4. Detection step
The control, background and experimental groups were each prepared by removing Triton X-100 solution, washing twice with 200. Mu.L of PBS (300 rpm,1 min each), adding blocking solution (PBS solution containing 1% (w/v) BSA) (GENVIEW FA 016-100G), incubating at room temperature for 2 hours, removing, washing once with 200. Mu.L of PBS (300 rpm,1 min), adding 30. Mu.L of Ser473-p-Akt antibody 0.1% (w/v) BSA diluent (CELL SIGNALING 4060L), and incubating overnight at 4 ℃. The Ser473-p-Akt antibody was removed, washed twice with 200. Mu.L PBS (300 rpm for 1 min each), and 100. Mu.L horseradish peroxidase-labeled anti-rabbit immunoglobulin G (HRP anti-rabit IgG), 0.1% BSA dilution (w/v) (CST 7074S) were added and incubated for 1.5 hours at room temperature. The reaction was stopped by washing twice with 200. Mu.L of PBS (300 rpm,1 min each), adding 100. Mu.L of the ELISA substrate Tetramethylbenzidine (TMB), incubating at room temperature for 0.5 h, adding 50. Mu.L of 2N sulfuric acid, incubating at room temperature for 20min, and reading OD on a INFINITE M1000 fluorescence microplate reader (Tecan) at an absorption wavelength of 450nm.
The TMB solution was taken out, washed three times with 200. Mu.L of PBS (300 rpm each for 1 minute), added with 100. Mu.L of 0.1% Janus aqueous solution (w/v) (Abcam ab 111622), incubated at room temperature for 10 minutes, taken out, washed 5 times with 200. Mu.L of double water (300 rpm each for 1 minute), added with 100. Mu.L of 1.5M hydrochloric acid, incubated at room temperature for 10 minutes, and OD was read on a INFINITE M1000 fluorescence microplate reader (Tecan) at an absorption wavelength of 595nm.
5. Data analysis
The background group was 10. Mu.m YZJ-0673 and the control group was DMSO.
Inhibition% = [1- (Ratio (experimental group) -Ratio (background group))/(Ratio (control group) -Ratio (background group)) ] ×100%, where Inhibition% is the percentage of Inhibition of compounds on T47D and PC3 cell activity, ratio=od450/OD 595. IC 50 values were calculated using XLFIT 5.0.0 software from the calculated inhibition and the test results are shown in table 2.
Compounds of table 2 inhibit IC 50 values for T47D and PC3 cell activity
From tables 1 and 2, it can be seen that the compounds of the present application have higher inhibitory activity on the Akt S473 phosphorylation level of raw264.7 cells, and lower inhibitory activity on T47D and PC3 cells, indicating that the compounds of the present application have higher selective inhibitory activity on PI3kγ.
Test example 3 measurement of PI3K kinase inhibitory Activity
1. Sources of materials and instruments
Human pi3kγ and pi3kδ kinases were purchased from Millipore, human pi3kα kinase from Invitrogen, human pi3kβ kinase from Eurofins, ADP-Glo kinase assay kit from Promega, PIP2 lipid substrate from Life Technologies,384 well plates from PE.
2. Test method steps
The test uses ADP-Glo method to detect the inhibition of the activity of the compounds on PI3K gamma, PI3K alpha, PI3K beta and PI3K delta, and uses the reagent PI3Kγ(Millipore 14-558),PI3Kα(Invitrogen PV4788)、PI3Kβ(Eurofins 14-603M)、PI3Kδ(Millipore 14-604M)、PIP2(Life Technologies PR8982B),ADP-Glo(Promega V9102/3).
Inhibition of PI3K kinase activity by test compounds in table 3 test compounds were formulated with 100% DMSO as 100 x DMSO solutions at the maximum concentration required for the experiment, and then 3-fold gradient dilutions were performed with 100% DMSO for a total of 10 concentration points, by the following method. 50nL of the formulated test compound solution was added to 384 well plates (PE 6008289), with blank wells and control wells added with DMSO. 1 Xbuffer was prepared and 2 Xkinase enzyme dilutions (reaction concentration: PI3Kγ2.5. Mu.g/mL, PI3Kα 0.15. Mu.g/mL, PI3Kβ 0.3. Mu.g/mL, and PI3Kδ1.2. Mu.g/mL) and 2 Xsubstrate dilutions ATP (reaction concentration: 25. Mu.M)/PIP 2 were prepared with 1 Xbuffer, and 2.5. Mu.L of kinase dilution and 2.5. Mu.L of substrate dilution were added to each well, respectively, with blank wells containing 2.5. Mu.L of 1 Xbuffer and 2.5. Mu.L of substrate dilution. After shaking and mixing, incubation is carried out for 1 hour at 25 ℃, the reaction is stopped by adding 5 mu L of ADP-Glo reagent in the ADP-Glo kit, shaking and mixing are carried out for 2 hours at 25 ℃, kinase Detection Reagent mu L of ADP-Glo reagent in the ADP-Glo kit is added, shaking and mixing are carried out, incubation is carried out for 30 minutes at 25 ℃, a control group, a blank group and an experimental group are respectively formed, and the chemiluminescent intensity of each hole is measured by an Envision fluorescent enzyme-labeled instrument.
3. Data analysis
Inhibition% = [1- (average (experimental group) -average (blank group))/(average (control group) -average (blank group)) ] ×100%
2 Parallel wells were provided for each test compound at different concentrations. The IC 50 values of the inhibitory activity of the compounds of the application on the 4 isoforms of PI3K kinase are shown in Table 3 and Table 4.
TABLE 3 Compounds inhibit IC 50 values for PI3K gamma kinase Activity
TABLE 4 Compounds inhibit IC 50 values for PI3K alpha, PI3K beta and PI3K delta kinase Activity
From tables 3 and 4, it can be seen that the compounds of the present application have higher inhibitory activity on pi3kγ kinase and lower inhibitory activity on pi3kα, pi3kβ and pi3kδ kinase, indicating that the compounds of the present application have higher selective inhibitory activity on pi3kγ.
Test example 4 in vivo drug substitution test in mice
The LC/MS method was used to determine the drug concentration in plasma at different times after intravenous administration and oral gavage administration of the compound of the present application, and the pharmacokinetic behavior of the compound of the present application in mice was studied using the known compound D1 and the compounds S11, S5 and S18 of the present application as examples, and the pharmacokinetic characteristics were evaluated.
The structure of compound D1 (CAS No. 2504036-13-7) is shown below, and can be prepared by referring to the prior art or obtained by commercial purchase.
1. Material and instrument sources
Adult male ICR mice were supplied by Peking Vitolith laboratory animal technologies Co., ltd., polyethylene glycol 15-hydroxystearate (Solutol HS 15) from Sigma under the accession number 42966, sulfobutyl-beta-cyclodextrin (Captisol) from Sigma-ALDRICH under the accession number 332607, the anticoagulant ethylenediamine tetraacetic acid dipotassium salt (EDTA-K 2) from Sigma under the accession number V900157, dimethyl sulfoxide from Sigma under the accession number D5879, dexamethasone (brand NIFDC, lot number 6TUC-T4C 2).
2. Experimental protocol:
2.1 test animals healthy adult male ICR mice (25-40 g body weight, 6 groups each, free drinking and eating by intravenous group mice; overnight fast by gavage administration group, free drinking and eating after 4h administration);
2.2 dosing mode and dosage, selecting animals meeting experimental requirements (quarantine qualification, adaptation period through laboratory and weight meeting requirements) before dosing, and weighing and marking. ICR mice were dosed intravenously at 2mg/kg, vehicle 5% DMSO+30% (10% Solutol HS15) +65% (20%Captisol,pH 7.4), v/v/v, and lavage at 10mg/kg, vehicle 5% DMSO+30% (10% Solutol HS15) +65% (20%Captisol,pH 7.4), v/v/v. The compound is weighed and dissolved in a solvent according to the concentration of 0.4mg/mL (intravenous injection group) or 1mg/mL (intragastric administration group), the administration amount of the compound is calculated according to the weight conversion of 5mL/kg (intravenous injection group) or 10mL/kg (intragastric administration group), namely, the weight of the mice is 25g after weighing, the intravenous administration amount of the compound is 25/1000kg multiplied by 0.4mg/mL multiplied by 5 mL/kg=0.05 mg, and the intragastric administration amount of the compound is calculated by the like.
2.3 Blood sample collection binding mice prior to blood sample collection, each of the dosed mice was bled at predetermined time points (intravenous administration: 0.083,0.25,0.5,1,2,4,7,24h post-administration, respectively, for 8 time points; intragastric administration: 0.083,0.25,0.5,1,2,4,7,24h post-administration, respectively, for 8 time points), by blood collection through the canthus vein of about 100. Mu.L. Whole blood was transferred to a 1.5mL tube with pre-addition of anticoagulant EDTA-K 2, centrifuged for 6min (8000 rpm,4 ℃) to separate plasma, and the whole process was completed within 15min after blood collection. All samples were stored in a-20 ℃ refrigerator until the samples were analyzed.
2.4 Analysis of plasma samples by liquid chromatography-tandem mass spectrometry (model: triple Quad TM 4000) under chromatographic conditions of a column of Waters XB ridge-C18 (2.1X105 mm,3.5 μm), mobile phase A of an aqueous solution (v/v) containing 0.05% formic acid and 5mmol/L ammonium acetate, mobile phase B of a methanol solution (v/v) containing 0.05% formic acid and 5mmol/L ammonium acetate, flow rate of 0.6mL/min, column temperature of 40 ℃, sample introduction amount of 2. Mu.L, liquid phase gradient:
the mass spectrum conditions are scanning mode, multi-reaction ion monitoring (positive ion mode);
Ion source, turbine spray, ionization mode, electrospray ionization.
2.5 Preparation of a Standard Curve the test compound was weighed into the appropriate powder and dimethyl sulfoxide was added at a concentration of 2mg/mL to give a stock solution for the test. Stock solutions were formulated into standard curve working solutions at concentrations of 20ng/mL,40ng/mL,100ng/mL,400ng/mL,2000ng/mL,10000ng/mL,34000ng/mL,40000ng/mL and quality control sample working solutions at concentrations of 60ng/mL,16000ng/mL,30000ng/mL, respectively, using pure acetonitrile. The standard curve working solution and the quality control sample working solution are diluted 20 times by using a blank ICR mouse plasma matrix, and low, medium and high concentration quality control samples of which the concentration of the compound to be tested in the blank ICR mouse plasma matrix is 1.00ng/mL,2.00ng/mL,5.00ng/mL,20.0ng/mL,100ng/mL,500ng/mL,1700ng/mL,2000ng/mL and 3ng/mL,800ng/mL and 1500ng/mL are prepared.
2.6 Processing biological samples, namely placing frozen plasma samples on wet ice for thawing, and placing on a vortex instrument for vortex for 5min after the samples are thawed. Respectively taking 20 mu L of plasma sample, standard curve and quality control instruction sample, adding into a 96-well plate, adding 200 mu L of acetonitrile solution containing internal standard dexamethasone (with the concentration of 2000 ng/mL), precipitating protein, mixing for 5min by vortex, centrifuging at 3700rpm and 4 ℃ for 15min, taking supernatant, centrifuging again under the same condition, and finally carrying out LC-MS/MS analysis by adding 2 mu L of supernatant solution. The parameters of the results and the oral bioavailability F are shown in Table 5.
Table 5 in vivo Pharmacokinetic (PK) parameters in compound mice
Note that in table 5, "C max" is the peak concentration, "AUC 0-t" is the area under the blood concentration-time curve from 0 to the final quantifiable time point, and "F" is the oral bioavailability.
As can be seen from table 5, both the exposure (AUC 0-t) and bioavailability (F) of compounds S11, S18 and compound S5 were higher than compound D1. The compound of the application has more excellent in vivo drug generation parameters.
Test example 5 pharmacodynamic test in tumor model of subcutaneous transplantation of mouse colon cancer cell MC38 and in vitro differentiated M2 macrophage isotype
1. Purpose of experiment
The antitumor effect of the compounds of the present application in a tumor model of subcutaneous transplantation of mouse colon cancer cell MC38 in combination with in vitro cultured macrophage isotype was evaluated.
2. Test object preparation
2.1 Solvent preparation
5% DMSO (dimethyl sulfoxide) (v/v) +40% PEG400 (v/v) +55% pure water (v/v), and the administration volume is 10. Mu.L/g.
2.2 Preparation of S11 Compounds for administration
Weighing a proper amount of compound S11 into a centrifuge tube, adding a solvent, and swirling to obtain a full-solution, wherein the concentration is 1mg/mL, and preparing twice a week.
2.3 Preparation of compound D1 administration preparation
An appropriate amount of compound D1 (CAS number 2504036-13-7, see test example 4 for structure) was weighed into a centrifuge tube, added with solvent, and vortexed to a total solution at a concentration of 1mg/mL.
3. Experimental animal
C57BL/6 mice, females, 8-9 weeks (the week of mice at tumor cell inoculation), body weight 18.0-20.0g, 8 per group. Purchased from Shanghai Ji Hui laboratory animal feeding Co.
4. Cell MC38 culture of colon cancer of mice
Mouse colon cancer cells MC38 (purchased from Shanghai transportation university) were cultured in RPMI-1640 medium (Gibco; 61870-036) containing 10% (v/v) fetal bovine serum (Gibco; 10099-141C), 100U/mL penicillin and 100pg/mL streptomycin (Gibco; 15140122). MC38 cells in exponential growth phase were collected, and PBS was resuspended to a suitable concentration for subcutaneous tumor inoculation in mice.
5. In vitro culture of mouse macrophages
Bone marrow cells are separated from the leg bones of C57BL/6 mice to prepare single cell suspension, 50ng/mL M-CSF (Peprotech; 315-02) is added to induce the single cell suspension into M0 macrophages derived from bone marrow, 20ng/mL interleukin 4 (IL 4) (Peprotech; 214-14) is added in the induction process of the M0 macrophages to culture for 48 hours to obtain polarized M2 macrophages, and the in vitro cultured M0 macrophages and M2 macrophages are respectively mixed with a colon cancer cell line MC38 of the mice according to the ratio of 1:1 (number), and inoculated subcutaneously, wherein the total number of the M0 macrophages and the M2 macrophages is 10 6 cells/mouse.
6. Animal inoculation group and administration
6.1 Animal Vaccination group
8 Female C57BL/6 mice were randomly selected and inoculated with 5X 10 5 MC38 cells subcutaneously on their backs as control groups, 8 female C57BL/6 mice were randomly selected and inoculated with 5X 10 5 MC38 cells+5X 10 5 M0 macrophages subcutaneously on their backs as M0 groups, 24 female C57BL/6 mice were randomly selected and inoculated with 5X 10 5 MC38 cells+5X 10 5 M2 macrophages subcutaneously on their backs (tumor cells and M2 macrophages differentiated in vitro) as M2 groups, and the method of setting up the tumor model was referred to as Nature.2016;539 (7629): 437-442.) (day of inoculation was defined as day 0, and dosing was started on day 1.
6.2 Modes of administration and dosage
The experiment adopts a mode of gastric lavage administration, wherein a control group of mice respectively irrigates a gastric solvent as a solvent group, the administration volume is 10 mu L/g, an M0 group of mice respectively irrigates a gastric solvent as a solvent group mixed with M0 macrophages, the administration volume is 10 mu L/g, 8M 2 groups of mice respectively irrigates a gastric solvent as a solvent group mixed with M2 macrophages, the administration volume is 10 mu L/g, 8M 2 groups respectively irrigate a 1mg/mL compound S11 administration preparation as a compound S11 group mixed with M2 macrophages, the administration dosage is 10mg/kg, the rest M2 groups of mice respectively irrigate a 1mg/mL compound D1 administration preparation as a compound D1 group mixed with M2 macrophages, and the administration dosage is 10mg/kg.
7. Post-dosing observation of animals
Conventional monitoring includes observing the effects of tumor growth and treatment on normal animal behavior, including, in particular, the activity of experimental animals, feeding and drinking conditions, weight gain or loss, eyes, hair and other anomalies. Tumor volume was weighed and measured 2 times per week for a dosing period of 27 days, tumor mass weighing (TW) was sacrificed after weighing tumor volume at day 27, tumor Volume (TV), relative tumor weight increase rate (T/C), and tumor growth inhibition rate (tumor weight) (TGI) were calculated, and statistical measurements were made. T/C (%) = T/c×100%, TGI (%) = (1-T/C) ×100%, and T and C are the relative tumor volumes or weights of a treatment group (compound S11 group mixed with M2 macrophages or compound D1 group mixed with M2 macrophages) and a control group (vehicle group), respectively, at a specific time point.
8. Results
During the experiment, the tumor growth curves of the mice are shown in FIG. 1. In FIG. 1, vehicle group, M0+ vehicle group, M2+ compound D1 group, and M2 macrophage group, respectively, with 10mg/kg (mpk), administration by stomach irrigation, administration once daily, and M2+ compound S11 group, administration by stomach irrigation, administration once daily, respectively, with M2 macrophage group, compound S11 group, 10mg/kg, administration by stomach irrigation, administration once daily. * P <0.01, P <0.001, ns, P >0.05. (n=8, mean ± standard error).
Tumor gravimetric analysis of mice in each group on day 27 of dosing model is shown in figure 2. In FIG. 2, vehicle group, M0+ vehicle group, M2+ compound D1 group, and M2 macrophage group, respectively, were administered by intragastric administration at a dose of 10mg/kg and at a dose of once daily, and M2+ compound S11 group, respectively, was administered by intragastric administration at a dose of once daily, wherein M2+ compound S11 group, and M2 macrophage group were administered by intragastric administration at a dose of once daily. * P <0.01; ns, P >0.05. (n=8, mean ± standard error).
The pharmacodynamic parameters of each group in the model at day 27 after dosing are shown in table 6 below:
table 6 table of pharmacodynamic analyses of groups in mice MC38 and macrophage mixed vaccination model at day 27 post-dose
Note that in Table 6, "T/C" indicates the relative tumor weight increase rate, "TGI" indicates the tumor growth inhibition rate (tumor weight), and "\" indicates no corresponding parameter.
As can be seen from fig. 1,2 and table 6, the compound S11 of the present embodiment showed a remarkable tumor growth inhibition effect in the mouse MC38 and macrophage mixed inoculation model. The compound has more excellent tumor growth inhibition effect.
All documents mentioned in this disclosure are incorporated by reference in this disclosure as if each were individually incorporated by reference. Further, it will be appreciated that various changes and modifications may be made by those skilled in the art after reading the above teachings, and such equivalents are intended to fall within the scope of the application as defined in the appended claims.
The technical features of the above-described embodiments may be arbitrarily combined, and all possible combinations of the technical features in the above-described embodiments are not described for brevity of description, however, as long as there is no contradiction between the combinations of the technical features, they should be considered as the scope of the description.
The above examples illustrate only a few embodiments of the application, which are described in detail and are not to be construed as limiting the scope of the claims. It should be noted that it will be apparent to those skilled in the art that several variations and modifications can be made without departing from the spirit of the application, which are all within the scope of the application. Accordingly, the scope of protection of the present application is to be determined by the appended claims.
Claims (15)
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2022110625850 | 2022-09-01 |
Publications (1)
Publication Number | Publication Date |
---|---|
CN119768404A true CN119768404A (en) | 2025-04-04 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN110088099B (en) | Amine-substituted heterocyclic compounds as EHMT2 inhibitors and methods of use thereof | |
ES2957233T3 (en) | Activin receptor-like kinase inhibitors | |
CN110117284B (en) | Nitrogen-containing heterocyclic compound and preparation method and application thereof | |
TW202214605A (en) | Immunomodulators, compositions and methods thereof | |
IL268366B2 (en) | Inhibitors of viral replication | |
CN110267954B (en) | Aniline substituted 1, 2-dihydropyrrolo [3,4-c ] pyridine/pyrimidine-3-one derivatives and application thereof | |
CN102858175A (en) | Triazolones as fatty acid synthase inhibitors | |
CN107921044B (en) | GLS1 inhibitors for the treatment of disease | |
CN113412263B (en) | Azabicyclo-substituted oxaspiro derivatives, preparation method and medical application thereof | |
CN113646050A (en) | Thieno [3,2-B ] pyrrolo [3,2-D ] pyridazinone derivatives and their use as PKM2 derivatives for the treatment of cancer, obesity and diabetes related disorders | |
CN114269751B (en) | Optically pure oxaspiro-substituted pyrrolopyrazoles derivatives, process for their preparation and their use in medicine | |
JP2023501799A (en) | ROCK INHIBITOR AND PRODUCTION METHOD AND USE THEREOF | |
EP3350185A1 (en) | 1-phenylpyrrolidin-2-one derivatives as perk inhibitors | |
WO2023165551A1 (en) | Six-membered aromatic ring-pyrrolidone derivative, and pharmaceutical composition thereof and use thereof | |
KR20200097294A (en) | 1,2,4-oxadiazole derivatives as histone deacetylase 6 inhibitors | |
CN115991706A (en) | PIM kinase inhibitors | |
EP4358954A1 (en) | Cdk2 inhibitors and methods of using the same | |
WO2020094156A1 (en) | Diheterocycle-substituted pyridine-2(1h)-ketone derivative, preparation method therefore and pharmaceutical use thereof | |
JP7599039B2 (en) | Heterocyclic substituted ketone derivatives, compositions thereof and medicinal uses thereof | |
WO2017046739A1 (en) | Imidazolidinone derivatives as inhibitors of perk | |
TW202214634A (en) | Heterocyclic compound and derivative thereof | |
CN110256446B (en) | Benzo-heterocycle substituted cyclopenta [4,5] pyrrolopyrazine-1-one derivatives and application thereof | |
CN114085220B (en) | Substituted morpholine-4-carboxylic acid ester derivatives, compositions and pharmaceutical uses thereof | |
CN119768404A (en) | Heteroaryl-substituted pyridopyrrolone derivatives, pharmaceutical compositions and applications thereof | |
WO2024046454A1 (en) | Heteroaryl-substituted pyridopyrrolidone derivative, and pharmaceutical composition and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication |