CN116003397B - Benzo-polycyclic thiazoline amide compound and application thereof - Google Patents

Benzo-polycyclic thiazoline amide compound and application thereof Download PDF

Info

Publication number
CN116003397B
CN116003397B CN202310294139.0A CN202310294139A CN116003397B CN 116003397 B CN116003397 B CN 116003397B CN 202310294139 A CN202310294139 A CN 202310294139A CN 116003397 B CN116003397 B CN 116003397B
Authority
CN
China
Prior art keywords
compound
replaced
equivalent
ring
thiazoline
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.)
Active
Application number
CN202310294139.0A
Other languages
Chinese (zh)
Other versions
CN116003397A (en
Inventor
施晶晶
龚林培
张正
杨生生
潘华玲
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd
Kaisi Kaixu Shanghai Pharmaceutical Technology Co ltd
Original Assignee
Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd
Kaisi Kaixu Shanghai Pharmaceutical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd, Kaisi Kaixu Shanghai Pharmaceutical Technology Co ltd filed Critical Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd
Priority to CN202310294139.0A priority Critical patent/CN116003397B/en
Publication of CN116003397A publication Critical patent/CN116003397A/en
Application granted granted Critical
Publication of CN116003397B publication Critical patent/CN116003397B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a benzo-multi-ring thiazoline amide compound and application thereof, wherein the benzo-multi-ring thiazoline amide compound has a structure shown in the following formula I:
Figure ZY_1
the method comprises the steps of carrying out a first treatment on the surface of the Wherein the ring A is selected from any one of C5-C8 cyclodiene, C4-C8 epoxydiene or C6-C10 aromatic ring; the B ring is selected from any one of substituted or unsubstituted C6-C12 aryl, substituted or unsubstituted C4-C12 nitrogen-containing heteroaryl and substituted or unsubstituted C4-C12 oxygen-containing heteroaryl; r is R 1 、R 2 Independently selected from any one of hydrogen, substituted or unsubstituted C1-C10 straight or branched chain alkyl, substituted or unsubstituted C7-C10 arylalkyl, substituted acyl and substituted sulfonyl. The benzo-polycyclic thiazoline amide compound provided by the invention can effectively inhibit the activities of COUP-TFII and NRF2, and has an excellent inhibition effect on the proliferation of prostate cancer cells.

Description

Benzo-polycyclic thiazoline amide compound and application thereof
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a benzo-multi-ring thiazoline amide compound and application thereof, in particular to a benzo-multi-ring thiazoline amide compound with high activity and application thereof.
Background
And regulating the growth and development of the organism and the cell differentiation, thereby affecting various physiological and metabolic processes in the body. Chicken ovalbumin upstream promoter transcription factor 2 (chicken ovalbumin upstream promoter transcription factor 2, coup-TFII) is one of the important members of the nuclear receptor family. The COUP-TF family includes two highly homologous subtypes, COUP-TFI and COUP-TFII, located on chromosomes 5 and 15, respectively, also referred to as nuclear receptor 2 families 1 and 2 (NR 2F1 and NR2F 2). COUP-TF is currently considered to be an orphan nuclear receptor because no ligand has been identified for its specific binding.
Current studies indicate that COUP-TFI plays an important role in neural development and COUP-TFII plays an important role in organ development, respectively. Further research shows that COUP-TFII plays an important role in various aspects of the physiological growth and development process of the organism, can regulate various signal paths, and participates in controlling tumor growth, angiogenesis, regeneration of organism tissues or cells and the like.
It is worth mentioning that the expression of COUP-TFII is abnormally up-regulated in the states of embryo development stage, tissue regeneration, diseases and the like; whereas in normal tissue or body maturation stage, COUP-TFII expression is maintained at a lower level. The current research result also shows that the COUP-TFII gene is knocked out from adult mice, and the mice do not show abnormal physiological phenomena such as phenotype change and the like. This demonstrates to some extent that inhibition of COUP-TFII does not produce toxic side effects. Therefore, small molecule drugs which target binding and inhibit COUP-TFII activity are developed, i.e. have practical and potential feasibility.
COUP-TFII is a necessary condition for angiogenesis during tumor growth, and its abnormal expression can lead to the occurrence and metastasis of cancer. Therefore, COUP-TFII is an ideal completely new target for potential cancer treatment.
NRF2 (nuclear factor E rythroid 2 related factor, nuclear factor E2 related factor) was cloned in 1994 from Moi et al as a factor binding to the nuclear factor E2 repeat of the beta globin gene promoter. Belongs to the CNC family of transcription factors, and contains a leucine zipper structure.
The NRF2/Keap1 (Kelch-like ECH-associated protein 1, kelch-like epichlorohydrin-related protein 1) -ARE (antioxidant response element ) signaling pathway is an endogenous antioxidant signaling pathway of the organism discovered in recent years.
Activation of the NRF2/Keap 1-ARE pathway is one of the important mechanisms for anti-tumorigenesis. Activation of NRF2 enables cancer cells to adapt to adverse microenvironments and high endogenous ROS levels during early stages of tumorigenesis; whereas in rapidly proliferating cells, NRF2 can support intermediary metabolism by promoting nucleotide and amino acid biosynthesis. Studies have shown that low activity NRF2 promotes tumorigenesis, while sustained high activity NRF2 can accelerate cancer progression and tolerance to treatment.
Therefore, the development of the compound which can inhibit COUP-TFII and NRF2 has very important practical value and wide application prospect in tumor treatment.
Disclosure of Invention
Aiming at the defects of the prior art, the invention aims to provide a benzo-multi-ring thiazoline amide compound and application thereof, in particular to a benzo-multi-ring thiazoline amide compound with high activity and application thereof. The benzo-polycyclic thiazoline amide compound provided by the invention can effectively inhibit the activities of COUP-TFII and NRF2, and has an excellent inhibition effect on the proliferation of prostate cancer cells.
In order to achieve the aim of the invention, the invention adopts the following technical scheme:
in a first aspect, the invention provides a benzo-multi-ring thiazoline amide compound, which has a structure shown in the following formula I:
Figure SMS_1
wherein the ring A is selected from any one of C5-C8 cyclodiene, C4-C8 epoxydiene or C6-C10 aromatic ring.
The B ring is selected from any one of substituted or unsubstituted C6-C12 aryl, substituted or unsubstituted C4-C12 nitrogen-containing heteroaryl and substituted or unsubstituted C4-C12 oxygen-containing heteroaryl.
R 1 、R 2 Independently selected from any one of hydrogen, substituted or unsubstituted C1-C10 straight or branched chain alkyl, substituted or unsubstituted C7-C10 arylalkyl, substituted acyl and substituted sulfonyl.
The benzo multi-ring thiazoline amide compound with the specific structure can effectively inhibit the activities of COUP-TFII and NRF2, and has excellent inhibition effect on the proliferation of prostate cancer cells.
Preferably, the substituents of the C6-C12 aryl, C4-C12 nitrogen-containing heteroaryl or C4-C12 oxygen-containing heteroaryl are independently selected from hydroxy, halogen, unsubstituted or R a Substituted C1-C10 straight-chain or branched alkyl, unsubstituted or R a Substituted C1-C10 alkoxy, unsubstituted or R b Substituted C6-C12 aryl, unsubstituted or R b Any one of substituted C5-C12 nitrogen-containing heteroaryl groups.
The R is a Selected from halogen or deuterium, said R b Selected from the group consisting of C1-C10 straight or branched alkyl, C1-C10 haloalkyl, C1-C10 alkoxy,
Figure SMS_2
or-COOR c Any one of the above, R c Any one selected from hydrogen, C1-C10 straight-chain alkyl or branched-chain alkyl, C1-C10 alkoxy, C1-C10 alkylamino or C1-C10 heterocyclic group.
Figure SMS_3
Representing the attachment site of the group.
Preferably, the substituents of the linear or branched alkyl groups are selected from halogen, hydrogen or deuterium.
Preferably, the acyl, sulfonyl, C1-C10 straight or branched alkyl, C7-C10 arylalkyl substituents are independently selected from C1-C10 straight or branched alkyl, C1-C10 alkoxy, C1-C10 haloalkyl, C3-C10 cycloalkyl, unsubstituted or R d Substituted C6-C12 aryl, unsubstituted or R d Any of the substituted C5-C12 heteroaryl groups; the R is d Any one of halogen, cyano, C1-C10 straight-chain alkyl or branched-chain alkyl.
Preferably, the A ring is selected from benzene rings or
Figure SMS_4
X is selected from-CH 2 -or-O-.
n is selected from 0, 1 or 2, and n is 0, where no group is present and X is directly attached to the other end.
Wherein, the liquid crystal display device comprises a liquid crystal display device,
Figure SMS_5
represents the attachment site of the fused ring structure.
Preferably, the B ring is selected from
Figure SMS_6
Any one of the following.
Wherein Y is selected from-ch=or-n=, and Z' is selected from-O-or-NH-.
R 3 、R 4 Independently selected from the group consisting of-H, -OH, -OMe, -OCD 3 、-F、-Cl、-CF 3
Figure SMS_7
Any one of the following.
Z is selected from-CH=or-N=.
R e Selected from-H, -CF 3 、-COOR f Or (b)
Figure SMS_8
Any one of the following.
The R is f Selected from the group consisting of-H, -Me, -Et, -i-Pr, -n-Bu, -i-Bu, -CH 2 -CHEt 2 、-(CH 2 ) 2 -OMe、-(CH 2 ) 2 -NMe 2 Or (b)
Figure SMS_9
Any one of them; />
Figure SMS_10
Representing the attachment site of the group.
Preferably, said R 1 、R 2 Independently selected from-H, -Me, -CD 3 、-Et、-i-Pr、-Bn、
Figure SMS_11
Any one of the following.
Wherein R is g Selected from-Me or MeOCH 2 -,R h Selected from-Me, -Et, -n-Pr, -i-Bu, -CF 3 、-Bn、
Figure SMS_12
Or->
Figure SMS_13
Any one of them; r is R i Selected from any one of-H, -Me, -F or-CN.
Figure SMS_14
Representing the attachment site of the group.
Preferably, the benzo-polycyclic thiazoline amide compound is selected from any one of the following formulas 1 to 58:
Figure SMS_15
Figure SMS_16
Figure SMS_17
Figure SMS_18
Figure SMS_19
Figure SMS_20
Figure SMS_21
Figure SMS_22
illustratively, the compounds of formula I may be prepared by the steps of:
Figure SMS_23
the initial compound I-1 is subjected to two-step substitution to obtain a compound III, then a ring is closed to obtain an intermediate M, and the intermediate M is subjected to condensation to obtain the compound shown in the formula I.
In a second aspect, the present invention provides stereoisomers of the benzomulti-ring thiazoline amides described above, pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same.
Preferably, the pharmaceutical composition further comprises pharmaceutically acceptable pharmaceutical excipients.
In a third aspect, the invention provides the use of a benzomulti-ring thiazoline amide compound as described above or a stereoisomer of a benzomulti-ring thiazoline amide compound as described above, a pharmaceutically acceptable salt thereof, a pharmaceutical composition comprising the same, for the manufacture of a medicament for the treatment and/or prevention of a disease associated with excessive activity of COUP-TFII or overexpression of COUP-TFII.
Preferably, the invention provides the use of a benzo-multi-ring thiazoline amide compound as described above or a stereoisomer of a benzo-multi-ring thiazoline amide compound as described above, a pharmaceutically acceptable salt thereof, a pharmaceutical composition comprising the same, for the preparation of a COUP-TFII inhibitor for the purpose of diagnosis or treatment of non-diseases.
Preferably, the disease associated with excessive COUP-TFII activity or COUP-TFII overexpression comprises prostate cancer.
In a fourth aspect, the present invention also provides the use of a benzomulti-ring thiazoline amide compound as described above or a stereoisomer of a benzomulti-ring thiazoline amide compound as described above, a pharmaceutically acceptable salt thereof, a pharmaceutical composition comprising the same, for the preparation of a medicament for the treatment and/or prevention of a disease associated with NRF2 over-activity or NRF2 over-expression.
Preferably, the diseases related to the excessive activity of NRF2 or the over expression of NRF2 comprise liver diseases such as NASH and drug liver injury, respiratory diseases such as lung injury and pulmonary fibrosis, and malignant tumors such as lung cancer, pancreatic cancer, liver cancer, colorectal cancer and prostate cancer.
Compared with the prior art, the invention has the following beneficial effects:
The invention provides a benzo multi-ring thiazoline amide compound with a specific structure, which can effectively inhibit the activities of COUP-TFII and NRF2 and has an excellent inhibition effect on the proliferation of prostate cancer cells.
Detailed Description
In order to further describe the technical means adopted by the present invention and the effects thereof, the following describes the technical scheme of the present invention in combination with the preferred embodiments of the present invention, but the present invention is not limited to the scope of the embodiments.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used herein in the description of the invention is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention.
The term "pharmaceutically acceptable salt" means that the compound can be converted by conventional means into the corresponding salt, which is chemically or physically compatible with the other ingredients comprising the pharmaceutical dosage form, and physiologically compatible with the recipient. The salts may be acid and/or base salts of the compounds with inorganic and/or organic acids and/or with inorganic and/or organic bases, and also include zwitterionic salts (inner salts) and also include quaternary ammonium salts, such as alkylammonium salts. These salts may be obtained directly in the final isolation and purification of the compounds. Or by appropriately mixing the compound of the present invention or a stereoisomer or solvate thereof with a certain amount of an acid or a base. These salts may be obtained by precipitation in solution and collected by filtration, or recovered after evaporation of the solvent, or by reaction in an aqueous medium and then cooled and dried. In particular, the salt is preferably a water-soluble pharmaceutically acceptable non-toxic acid addition salt, examples being salts of amino groups with inorganic acids (such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid) or with organic acids (such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid), or by using other methods conventional in the art (e.g. ion exchange methods).
The term "alkyl" refers to a saturated hydrocarbon containing primary (positive) carbon atoms, or secondary carbon atoms, or tertiary carbon atoms, or quaternary carbon atoms, or a combination thereof. The phrase containing the term, for example, "C1-C3 alkyl" refers to an alkyl group containing 1 to 3 carbon atoms, which may be, for each occurrence, independently of each other, C1 alkyl, C2 alkyl, C3 alkyl. Suitable examples include, but are not limited to: methyl (Me, -CH) 3 ) Ethyl (Et, -CH) 2 CH 3 ) 1-propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ) 2-propyl (i-Pr, i-propyl, -CH (CH) 3 ) 2 )。
The term "heterocycloalkyl" refers to a non-aromatic cyclic group in which one or more of the atoms making up the ring is a heteroatom and the remainder is carbon, including but not limited to nitrogen, oxygen, sulfur, and the like. Preferred heterocycloalkyl groups are 3-10 membered saturated heterocycloalkyl groups. Unless specifically indicated otherwise in this specification, heterocycloalkyl groups may be monocyclic ("monocyclic heterocycloalkyl"), or bicyclic, tricyclic or more ring systems which may include a fused, bridged or spiro ring system (e.g., a bicyclic system ("bicyclic heterocycloalkyl"). Heterocycloalkyl bicyclic ring system may include one or more heteroatoms in one or both rings, and saturated.
The term "alkoxy" refers to a group having an-O-alkyl group, i.e. an alkyl group as defined above, attached to the parent core structure via an oxygen atom. The phrase containing the term, for example, "C1-C3 alkoxy" means that the alkyl moiety contains 1 to 3 carbon atoms.
The term "aryl" refers to an aromatic hydrocarbon radical derived from the removal of one hydrogen atom on the basis of an aromatic ring compound, which may be a monocyclic aryl radical, or a fused ring aryl radical, or a polycyclic aryl radical, at least one of which is an aromatic ring system for a polycyclic species. Preferred aryl groups are 6-10 membered aryl groups, which may be selected from phenyl and naphthyl, as examples.
The term "heteroaryl" refers to an aryl group containing heteroatoms, which may be a single ring or a fused ring, independently selected from N, O and S, preferably 5-12 membered heteroaryl groups, including but not limited to pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, quinolinyl, isoquinolinyl, triazolyl, tetrahydropyrrolyl. In one embodiment, a 5-6 membered monocyclic heteroaryl group typically containing 1 or more heteroatoms independently selected from N, O and S. Exemplary 5-membered heteroaryl groups containing one heteroatom, such as "5-membered heteroaryl" unless otherwise specified, include, but are not limited to, pyrrolyl, furanyl, and thienyl; exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolinyl, isoxazolyl, thiazolyl, and isothiazolyl; exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, thiadiazolyl; exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl.
In the various parts of the invention, linking substituents are described. When the structure clearly requires a linking group, the markush variables recited for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for that variable enumerates an "alkyl" or "aryl" group, it will be understood that the "alkyl" or "aryl" represents a linked alkylene group or arylene group, respectively. In some specific structures, when an alkyl group is explicitly represented as a linking group, then the alkyl group represents a linked alkylene group, e.g., the alkyl in the group "-C1-C3 haloalkyl" is to be understood as alkylene.
Furthermore, the term "comprising" is an open-ended limitation and does not exclude other aspects, i.e. it includes the content indicated by the invention.
Unless otherwise indicated, the present invention employs conventional methods of mass spectrometry, nuclear magnetism, and the like to identify compounds, and the procedures and conditions may be referred to procedures and conditions conventional in the art.
As will be appreciated by those skilled in the art, the present application describes "as used in the structural formula of a group" in accordance with convention used in the art "
Figure SMS_24
By "is meant that the corresponding group is attached to other fragments, groups in the compound through that site. The above preferred conditions can be arbitrarily combined on the basis of not deviating from the common knowledge in the art, and thus, each preferred embodiment of the present invention can be obtained.
The terms "halo", "halogen" refer to halogen atoms or are substituted with halogen atoms including fluorine, chlorine, bromine or iodine atoms.
The term "cyclodiene" refers to a cycloalkane having two double bonds, such as cyclopentadiene, cyclohexadiene, cycloheptadiene, or cyclononadiene; the term "epoxydiene" refers to a structure in which at least one carbon atom is replaced by an oxygen atom on the basis of the term "cyclodiene".
The reagents and materials used in the present invention are commercially available.
Intermediate M-synthesis of a:
Figure SMS_25
step M-1: compound I-a (8.0 g,49.6 mmol) and pyridine (4.31 g,54.56 mmol) were added to dichloromethane (20 mL), methanesulfonyl chloride (5.68 g,49.6 mmol) was slowly added under ice-bath conditions, and then stirred overnight at 20 ℃. After the reaction, adding 1.0M sodium hydroxide solution to adjust the pH value of the system to be between 11 and 12, extracting with dichloromethane and water, adjusting the pH value of the obtained water phase part to be between 5 and 6 with 1.0M hydrochloric acid, filtering and drying to obtain a yellow solid compound II-a (9.7 g, yield 82%). LCMS [ M+H ] + = 240.1。
Step M-2: a mixed solution of methyl iodide (6.9 g,48.7 mmol) and N, N-dimethylformamide (10 mL) was slowly added to N, N-dimethylformamide (20 mL) with ice bath, followed by stirring overnight at 20 ℃. After the reaction, the reaction solution was added to a sufficient amount of water and stirring was continued for 30 minutes, and then filtered and dried to obtain a yellow solid compound III-a (10.1. 10.1 g, yield 99%). LCMS [ M+H] + = 254.1。
Step M-3: compound III-a (3.0 g,11.86 mmol) and elemental iodine (4.5 g,17.79 mmol) were added to ethanol (20 mL), 95 o Stirring for 30 min under C, then adding thiourea (1.80 g,23.72 mmol), 95 o Stirring overnight under the condition C, adding 1.0M sodium hydroxide solution to quench the reaction after the reaction is finished, regulating the pH value of the system to 10-11, adding water (20 mL), stirring at 20 ℃ for 30 minutes, filtering, and drying to obtain a coffee solid intermediate M-a (2.38 g, yield 64%). LCMS [ M+H] + = 310.1。
Example 1:
Figure SMS_26
the synthetic route is as follows:
Figure SMS_27
intermediate M-a (92.7 mg,0.3 mmol), starting compound 1-a (60 mg,0.3 mmol), N, N-diisopropylethylamine (116 mg, 0.9 mmol) and 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (171 mg,0.45 mmol) were added to N, N-dimethylformamide (1.5 mL) and stirred overnight at 20 ℃. After the completion of the reaction, compound 1 was purified by reverse phase column chromatography to give yellow solid (27, mg, yield 18%). LCMS [ M+H ] + = 491.1。
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.71 (s, 1H), 8.26-8.21 (m, 4H), 8.09 (d, J = 6.0 Hz, 1H), 7.92 (t, J = 6.0 Hz, 1H), 7.72 (d, J = 6.0 Hz, 1H), 7.41 (t, J = 6.0 Hz, 1H), 7.33-7.30 (m, 2H), 3.23 (s, 3H), 3.02-2.98 (m, 4 H), 2.95 (s, 3H)。
Example 2:
Figure SMS_28
the synthetic route is as follows:
Figure SMS_29
intermediate M-a (927 mg,3 mmol), starting compound 2-a (600 mg,3 mmol), N, N-diisopropylethylamine (1.16 g,9 mmol) and 2- (7-azobenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (1.71 g,4.5 mmol) were added to N, N-dimethylformamide (15 mL), followed by stirring overnight at 20 ℃. After the completion of the reaction, the compound 2-b was purified by reverse phase column chromatography to give a pink solid (840, mg, yield 57%). LCMS [ M+H] + = 493.1。
1H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 8.03 (d, J = 6.0 Hz, 2H), 7.74 (d, J = 6.0 Hz, 2H), 7.70 (d, J = 6.0 Hz, 1H), 7.31 (d, J = 6.0 Hz, 2H), 3.22 (s, 3H), 3.02-2.97 (m, 4H), 2.95 (s, 3H).
Compound 2-b (150 mg,0.3 mmol), compound 2-c (74 mg,0.36 mmol), [1,1' -bis (diphenylphosphine) ferrocene]Palladium dichloride dichloromethane complex (25 mg,0.03 mmol) and sodium carbonate (63.6 mg,0.6 mmol) were added to a mixed solvent of 1, 4-dioxane (5 mL) and water (0.5 mL), and the mixture was heated to 100℃under nitrogen to react overnight. After the completion of the reaction, ethyl acetate (20 mL) was added to dilute, filter and extract, and the organic phase was concentrated and purified by column chromatography to give compound 2 as a yellow solid (104, mg, yield 70%). LCMS [ M+H] + = 491.1。
1H NMR (400 MHz, DMSO-d6) δ 12.83 (s, 1H), 8.68-8.66 (m, 2H), 8.25 (d, J = 6.0 Hz, 2H), 7.98 (d, J = 6.0 Hz, 2H), 7.81-7.79 (m, 2H), 7.72 (d, J = 6.0 Hz, 1H), 7.32 (d, J = 6.0 Hz, 2H), 3.23 (s, 3H), 3.04-2.98 (m, 4 H), 2.96 (s, 3H)。
Example 3:
Figure SMS_30
the synthetic route is as follows:
Figure SMS_31
referring to example 2, compound 2-c was replaced with an equivalent amount of compound 3-a, and compound 3 was synthesized as a yellow solid (27, mg, yield 18%). LCMS [ M+H ] + = 491.1。
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.99 (s, 1H), 8.62-8.61 (m, 1H), 8.24 (d, J = 6.0 Hz, 2H), 8.18 (d, J = 6.0 Hz, 1H), 7.92 (d, J = 6.0 Hz, 2H), 7.72 (d, J = 6.0 Hz, 1H), 7.53-7.50 (m, 1H), 7.32 (d, J = 6.0 Hz, 2H), 3.23 (s, 3H), 3.03-2.98 (m, 4 H), 2.96 (s, 3H)。
Example 4:
Figure SMS_32
the synthetic route is as follows:
Figure SMS_33
compound 2 (20 mg,0.04 mmol) and 2, 3-dichloro-5, 6-dicyanobenzoquinone (14 mg,0.06 mmol) were added to 1, 2-dichloroethane (3 mL), and the mixture was heated to 60℃under nitrogen to react overnight. After the completion of the reaction, N-dimethylformamide (5 mL) was added thereto for dilution, and the mixture was purified by reverse phase column chromatography to give compound 4 (12, 12 mg, yield 61%) as a yellow solid. LCMS [ M+H] + = 489.1。
1H NMR (400 MHz, DMSO-d6) δ 12.22 (s, 1H), 8.69 - 8.66 (m, 2H), 8.58 (d, J = 6.8 Hz, 1H), 8.32 (d, J = 6.0 Hz, 2H), 8.14 (d, J = 6.8 Hz, 1H), 8.09 (s, 1H), 8.02 (d, J = 6.0 Hz, 2H), 7.86 (d, J = 6.8 Hz, 1H), 7.82 (d, J = 4.4 Hz, 2H), 7.74 - 7.71 (m, 1 H), 3.36 (s, 3H), 3.01 (s, 3H)。
Example 5:
Figure SMS_34
the synthetic route is as follows:
Figure SMS_35
referring to example 2, the compound 2-a was replaced with an equivalent of 5-a and the compound 2-c was replaced with an equivalent of 3-a, to thereby obtain a yellow solid compound 5 (100 mg, yield 91%). LCMS [ M+H] + =521.1。
1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 9.02 (s, 1H), 8.63 (d, J = 3.3 Hz, 1H), 8.20 (d, J = 6.0 Hz, 1H), 7.83 (d, J = 6.0 Hz, 1H), 7.69 (d, J = 6.0 Hz, 1H), 7.55 - 7.52 (m, 2H), 7.45 (d, J = 6.0 Hz, 1H), 7.32 - 7.30 (m, 2H), 4.06 (s, 3H), 3.24 (s, 3H), 3.03 - 2.99 (m, 4H), 2.96 (s, 3H)。
Example 6:
Figure SMS_36
the synthetic route is as follows:
Figure SMS_37
compound II-a (2.0 g,8.37 mmol) and elemental iodine (2.1 g,8.37 mmol) were added to ethanol (20 mL), stirred at 95℃for 30 min, then thiourea (1.3 g,16.7 mmol) was added, stirred at 95℃overnight, after completion of the reaction, concentrated, and purified by reverse phase column chromatography to give compound 6-a (2.1 g, yield 85%) as a yellow solid. LCMS [ M+H ]] + = 296.1。
Compound 6-a (2.1 g,7.12 mmol) and potassium carbonate (1.96 g,14.2 mmol) were added to N, N-dimethylformamide (20 mL), followed by addition of di-tert-butyl dicarbonate (1.7 g,7.83 mmol), stirring overnight at 40℃and, after completion of the reaction, water (100 mL) was added for dilution, and after extraction with ethyl acetate, the organic phase was concentrated and purified by column chromatography to give compound 6-b as a grey solid (2 g, yield 71.4%). LCMS [ M+H ] ] + = 396.1。
Then referring to example 2, compound 2-a was replaced with an equivalent of compound 6-c, compound M-a was replaced with an equivalent of compound 6-b, and compound 2-c was replaced with an equivalent of compound 6-e to give compound 6-f, which was deprotected to synthesize compound 6 as a yellow solid (700 mg, yield 83.8%). LCMS [ M+H ]] + = 507.1。
1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 9.73 (s, 1H), 9.02 (s, 1H), 8.63 (d, J = 4.8 Hz, 1H), 8.20 (d, J = 8.4 Hz, 1H), 7.83 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 8.0 Hz, 1H), 7.55 - 7.52 (m, 2H), 7.45 (d, J = 8.4 Hz, 1H), 7.13 - 7.11 (m, 2H), 4.06 (s, 3H), 3.00 - 2.94 (m, 7H)。
Example 7:
Figure SMS_38
the synthetic route is as follows:
Figure SMS_39
referring to the synthesis method of intermediate M-a, methyl iodide was replaced with an equivalent amount of trideuterated methyl iodide to obtain intermediate M-b, and then referring to example 2, compound 2-a was replaced with an equivalent amount of compound 7-a, compound M-a was replaced with an equivalent amount of M-b, and compound 2-c was replaced with an equivalent amount of 7-c, to obtain compound 7 as a yellow solid (80 mg, yield 40%). LCMS [ M+H] + = 524.1。
1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 9.01 (s, 1H), 8.63 (d, J = 3.0 Hz, 1H), 8.20 (d, J = 6.0 Hz, 1H), 7.83 (d, J = 6.0 Hz, 1H), 7.69 (d, J = 5.7 Hz, 1H), 7.54 - 7.52 (m, 2H), 7.45 (d, J = 5.7 Hz, 1H),7.32 - 7.29 (m, 2H), 4.05 (s, 3H), 3.02 - 2.98 (m, 4H), 2.96 (s, 3H)。
Example 8:
Figure SMS_40
the synthetic route is as follows:
Figure SMS_41
referring to the synthesis of intermediate M-a, methyl iodide was replaced with an equivalent amount of ethyl iodide to give intermediate M-c, and then referring to example 2, compound 2-a was replaced with an equivalent amount of compound 8-a, compound M-a was replaced with an equivalent amount of M-c, and compound 2-c was replaced with an equivalent amount of 8-c, to thereby obtain compound 8 as a yellow solid (10.4. 10.4 mg, yield 41%). LCMS [ M+H] + = 535.1。
1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 9.01 (s, 1H), 8.63 (d, J = 4.8 Hz, 1H), 8.20 (d, J = 8.0 Hz, 1H), 7.84 (d, J = 8.0 Hz, 1H), 7.71 (d, J = 8.0 Hz, 1H), 7.55 - 7.52 (m, 2H), 7.45 (d, J = 8.4 Hz, 1H), 7.27 (d, J = 9.6 Hz, 2H), 4.06 (s, 3H), 3.66 (q, J = 7.2 Hz, 2H), 3.04 - 2.99 (m, 7H), 1.02 (t, J = 7.2 Hz, 3H)。
Example 9:
Figure SMS_42
the synthetic route is as follows:
Figure SMS_43
referring to the synthesis of intermediate M-a, methyl iodide was replaced with an equivalent amount of 2-iodopropane to give intermediate M-d, and then referring to example 2, compound 2-a was replaced with an equivalent amount of compound 9-a, compound M-a was replaced with an equivalent amount of M-d, and compound 2-c was replaced with an equivalent amount of 9-c, to thereby obtain yellow solid compound 9 (10.4. 10.4 mg, yield 41%). LCMS [ M+H ] + = 549.1。
1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 9.01 (s, 1H), 8.63 (d, J = 4.0 Hz, 1H), 8.20 (d, J = 7.2 Hz, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.72 (d, J = 8.4 Hz, 1H), 7.55 - 7.52 (m, 2H), 7.46 (d, J = 8.0 Hz, 1H), 7.18 (d, J = 7.2 Hz, 2H), 4.36 - 4.29 (m, 1H), 4.06 (s, 3H), 3.07 - 3.00 (m, 7H), 1.10 (d, J = 6.8 Hz, 6H)。
Example 10:
Figure SMS_44
the synthetic route is as follows:
Figure SMS_45
referring to the synthesis method of intermediate M-a, methyl iodide was replaced with an equivalent amount of benzyl iodide to obtain intermediate M-e, and then referring to example 2, compound 2-a was replaced with an equivalent amount of Compound 10-a, compound M-A is replaced by an equivalent M-e, and the compound 2-c is replaced by an equivalent 10-c, thus obtaining the yellow solid compound 10 (23.2 mg, yield 51%). LCMS [ M+H] + = 597.1。
1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 9.01 (s, 1H), 8.62 (d, J = 4.8 Hz, 1H), 8.20 (d, J = 8.0 Hz, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.61 (d, J = 8.4 Hz, 1H), 7.54 - 7.51 (m, 2H), 7.44 (d, J = 8.0 Hz, 1H), 7.31 - 7.25 (m, 6H), 7.21 (q, J = 4.4 Hz, 1H), 4.86 (s, 2H), 4.04 (s, 3H), 3.10 (s, 3H), 2.99 - 2.92 (m, 4H)。
Example 11:
Figure SMS_46
/>
the synthetic route is as follows:
Figure SMS_47
referring to example 2, compound 2-a was replaced with an equivalent of compound 11-a and compound 2-c was replaced with an equivalent of 11-c, to thereby obtain compound 11 as a yellow solid (50 mg, yield 22%). LCMS [ M+H] + = 527.1。
1H NMR (400 MHz, DMSO-d6) δ 11.93 (s, 1H), 9.01 (s, 1H), 8.63 (d, J = 2.7 Hz, 1H), 8.20 (d, J = 6.0 Hz, 1H), 7.84 (d, J = 6.0 Hz, 1H), 7.69 (d, J = 6.0 Hz, 1H), 7.54-7.51 (m, 2H), 7.45 (d, J = 5.7Hz, 1H), 7.32 - 7.29 (m, 2H), 3.02 - 2.98 (m, 4H), 2.96 (s, 3H)。
Example 12:
Figure SMS_48
the synthetic route is as follows:
Figure SMS_49
compound 5 (50 mg,0.1 mmol) was added to dichloromethane (3 mL)Boron tribromide (0.2 mL) was added to the ice bath, followed by stirring at 20℃for 30 minutes, and after completion of the reaction, methanol was added to quench the reaction, followed by concentration and purification by reverse phase column chromatography to give compound 12 (20 mg, yield 39.5%) as a yellow solid. LCMS [ M+H] + = 507.1。
1H NMR (400 MHz, DMSO-d6) δ 12.08 (s, 1H), 8.93 (s, 1H), 8.63 (s, 1H), 8.16 - 8.11 (m, 2H), 7.43 (d, J = 6.0 Hz, 1H), 7.54 - 7.51 (m, 1H), 7.39 - 7.31 (m, 4H), 3.24 (s, 3H), 3.04 - 2.99 (m, 4H), 2.97 (s, 3H)。
Example 13:
Figure SMS_50
the synthetic route is as follows:
Figure SMS_51
referring to example 12, compound 5 was replaced with an equivalent of compound 7 to yield compound 13 as a yellow solid (200 mg, 93% yield). LCMS [ M+H ] + = 510.1。
1H NMR (400 MHz, DMSO-d6) δ 12.25 (s, 1H), 9.13 (s, 1H), 8.80 (d, J = 3.6 Hz,1H), 8.57 (d, J = 6.6 Hz,1H), 8.17 (d, J = 6.0 Hz, 1H), 7.89 (t, J = 5.7 Hz, 1H), 7.71 (d, J = 6.0 Hz, 1H), 7.45-7.41 (m, 2H), 7.33 - 7.31 (m, 2H), 3.03 - 2.98 (m, 4H), 2.96 (s, 3H)。
Example 14:
Figure SMS_52
the synthetic route is as follows:
Figure SMS_53
referring to example 2, the compound 2-b was replaced with an equivalent of 14-a (i.e., 5-b) and the compound 2-c was replaced with an equivalent of 14-b, to obtain yellowColored solid compound 14 (201 mg, yield 89.5%). LCMS [ M+H] + = 589.1。
1H NMR (400 MHz, DMSO-d6) δ 12.05 (s, 1H), 9.33 (s, 1H), 9.04 (s, 1H), 8.62 (s, 1H), 7.84 (d, J = 5.6 Hz, 1H), 7.70 (d, J = 6.0 Hz, 1H), 7.64 (s, 1H), 7.58 (d, J = 6.0 Hz, 1H), 7.33 - 7.31 (m, 2H), 4.08 (s, 3H), 3.24 (s, 3H), 3.04 - 3.00 (m, 4H), 2.97 (s, 3H)。
Example 15:
Figure SMS_54
the synthetic route is as follows:
Figure SMS_55
referring to example 2, compound 2-b was replaced with an equivalent of 15-a (i.e., 7-b) and compound 2-c was replaced with an equivalent of 15-b, thus obtaining compound 15 (60 mg, yield 37.6%) as a yellow solid. LCMS [ M+H] + = 592.1。
1H NMR (400 MHz, DMSO-d6) δ 12.00 (s, 1H), 9.31 (s, 1H), 9.01 (s, 1H), 8.60 (s, 1H), 7.83 (d, J = 6.4 Hz, 1H), 7.68 (d, J = 6.0 Hz, 1H), 7.62 (s, 1H), 7.56 (d, J = 5.6 Hz, 1H), 7.31 - 7.29 (m, 2H), 4.06 (s, 3H), 3.02 - 2.98 (m, 4H), 2.95 (s, 3H)。
Example 16:
Figure SMS_56
the synthetic route is as follows:
Figure SMS_57
referring to example 2, compound M-a was replaced with an equivalent of M-b, compound 2-a was replaced with an equivalent of 16-a, and compound 2-c was replaced with an equivalent of 16-c, to thereby obtain compound 16 (55.9. 55.9 mg, yield 24.5%) as a yellow solid. LCMS [ M+H] + = 566.1。
1H NMR (400 MHz, DMSO-d6) δ 12.85 (s, 1H), 9.14 (s, 1H), 8.46 – 8.35 (m, 1H), 8.28 (d, J = 8.0 Hz, 2H), 8.15 (d, J = 8.4 Hz, 1H), 8.01 (d, J = 8.4 Hz, 2H), 7.73 (d, J = 8.0 Hz, 1H), 7.34 - 7.31 (m, 2H), 4.37 (q, J = 7.2 Hz, 2H), 3.05 - 2.97 (m, 7H), 1.35 (t, J = 7.2 Hz, 3H)。
Example 17:
Figure SMS_58
the synthetic route is as follows:
Figure SMS_59
compound 17-a (i.e., 16-b,690 mg, 1.39 mmol), pinacol biborate (708 mg,2.79 mmol), 1' -bis (diphenylphosphine) ferrocene]Palladium dichloride (102 mg,0.14 mmol) and potassium acetate (273 mg,2.79 mmol) were added to 1, 4-dioxane (10 mL), stirred at 10℃for 6 hours, after completion of the reaction, concentrated and purified by column chromatography to give compound 17-b (750 mg, 99.4% yield) as a red oil, LCMS: [ M+H ] + = 543.1。
Compound 17-b (120 mg,0.22 mmol), compound 17-c (72 mg,0.33 mmol), [1,1' -bis (diphenylphosphine) ferrocene]Palladium dichloride (24 mg,0.022 mmol) and potassium carbonate (92 mg,0.66 mmol) were added to a mixed solvent of 1, 4-dioxane (4 mL) and water (0.5 mL), stirred overnight at 95℃and after completion of the reaction, concentrated and purified by column chromatography to give compound 17 (24.2 mg, yield 19.8%) as a yellow solid, LCMS: [ M+H] + = 552.1。
1H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 9.20 (s, 1H), 8.46 – 8.35 (m, 1H), 8.33 (d, J = 8.4 Hz, 2H), 8.27 (d, J = 8.4 Hz, 3H), 7.73 (d, J = 8.0 Hz, 1H), 7.34 - 7.31 (m, 2H), 3.91 (s, 3H), 3.04 - 2.97 (m, 7H)。
Example 18:
Figure SMS_60
the synthetic route is as follows:
Figure SMS_61
referring to example 2, compound M-a was replaced with an equivalent of M-b, compound 2-a was replaced with an equivalent of 18-a, and compound 2-c was replaced with an equivalent of 18-c, to thereby obtain compound 18 (57.1. 57.1 mg, yield 13.6%) as a yellow solid. LCMS [ M+H] + = 566.1。
1H NMR (400 MHz, DMSO-d6) δ 12.08 (s, 1H), 9.14 (s, 1H), 8.46 (d, J = 8.4 Hz, 1H), 8.28 (d, J = 8.4 Hz, 1H), 8.11 (d, J = 8.0 Hz, 2H), 8.02 (d, J = 8.0 Hz, 2H), 7.75 (d, J = 8.8 Hz, 1H), 7.34 - 7.32 (m, 2H), 4.36 (q, J = 7.2 Hz, 2H), 3.06 - 3.01 (m, 4H), 2.97 (s, 3H), 1.35 (t, J = 7.2 Hz, 3H)。
Example 19:
Figure SMS_62
the synthetic route is as follows:
Figure SMS_63
compound 18 (100 mg,0.18 mmol) was added to a mixture of tetrahydrofuran (2 mL) and ethanol (2 mL), then an aqueous solution of sodium hydroxide (35 mg,0.88 mmol) was added (1 mL), stirred overnight at 45℃and after the reaction was completed, concentrated and purified by column chromatography to give Compound 19 (31.1 mg, yield 32.7%) as a yellow solid, LCMS: [ M+H] + = 538.1。
1H NMR (400 MHz, DMSO-d6) δ 8.88 (s, 1H), 8.32 (d, J = 8.4 Hz, 1H), 8.19 – 8.03 (m, 4H), 7.97 (d, J = 8.0 Hz, 2H), 7.73 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 8.0 Hz, 2H), 7.27 - 7.23 (m, 2H), 3.00 - 2.94 (m, 5H), 2.88 - 2.84 (m, 2H)。
Example 20:
Figure SMS_64
the synthetic route is as follows:
Figure SMS_65
referring to example 2, compound 2-c was replaced with an equivalent amount of 20-c, and thus, compound 20 (346 mg, yield 76%) was synthesized as a yellow solid. LCMS [ M+H ] + = 562.1。
1H NMR (400 MHz, DMSO-d6) δ 12.80 (s, 1H), 8.24 (d, J = 6.0 Hz, 2H), 8.05 (d, J = 6.0 Hz, 2H), 7.91 (d, J = 6.0 Hz, 4H), 7.72 (d, J = 6.0 Hz, 1H), 7.32 (d, J = 6.0 Hz, 2H), 4.38 – 4.27 (m, 2H), 3.24 (s, 3H), 3.02 - 2.98 (m, 4H), 2.96 (s, 3H), 1.33 (t, J = 5.6 Hz, 3H)。
Example 21:
Figure SMS_66
the synthetic route is as follows:
Figure SMS_67
referring to example 2, compound 2-c was replaced with an equivalent amount of 21-c, and yellow solid compound 21 (100 mg, yield 46.7%) was synthesized. LCMS [ M+H] + = 565.1。
1H NMR (400 MHz, DMSO-d6) δ 12.78 (s, 1H), 8.24 (d, J = 6.0 Hz, 2H), 8.06 (d, J = 6.4 Hz, 2H), 7.92 (d, J = 6.0 Hz, 4H), 7.73 (d, J = 6.4 Hz, 1H), 7.32 (d, J = 5.6 Hz, 2H), 4.36 - 4.31 (m, 2H), 3.03 - 2.98 (m, 4H), 2.96 (s, 3H), 1.33 (t, J = 5.6 Hz, 3H)。
Example 22:
Figure SMS_68
the synthetic route is as follows:
Figure SMS_69
synthesis of Compound I-a with respect to intermediate M-a, substituting equivalent amounts of 5-amino-2, 3-dihydro-1H-inden-1-one to give intermediate M-f, further with respect to example 2, substituting equivalent amounts of M-f for Compound M-a and 22-c for Compound 2-c to give Compound 22-d, and further with respect to example 19, substituting equivalent amounts of 22-d for Compound 18 afforded Compound 22 as a yellow solid (10.2. 10.2 mg, yield 26%). LCMS [ M+H] + = 520.1。
1H NMR (400 MHz, DMSO-d6) δ 12.95 (s, 1H), 8.26 (d, J = 8.4 Hz, 2H), 8.05 (d, J = 8.4 Hz, 2H), 7.92 (t, J = 8.4 Hz, 4H), 7.64 (s, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.41 (d, J = 8.0 Hz, 1H), 3.96 (s, 2H), 3.27 (s, 3H), 2.97 (s, 3H)。
Example 23:
Figure SMS_70
the synthetic route is as follows:
Figure SMS_71
referring to the synthesis of intermediate M-a and example 22, substitution of methyl iodide with an equivalent amount of trideuterated methyl iodide gave intermediate M-g, and referring again to example 2, substitution of compound M-a with an equivalent amount of M-g and substitution of compound 2-c with an equivalent amount of 23-c, yellow solid compound 23 was synthesized (10 mg, 14% yield). LCMS [ M+H] + = 551.1。
1H NMR (400 MHz, CDCl3) δ 10.19 (s, 1H), 8.14 (d, J = 6.0 Hz, 2H), 8.06 (d, J = 6.0 Hz, 2H), 7.54 (d, J = 6.0 Hz, 2H), 7.66 (d, J = 6.0 Hz, 2H), 7.56 (s, 1H), 7.50 (d, J = 6.0 Hz, 1H), 7.29 (d, J = 6.0 Hz, 1H), 7.45 - 7.39 (m, 2H), 3.90 (s, 2H), 2.84 (s, 3H), 1.14 (t, J = 5.2 Hz, 3H)。
Example 24:
Figure SMS_72
the synthetic route is as follows:
Figure SMS_73
referring to the synthesis of intermediate M-a, compound I-a is replaced with an equivalent amount of 2-amino-6, 7,8, 9-tetrahydro-5H-benzo [7 ]]The intermediate M-h was obtained by substituting equivalent amount of trideuterated iodomethane for cycloolefin-5-one and equivalent amount of trideuterated iodomethane, and referring to example 2, compound 24 was synthesized by substituting equivalent amount of M-h for compound M-a and equivalent amount of 24-c for compound 2-c (3.3 mg, yield 18.4%). LCMS [ M+H ] + = 579.1。
1H NMR (400 MHz, DMSO-d6) δ 12.71 (s, 1H), 8.24 (d, J = 8.4 Hz, 2H), 8.07 - 8.04 (m, 3H), 7.97 – 7.89 (m, 4H), 7.42 – 7.31 (m, 1H), 7.29 (s, 1H), 4.34 (q, J = 7.2 Hz, 2H), 3.01 - 2.97 (m, 5H), 2.82 (d, J = 8.0 Hz, 2H), 2.12 - 2.06 (m, 2H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 25:
Figure SMS_74
the synthetic route is as follows:
Figure SMS_75
the intermediate M-I was obtained by substituting an equivalent amount of 7-aminochroman-4-one for compound I-a and an equivalent amount of tridentate iodomethane for iodomethane by referring to the synthesis of intermediate M-a, the yellow solid compound was obtained by substituting an equivalent amount of M-I for compound M-a and an equivalent amount of 25-c for compound 2-c by referring to example 2, 25-d for compound 25-d, and 25-d for compound 18 by referring to example 1925 (2.5. 2.5 mg, yield 26%). LCMS [ M+H] + = 539.1。
1H NMR (400 MHz, DMSO-d6) δ 8.23 (d, J = 8.0 Hz, 2H), 8.04 (d, J = 8.0 Hz, 2H), 7.90 (d, J = 8.4 Hz, 2H), 7.85 (d, J = 8.0 Hz, 2H), 7.62 (d, J = 8.4 Hz, 1H), 7.08 (d, J = 8.4 Hz, 1H), 7.00 (s, 1H), 5.53 (s, 2H), 2.95 (s, 3H)。
Example 26:
Figure SMS_76
the synthetic route is as follows:
Figure SMS_77
referring to example 2, compound M-a was replaced with equivalent M-b, compound 2-a was replaced with equivalent 26-a, and compound 2-c was replaced with equivalent 26-c, and then referring to example 19, compound 18 was replaced with equivalent 26-d, so that compound 26 (51.2. 51.2 mg, yield 77%) as a white solid was synthesized. LCMS [ M+H] + = 540.1。
1H NMR (400 MHz, DMSO-d6) δ 12.33 (s, 1H), 8.03 (d, J = 8.0 Hz, 2H), 7.72 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 8.0 Hz, 2H), 7.47 (d, J = 4.0 Hz, 1H), 7.32 - 7.30 (m, 2H), 6.41 (d, J = 4.0 Hz, 1H), 3.90 (s, 3H), 3.02 - 2.96 (m, 7H)。
Example 27:
Figure SMS_78
the synthetic route is as follows:
Figure SMS_79
referring to example 2, compound 2-b was replaced with an equivalent of 27-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 27-b, thus obtaining compound 27 (50. 50 mg, yield 50.5%) as a yellow solid. LCMS [ M+H] + = 551.1。
1H NMR (400 MHz, DMSO-d6) δ 12.80 (s, 1H), 8.25 (d, J = 6.0 Hz, 2H), 8.07 (d, J = 6.4 Hz, 2H), 7.95 - 7.92 (m, 4H), 7.73 (d, J = 6.0 Hz, 1H), 7.33 - 7.31 (m, 2H), 3.88 (s, 3H), 3.05 - 2.98 (m, 4H), 2.96 (s, 3H)。
Example 28:
Figure SMS_80
the synthetic route is as follows:
Figure SMS_81
referring to example 2, compound 2-b was replaced with an equivalent of 28-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 28-b, thus obtaining compound 28 (80 mg, yield 76.9%) as a yellow solid. LCMS [ M+H ] + = 579.1。
1H NMR (400 MHz, DMSO-d6) δ 12.80 (s, 1H), 8.25 (d, J = 6.4 Hz, 2H), 8.05 (d, J = 6.4 Hz, 2H), 7.94 - 7.91 (m, 4H), 7.73 (d, J = 6.0 Hz, 1H), 7.33 - 7.30 (m, 2H), 5.19 - 5.13 (m, 1H), 3.05 - 2.98 (m, 4H), 2.96 (s, 3H), 1.34 (d, J = 4.8 Hz, 6H)。
Example 29:
Figure SMS_82
the synthetic route is as follows:
Figure SMS_83
referring to example 2, compound 2-b was replaced with an equivalent of 29-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 29-b, thus obtaining compound 29 (60 mg, yield 56.3%) as a yellow solid. LCMS [ M+H] + = 593.1。
1H NMR (400 MHz, DMSO-d6) δ12.80 (s, 1H), 8.25 (d, J = 6.4 Hz, 2H), 8.06 (d, J = 6.0 Hz, 2H), 7.93 - 7.90 (m, 4H), 7.73 (d, J = 6.0 Hz, 1H), 7.33 - 7.31 (m, 2H), 4.30 (t, J = 4.8 Hz, 2H), 3.05-2.99 (m, 4H), 2.96 (s, 3H), 1.74 - 1.67 (m, 2H), 1.48 - 1.39 (m, 2H), 0.94 (t, J = 5.4 Hz, 3H)。
Example 30:
Figure SMS_84
the synthetic route is as follows:
Figure SMS_85
referring to example 2, compound 2-b was replaced with an equivalent of 30-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 30-b, thus obtaining compound 30 (55 mg, yield 51.6%) as a yellow solid. LCMS [ M+H] + = 593.1。
1H NMR (400 MHz, DMSO-d6) δ12.82 (s, 1H), 8.25 (d, J = 8.4 Hz, 2H), 8.08 (d, J = 8.4 Hz, 2H), 7.96 – 7.89 (m, 4H), 7.73 (d, J = 8.0 Hz, 1H), 7.33 (d, J = 7.6 Hz, 1H), 4.10 (d, J = 6.4 Hz, 2H), 3.08 – 2.98 (m, 4H), 2.97 (s, 3H), 2.12 – 1.99 (m, 1H), 0.99 (d, J = 6.8 Hz, 6H)。
Example 31:
Figure SMS_86
the synthetic route is as follows:
Figure SMS_87
referring to example 2, compound 2-b was replaced with an equivalent of 31-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 31-b, thus obtaining compound 31 (80 mg, yield 71.7%) as a yellow solid. LCMS [ M+H] + = 621.1。
1H NMR (400 MHz, DMSO-d6) δ12.80 (s, 1H), 8.25 (d, J = 6.0 Hz, 2H), 8.05 (d, J = 6.0 Hz, 2H), 7.95 - 7.91 (m, 4H), 7.73 (d, J = 6.0 Hz, 1H), 7.33 - 7.31 (m, 2H), 4.24 (d, J = 4.2 Hz, 2H), 3.05 - 2.98 (m, 4H), 2.96 (s, 3H), 1.68 - 1.62 (m, 1H), 1.46 - 1.38 (m, 4H), 0.92 (t, J = 5.6 Hz, 6H)。
Example 32:
Figure SMS_88
/>
the synthetic route is as follows:
Figure SMS_89
referring to example 2, compound 2-b was replaced with an equivalent of 32-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 32-b, thus obtaining compound 32 (78 mg, yield 70.2%) as a yellow solid. LCMS [ M+H] + = 595.1。
1H NMR (400 MHz, CDCl3) δ 9.99 (s, 1H), 8.18 (d, J = 8.0 Hz, 2H), 8.03 (d, J = 8.0 Hz, 2H), 7.76 – 7.66 (m, 5H), 7.23 (d, J = 8.0 Hz, 2H), 4.55 – 4.48 (m, 2H), 3.79 – 3.74 (m, 2H), 3.46 (s, 3H), 3.10 – 3.01 (m, 4H), 2.86 (s, 3H)。
Example 33:
Figure SMS_90
the synthetic route is as follows:
Figure SMS_91
referring to example 2, compound 2-b was replaced with an equivalent of 33-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 33-b, whereby compound 33 (43 mg, yield 66.2%) was synthesized as a yellow solid. LCMS [ M+H ] + = 609.1。
1H NMR (400 MHz, CDCl3) δ 9.97 (s, 1H), 8.18 (d, J = 8.0 Hz, 2H), 8.03 (d, J = 8.0 Hz, 2H), 7.78 – 7.65 (m, 5H), 7.23 (d, J = 8.4 Hz, 2H), 4.57 – 4.47 (m, 2H), 3.84 – 3.77 (m, 2H), 3.61 (q, J = 7.2 Hz, 2H), 3.11 – 2.99 (m, 4H), 2.86 (s, 3H), 1.26 (t, J = 7.2 Hz, 3H)。
Example 34:
Figure SMS_92
the synthetic route is as follows:
Figure SMS_93
referring to example 2, compound 2-b was replaced with an equivalent of 34-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 34-b, thus obtaining compound 34 (27 mg, yield 31%) as a yellow solid. LCMS [ M+H] + = 650.1。
1H NMR (400 MHz, CDCl3) δ 8.15 (d, J = 8.0 Hz, 2H), 8.08 (d, J = 8.0 Hz, 2H), 7.82 – 7.67 (m, 5H), 7.27 (s, 2H), 4.57 – 4.46 (m, 2H), 3.75 (s, br, 4H), 3.15 – 2.99 (m, 4H), 2.87 (s, 3H), 2.83 (s, 2H), 2.62 (s, 4H)。
Example 35:
Figure SMS_94
the synthetic route is as follows:
Figure SMS_95
referring to example 2, compound 2-b was replaced with an equivalent of 35-a (i.e., 16-b) and compound 2-c was replaced with an equivalent of 35-b, thus obtaining compound 35 (47 mg, yield 53.4%) as a yellow solid. LCMS [ M+H] + = 608.1。
1H NMR (400 MHz, CDCl3) δ10.23 (s, 1H), 8.15 (d, J = 8.0 Hz, 2H), 8.02 (d, J = 8.0 Hz, 2H), 7.82 – 7.58 (m, 5H), 7.28 – 7.08 (m, 2H), 4.48 (t, J = 5.6 Hz, 2H), 3.16 – 2.97 (m, 4H), 2.85 (s, 3H), 2.78 (t, J = 5.6 Hz, 2H), 2.38 (s, 6H)。
Example 36:
Figure SMS_96
the synthetic route is as follows:
Figure SMS_97
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent amount of acetyl chloride, methyl iodide was replaced with an equivalent amount of trideuterated methyl iodide to give intermediate M-j, and referring again to example 2, compound M-a was replaced with an equivalent amount of M-j, and compound 2-c was replaced with an equivalent amount of 36-c, to give compound 36 as a yellow solid (290 mg, yield 83.8%). LCMS [ M+H] + = 529.1。
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.06 (d, J = 8.4 Hz, 2H), 7.98 – 7.89 (m, 4H),7.76 (d, J = 8.0 Hz, 1H), 7.26 (s, 1H), 7.22 (d, J = 8.4 Hz, 1H), 4.34 (q, J = 7.2 Hz, 2H), 3.05 - 2.98 (m, 4H), 1.82 (s, 3H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 37:
Figure SMS_98
the synthetic route is as follows:
Figure SMS_99
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of 1-methoxyacetyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to give intermediate M-k, and referring again to example 2, compound M-a was replaced with an equivalent of M-k, and compound 2-c was replaced with an equivalent of 37-c, to give compound 37 as a yellow solid (33.7. 33.7 mg, yield 58.8%). LCMS [ M+H ] + = 559.1。
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.0 Hz, 2H), 7.98 – 7.88 (m, 4H), 7.75 (d, J = 8.0 Hz, 1H), 7.27 (s, 1H), 7.23 (d, J = 8.0 Hz, 1H), 4.34 (q, J = 7.2 Hz, 2H), 3.84 (s, 2H), 3.21 (s, 3H), 3.04 - 3.00 (m, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 38:
Figure SMS_100
the synthetic route is as follows:
Figure SMS_101
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of ethylsulfonyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to give intermediate M-l, and referring again to example 2, compound M-a was replaced with an equivalent of M-l, and compound 2-c was replaced with an equivalent of 38-c, to give compound 38 as a yellow solid (3 mg, yield 12.5%). LCMS [ M+H] + = 579.1。
1H NMR (400 MHz, DMSO-d6) δ 12.82 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 7.98 – 7.89 (m, 4H), 7.72 (d, J = 8.0 Hz, 1H), 7.34 - 7.32 (m, 2H), 4.34 (q, J = 7.2 Hz, 2H), 3.16 (q, J = 7.6 Hz, 2H), 3.05 - 2.97 (m, 4H), 1.34 (t, J = 7.2 Hz, 3H), 1.22 (t, J = 7.2 Hz, 3H)。
Example 39:
Figure SMS_102
the synthetic route is as follows:
Figure SMS_103
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of propylsulfonyl chloride and iodomethane was replaced with an equivalent of tridentate iodomethane to give intermediate M-M, which was again referred toIn example 2, compound M-a was replaced with an equivalent of M-M and compound 2-c was replaced with an equivalent of 38-c, to yield compound 39 as a yellow solid (3 mg, 12.5% yield). LCMS [ M+H] + = 593.1。
1H NMR (400 MHz, DMSO-d6) δ 12.82 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 7.98 – 7.87 (m, 4H), 7.72 (d, J = 8.4 Hz, 1H), 7.34 - 7.32 (m, 2H), 4.34 (q, J = 7.2 Hz, 2H), 3.12 (t, J = 7.6 Hz, 2H), 3.05 - 2.97 (m, 4H), 1.74 - 1.65 (m, 2H), 1.34 (t, J = 7.2 Hz, 3H), 0.97 (t, J = 7.2 Hz, 3H)。
Example 40:
Figure SMS_104
the synthetic route is as follows:
Figure SMS_105
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of isobutylsulfonyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to give intermediate M-n, and referring again to example 2, compound M-a was replaced with an equivalent of M-n, and compound 2-c was replaced with an equivalent of 40-c, to give compound 40 as a yellow solid (15.2. 15.2 mg, yield 17.4%). LCMS [ M+H ] + = 607.1。
1H NMR (400 MHz, DMSO-d6) δ 12.82 (s, 1H), 8.25 (d, J = 8.4 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 7.98 – 7.89 (m, 4H), 7.72 (d, J = 8.8 Hz, 1H), 7.34 - 7.31 (m, 2H), 4.35 (q, J = 7.2 Hz, 2H), 3.12 (t, J = 7.6 Hz, 2H), 3.05 - 2.97 (m, 6H), 2.15 - 2.08 (m, 1H), 1.35 (t, J = 7.2 Hz, 3H), 1.02 (s, 3H), 1.00 (s, 3H)。
Example 41:
Figure SMS_106
the synthetic route is as follows:
Figure SMS_107
referring to the synthesis of intermediate M-a, the replacement of methanesulfonyl chloride with an equivalent of trifluoromethanesulfonyl chloride and the replacement of iodomethane with an equivalent of trideuterated iodomethane gave intermediate M-o, and referring again to example 2, the replacement of compound M-a with an equivalent of M-o and the replacement of compound 2-c with an equivalent of 41-c gave compound 41 as a yellow solid (31.9. 31.9 mg, yield 41.8%). LCMS [ M+H] + = 619.1。
1H NMR (400 MHz, DMSO-d6) δ 12.85 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 8.04 – 7.85 (m, 4H), 7.78 (d, J = 8.4 Hz, 1H), 7.43 - 7.38 (m, 2H), 4.34 (q, J = 7.2 Hz, 2H), 3.06 - 3.01 (m, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 42:
Figure SMS_108
the synthetic route is as follows:
Figure SMS_109
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of cyclopropylsulfonyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to obtain intermediate M-p, and referring again to example 2, compound M-a was replaced with an equivalent of M-p, and compound 2-c was replaced with an equivalent of 42-c, to obtain compound 42 as a yellow solid (31.9. 31.9 mg, yield 41.8%). LCMS [ M+H] + = 591.1。
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.06 (d, J = 8.4 Hz, 2H), 7.97 – 7.89 (m, 4H), 7.73 (d, J = 8.8 Hz, 1H), 7.36 - 7.34 (m, 2H), 4.34 (q, J = 7.2 Hz, 2H), 3.05 - 2.98 (m, 4H), 2.73 - 2.66 (m, 1H), 1.34 (t, J = 7.2 Hz, 3H), 0.99 - 0.95 (m, 2H), 0.86 - 0.82 (m, 2H)。
Example 43:
Figure SMS_110
the synthetic route is as follows:
Figure SMS_111
referring to example 42, compound 42-c was replaced with an equivalent amount of 43-b (the same compound as that of compound 42-b and 43-a), and yellow solid compound 43 was synthesized (72.4. 72.4 mg, yield 60.8%). LCMS [ M+H] + = 676.1。
1 H NMR (400 MHz, CDCl3) δ 10.25 (s, 1H), 8.14 (d, J = 7.6 Hz, 2H), 8.02 (d, J = 7.2 Hz, 2H), 7.72 - 7.65 (m, 5H), 7.27 - 7.24 (m, 2H), 4.51 (t, J = 5.6 Hz, 2H), 3.74 (t, J = 4.0 Hz, 4H), 3.08 - 3.01 (m, 4H), 2.82 (t, J = 5.6 Hz, 2H), 2.60 (t, J = 4.0 Hz, 4H), 2.40 - 2.33 (m, 1H), 1.12 - 1.08 (m, 2H), 0.95 - 0.90 (m, 2H)。
Example 44:
Figure SMS_112
the synthetic route is as follows:
Figure SMS_113
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent amount of 6-sulfonylchlorobenzo [ d ] ]Thiazole, substitution of methyl iodide with equivalent amount of trideuterated methyl iodide to give intermediate M-q, and further reference to example 2, substitution of compound M-a with equivalent amount of M-q and substitution of compound 2-c with equivalent amount of 44-c, yellow solid compound 44 (47.9. 47.9 mg, yield 68.5%) was synthesized. LCMS [ M+H] + = 684.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 9.64 (s, 1H), 8.57 (s, 1H), 8.28 – 8.15 (m, 3H), 8.06 (d, J = 8.4 Hz, 2H), 8.04 – 7.85 (m, 4H), 7.65 (d, J = 8.0 Hz, 1H), 7.63 – 7.52 (m, 1H), 7.07 (s, 1H), 7.04 – 6.91 (m, 1H),4.34 (q, J = 7.2 Hz, 2H), 2.95 (s, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 45:
Figure SMS_114
the synthetic route is as follows:
Figure SMS_115
referring to example 44, compound 44-c was replaced with an equivalent amount of 45-b (the same compound as that of compound 44-b and 45-a), and thus, compound 43 (64.2. 64.2 mg, yield 55.8%) was synthesized as a yellow solid. LCMS [ M+H] + = 769.1。
1 H NMR (400 MHz, CDCl3) δ 10.07 (s, 1H), 9.20 (s, 1H), 8.27 (s, 1H), 8.18 (d, J = 8.4 Hz, 1H), 8.14 (d, J = 8.4 Hz, 2H), 8.03 (d, J = 8.0 Hz, 2H), 7.73 (d, J = 8.0 Hz, 2H), 7.69 (d, J = 8.0 Hz, 3H), 7.57 (d, J = 8.4 Hz, 1H), 7.07 (s, 1H), 6.81 (d, J = 8.4 Hz, 1H), 4.50 (t, J = 6.0 Hz, 2H), 3.74 (t, J = 4.8 Hz, 4H), 3.01 (s, 4H), 2.81 (t, J = 6.0 Hz, 2H), 2.60 (t, J = 4.4 Hz, 4H)。
Example 46:
Figure SMS_116
the synthetic route is as follows:
Figure SMS_117
referring to intermediate M-a, methanesulfonyl chloride was replaced with an equivalent amount of benzenesulfonyl chloride, methyl iodide was replaced with an equivalent amount of trideuterated methyl iodide, and intermediate M-r was synthesized, and referring to example 2, compound M-a was replaced with an equivalent amount of M-r, and compound 2-c was replaced with an equivalent amount of 46-c, yellow solid compound 46 (60.2 mg,yield 77.9%). LCMS [ M+H] + = 627.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.0 Hz, 2H), 8.02 – 7.85 (m, 4H), 7.71 (t, J = 7.2 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.61 - 7.53 (m, 4H), 7.04 (s, 1H), 7.01 – 6.85 (m, 1H), 4.34 (q, J = 7.2 Hz, 2H), 2.96 (s, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 47:
Figure SMS_118
the synthetic route is as follows:
Figure SMS_119
referring to the synthesis of intermediate M-a, the replacement of methanesulfonyl chloride with an equivalent of p-toluenesulfonyl chloride, the replacement of iodomethane with an equivalent of trideuterated iodomethane, to give intermediate M-s, and referring again to example 2, the replacement of compound M-a with an equivalent of M-s, and the replacement of compound 2-c with an equivalent of 47-c, yellow solid compound 47 was obtained (52.6 mg, yield 72.9%). LCMS [ M+H ] + = 641.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 8.04 – 7.85 (m, 4H), 7.65 (d, J = 8.0 Hz, 1H), 7.44 - 7.38 (m, 4H), 7.06 (s, 1H), 7.01 – 6.85 (m, 1H), 4.34 (q, J = 7.2 Hz, 2H), 2.97 (s, 4H), 2.39 (s, 3H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 48:
Figure SMS_120
the synthetic route is as follows:
Figure SMS_121
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of p-fluorobenzenesulfonyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to give intermediate M-t, and referring again to example 2, compound M-a was replaced with an equivalent of M-t, and compound 2-c was replaced with an equivalent of 48-c to give compound 48 as a yellow solid (47.5 mg, yield 72%). LCMS [ M+H] + = 645.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.4 Hz, 2H), 8.04 – 7.85 (m, 4H), 7.66 (d, J = 8.0 Hz, 1H), 7.62 - 7.58 (m, 2H), 7.44 (t, J = 8.4 Hz, 2H), 7.06 (s, 1H), 7.01 – 6.85 (m, 1H), 4.34 (q, J = 7.2 Hz, 2H), 2.97 (s, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 49:
Figure SMS_122
the synthetic route is as follows:
Figure SMS_123
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent amount of 3-cyano-4-fluorobenzenesulfonyl chloride, methyl iodide was replaced with an equivalent amount of trideuterated methyl iodide to obtain intermediate M-u, and referring again to example 2, compound M-a was replaced with an equivalent amount of M-u, and compound 2-c was replaced with an equivalent amount of 49-c, to obtain compound 49 (47.4 mg, yield 69%) as a yellow solid. LCMS [ M+H] + = 670.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.23 – 8.11 (m, 1H), 8.07 (d, J = 8.4 Hz, 2H), 7.99 – 7.89 (m, 4H), 7.83 - 7.78 (m, 1H), 7.72 (t, J = 8.0 Hz, 1H), 7.68 (d, J = 8.0 Hz, 1H), 7.09 (s, 1H), 7.05 – 6.79 (m, 1H), 4.34 (q, J = 7.2 Hz, 2H), 2.98 (s, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 50:
Figure SMS_124
the synthetic route is as follows:
Figure SMS_125
referring to the synthesis of intermediate M-a, methanesulfonyl chloride was replaced with an equivalent of benzylsulfonyl chloride, methyl iodide was replaced with an equivalent of trideuterated methyl iodide to give intermediate M-v, and referring again to example 2, compound M-a was replaced with an equivalent of M-v, and compound 2-c was replaced with an equivalent of 50-c to give compound 50 as a yellow solid (59.2 mg, yield 37.7%). LCMS [ M+H ] + = 641.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.25 (d, J = 8.0 Hz, 2H), 8.07 (d, J = 8.0 Hz, 2H), 7.99 – 7.89 (m, 4H), 7.68 (d, J = 8.4 Hz, 1H), 7.41 -7.36 (m, 5H), 7.25 – 7.13 (m, 4H),, 7.09 (s, 1H), 4.55 (s, 2H), 4.34 (q, J = 7.2 Hz, 2H), 2.98 (s, 4H), 1.34 (t, J = 7.2 Hz, 3H)。
Example 51:
Figure SMS_126
the synthetic route is as follows:
Figure SMS_127
referring to example 1, compound 1-a was replaced with an equivalent amount of 51-a, and thus, a yellow solid compound 51 was synthesized (35 mg, yield 30%). LCMS [ M+H] + = 448.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 8.11 (d, J = 6.0 Hz, 2H), 7.70 (d, J = 6.0 Hz, 1H), 7.60 (d, J = 6.8 Hz, 2H), 7.32-7.29 (m, 2H), 3.22 (s, 3H), 3.03-2.97 (m, 4H), 2.95 (s, 3H)。
Example 52:
Figure SMS_128
the synthetic route is as follows:
Figure SMS_129
referring to example 1, compound 1-a was replaced with an equivalent amount of 52-a to synthesize yellow solid compound 52 (84 mg, yield 54%). LCMS [ M+H] + = 482.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.98 (s, 1H), 8.49 (s, 1H), 8.38 (d, J = 6.0 Hz, 1H), 7.99 (d, J = 6.0 Hz, 1H), 7.78 (t, J = 6.0 Hz, 1H), 7.71 (d, J = 6.0 Hz, 1H), 7.32 (d, J = 4.4 Hz, 1H), 3.23 (s, 3H), 3.02-2.98 (m, 4H), 2.95 (s, 3H)。
Example 53:
Figure SMS_130
the synthetic route is as follows:
Figure SMS_131
referring to example 1, compound 1-a was replaced with an equivalent of 53-a to synthesize yellow solid compound 53 (70 mg, yield 45%). LCMS [ M+H] + = 432.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.72 (s, 1H), 8.20-8.17 (m, 2H), 7.71 (d, J = 6.0 Hz, 1H), 7.37 (t, J = 6.0 Hz, 2H), 7.32-7.29 (m, 2H), 3.23 (s, 3H), 3.02-2.97 (m, 4H), 2.95 (s, 3H)。
Example 54:
Figure SMS_132
the synthetic route is as follows:
Figure SMS_133
referring to example 19, compound 18 was replaced with an equivalent of 20 to afford compound 54 as a yellow solid (38 mg, 51% yield). LCMS [ M+H] + = 534.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.80 (s, 1H), 8.24 (d, J = 6.4 Hz, 2H), 8.05 (d, J = 6.4 Hz, 2H), 7.92 - 7.89 (m, 4H), 7.73 (d, J = 6.0 Hz, 1H), 7.32 (d, J = 6.4 Hz, 2H), 3.24 (s, 3H), 3.05 - 2.99 (m, 4H), 2.96 (s, 3H)。
Example 55:
Figure SMS_134
the synthetic route is as follows:
Figure SMS_135
referring to example 4, compound 2 was replaced with an equivalent of 54 to synthesize yellow solid compound 55 (20 mg, yield 52.3%). LCMS [ M+H] + = 532.1。
1 H NMR (400 MHz, DMSO-d6) δ 13.19 (s, 1H), 8.59 (d, J = 6.4 Hz, 1H), 8.31 (d, J = 6.4 Hz, 2H), 8.15 (d, J = 6.4 Hz, 1H), 8.10 (s, 1H), 8.06 (d, J = 6.0 Hz, 2H), 7.97 - 7.91 (m, 4H), 7.87 (d, J = 6.4 Hz, 1H), 7.73 (d, J = 6.8 Hz, 1H), 3.37 (s, 3H), 3.02 (s, 3H)。
Example 56:
Figure SMS_136
the synthetic route is as follows:
Figure SMS_137
reference example 19, compound 18 was replaced with an equivalent of 21, and yellow solid compound 56 (17 mg, yield 35.2%) was synthesized. LCMS [ M+H] + = 537.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.79 (s, 1H), 8.24 (d, J = 6.0 Hz, 2H), 8.05 (d, J = 6.4 Hz, 2H), 7.91 (t, J = 5.6 Hz, 4H), 7.73 (d, J = 6.4 Hz, 1H), 7.33 - 7.31 (m, 2H), 3.03 - 2.99 (m, 4H), 2.96 (s, 3H)。
Example 57:
Figure SMS_138
the synthetic route is as follows:
Figure SMS_139
referring to example 19, compound 18 was replaced with 42 equivalents to afford compound 57 as a yellow solid (18.6, mg, 17.8% yield). LCMS [ M+H ] + = 563.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.82 (s, 1H), 8.24 (d, J = 8.4 Hz, 2H), 8.05 (d, J = 8.0 Hz, 2H), 7.93 - 7.90 (m, 4H), 7.73 (d, J = 8.4 Hz, 1H), 7.36 - 7.34 (m, 2H), 3.06 - 2.97 (m, 4H), 2.73 - 2.66 (m, 1H), 1.00 - 0.95 (m, 2H), 0.86 - 0.82 (m, 2H)。
Example 58:
Figure SMS_140
the synthetic route is as follows:
Figure SMS_141
referring to example 19, compound 18 was replaced with an equivalent of 44 to afford compound 58 as a yellow solid (2.8, mg, yield 2.6%). LCMS [ M+H] + = 656.1。
1 H NMR (400 MHz, DMSO-d6) δ 12.83 (s, 1H), 9.64 (s, 1H), 8.58 (s, 1H), 8.24 (d, J = 8.0 Hz, 3H), 8.05 (d, J = 7.6 Hz, 2H), 7.91 (t, J = 7.2 Hz, 3H), 7.64 (d, J = 8.4 Hz, 1H), 7.59 (d, J = 7.6 Hz, 1H), 7.07 (s, 1H), 6.99 (d, J = 8.0 Hz, 1H), 2.95 (s, 4H)。
Effect of COUP inhibitors on proliferation Activity of prostate cancer cells
1. Method of
Prostate cancer cells LNCaP, PC-3, DU-145 were seeded in 96-well plates at a density of 1.5X104/well and incubated overnight at 37℃in 5% CO 2. The next day, the compounds were diluted at a maximum concentration of 100 uM at a 3-fold gradient, added to the cell culture medium at 10 total concentrations for a further treatment of 24 h, and split into 2 total wells.
After 24 h treatment of the cells with the compound, the cell viability was determined according to CellTiter 96 [ b ] AQueous One Solution Cell Proliferation Assay kit (Promega, G3580) as follows: mu.L MTS reagent is added into each hole, the mixture is placed in an incubator for incubation for 1 hour at 37 ℃, and the absorbance is measured by an MD i3x multifunctional enzyme label instrument with the wavelength of 490 and nM.
The effect of compounds on cell proliferation was evaluated as cell viability and IC50 values were calculated using GraphPad prism8.0 software to fit dose-response curves with three parameters. Cell viability= (signal value of sample-signal value of medium control)/(signal value of DMSO control-signal value of medium control) ×100%. Inhibition ratio = (1-cell viability) ×100%.
2. Results
Figure SMS_142
/>
Figure SMS_143
*: IC50 > 10μM; **:10μM ≥ IC50 > 1μM; ***:1μM ≥ IC50 > 0.1μM; ****:0.1μM ≥ IC50
†:30 ≥ Max;††:50 ≥Max > 30;†††:70 ≥Max > 50;††††: Max > 70
Summarizing: the compound provided by the invention has better inhibition activity on the proliferation of prostate cancer cells.
Method for detecting inhibition activity of compound on COUP-TF II based on reporter gene activity detection
1. Method of
1.1 Plasmid cotransfection HEK293T cells and compound treatment
HEK293T cells were seeded at a density of 1X 104/well in 96-well plates one day prior to plasmid transfection. Cell transfection was performed according to the instructions of transfection reagent F. Mu. GENE cube HD (Promega, # E2311). The method mainly comprises the following steps: taking a single well as an example, the plasmids pCR3.1-COUP-TFII and pXP2-NGFIA-Luc were added to 10. Mu.L of Opti-MEM ™ I medium (Gibco, # 11058021) at a ratio of 20ng to 5 ng and mixed well; adding 0.2 mu L of Fmu gENE cube HD, uniformly mixing, and standing at 20 ℃ for 5 min; this 10. Mu.L mixture was then added to the cell well containing 100. Mu.L of culture medium. After cell co-transfection 6 h, the compounds were diluted in a gradient at 100 uM at maximum concentration at half log dilution, 10 total concentrations were added to cell culture broth for treatment 24 h total of 2 multiplex wells.
1.2 One-Glo Luciferase assay
Cells were treated with the compound 24 h and examined according to ONE-Glo Luciferase Assay System (Promega, # E6120) instructions. The method mainly comprises the following steps: 50 mu L of culture solution is sucked and removed from each hole, 50 mu L of ONE-Glo Luciferase reagent is added, and the mixture is oscillated for 10 min at 20 ℃; mu.L of the cleavage reaction solution was transferred to a white opaque optiPlate-96 well plate, and the luminescence signal value (Firefly-Luc) of Firefly luciferase (Firefly luciferase) was detected by an MD i3x multifunctional microplate reader. EC50 values were calculated using the Firefly-Luc signal values as inhibitory activity of the compounds on COUP-TF II, normalized to the ratio of solvent DMSO groups, and dose-response curves fitted with three parameters using GraphPad prism8.0 software. And the inhibition ratio was calculated as the following formula = (Luc of Luc/DMSO group of 1-compound treated samples) ×100%. VCT-8 was used as a positive control compound (WO 2019/222134).
Figure SMS_144
2. Results
Experimental data are shown in the following table.
Figure SMS_145
*:20μM ≥ EC50 > 5μM; **:5μM ≥ EC50 > 1μM; ***:1μM ≥ EC50 > 0.1μM;
****:0.1μM ≥ EC50
†:20 ≥ Max;††:40 ≥ Max > 20;†††:70 ≥Max > 40;††††: Max > 70
Summarizing: the compound provided by the invention has better inhibition activity on COUP-TFII.
Method for detecting inhibition activity of compound on NRF2 based on reporter gene activity detection
1. Method of
1.1 Plasmid cotransfection HEK293T cells and compound treatment
pGL4.37[ luc2P/ARE ] (Promega, # E3641) and pRL-TK (Promega, # E2241) plasmids were purchased from Promega; the DH 5. Alpha. E.coli was transformed with the plasmid by CaCl2 method, and after further culture amplification, the corresponding plasmid DNA was obtained by purification using plasmid extraction kit (TIANGEN, #D107). HEK293T cells were seeded at a density of 1X 104/well in 96-well plates one day prior to plasmid transfection. Cell transfection was performed according to the instructions of transfection reagent F. Mu. GENE cube HD (Promega, # E2311). The method mainly comprises the following steps: taking a single well as an example, plasmids pGL4.37[ luc2P/ARE ] and pRL-TK were added to 10. Mu.L of Opti-MEM ™ I medium (Gibco, # 11058021) at a ratio of 80ng to 8 ng and mixed well; adding 0.2 mu L of Fmu gENE cube HD, uniformly mixing, and standing at 20 ℃ for 5 min; this 10. Mu.L mixture was then added to the cell well containing 100. Mu.L of culture medium. After cell co-transfection 6 h, the compounds were diluted in a gradient at 100 uM at maximum concentration at half log dilution, 10 total concentrations were added to cell culture broth for treatment 24 h total of 2 multiplex wells.
1.2 Dual-Glo Luciferase assay
Cells were treated with the compound 24 h and tested according to the Dual-Glo Luciferase Assay System (Promega, # E2940) instructions. The method mainly comprises the following steps: 50 mu L of culture solution is sucked and removed from each well, 50 mu L of Dual-Glo Luciferase reagent is added, and the mixture is oscillated for 10 min at 20 ℃; taking 80 mu L of the cleavage reaction solution to a white opaque optiPlate-96 well plate, and detecting a luminescence signal value (Firefly-Luc) of Firefly luciferase (Firefly luciferase) by using an MD i3x multifunctional enzyme-labeled instrument; then 40 mu L of Dual-Glo cube Stop & Glo cube reagent is added, and the mixture is oscillated for 10 min at 20 ℃; the luminescence signal value (Renilla-Luc) of Renilla luciferase (Renilla luciferase) was detected by an MD i3x multifunctional microplate reader. EC50 values were calculated using GraphPad prism8.0 software to fit dose-response curves with three parameters, with the ratio of Firefly-Luc/Renilla-Luc as the inhibitory activity of the compound on NRF2, and normalized to the ratio of solvent DMSO group. And the inhibition ratio (inhibition ratio) was calculated as follows, inhibition = (ratio of 1-compound treated sample of Firefly-Luc/Renila-Luc/ratio of DMSO group of Firefly-Luc/Renila-Luc) ×100%.
2. Results
Experimental data are given in the following table:
Figure SMS_146
*: EC50 > 20μM; **:20μM ≥ EC50 > 10μM; ***:10μM ≥ EC50 > 1μM;****:1μM ≥ EC50;
†:20 ≥ Max;††:40 ≥ Max > 20;†††:70 ≥Max > 40;††††: Max > 70
summarizing: the compound provided by the invention also has better inhibition activity on NRF 2.
Pharmacokinetic evaluation
In mice, the bioavailability and pharmacokinetic behavior of the compounds were evaluated. 6 male ICR mice of similar body weight were selected, of which 3 were given 10 mg/kg by single gavage and 3 were given intravenously at a single dose of 5 mg/kg. Blood samples were taken at time points of 5 minutes (intravenous), 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours and 7 hours post-dose, plasma samples were analyzed for concentration by LC-MS/MS and the pharmacokinetic parameters of the compounds were analyzed using PKSolver free tools and non-compartmental model (NCA) software.
Experimental protocol:
experimental animals: each compound test group consisted of 6 healthy male ICR mice, 18-25 g, purchased from Charles River, randomly divided into 2 groups of 3.
Preparation: weighing a certain amount of compound, adding into 2% DMSO+15% Solutol+83% physiological saline, and preparing into clear solution.
Dosage is as follows: ICR mice were fasted overnight and the compounds were administered at either a gastric lavage dose of 10 mg/kg or a dose of 5 mg/kg intravenously. The administration volumes for intragastric and intravenous administration were 10 mL/kg and 5 mL/kg, respectively. Unified feeding is performed 2 hours after administration.
Sample collection: about 30 μl of blood was collected through the great saphenous vein 5 minutes (intravenous), 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, and 7 hours after administration. Blood is put into a container containing K 2 In a commercially available tube of EDTA, the blood sample was then centrifuged at 4600 rpm at 4℃for 5 minutes to obtain a plasma sample, and all plasma samples were then flash frozen on dry ice and kept at-70℃until LC-MS/MS analysis was performed.
Sample preparation: mu.L of plasma sample was aspirated, precipitated with 50 nmol/L of alpha-naphthaleneflavone (internal standard) in methanol, the mixture was thoroughly mixed and centrifuged at 14000 rpm for 5 minutes at 4℃and 75. Mu.L of supernatant was then mixed with 75. Mu.L of methanol for LC-MS/MS analysis.
The pharmacokinetic parameter results are shown in table 1.
TABLE 1
Figure SMS_147
Conclusion: the compound has better absorption in mice, proper elimination, high exposure and higher bioavailability, and can be used for further research.
The applicant states that the present invention is illustrated by the above examples for the benzomulti-ring thiazoline amides of the present invention and the use thereof, but the present invention is not limited to the above examples, i.e., it is not meant that the present invention must be practiced by relying on the above examples. It should be apparent to those skilled in the art that any modification of the present invention, equivalent substitution of raw materials for the product of the present invention, addition of auxiliary components, selection of specific modes, etc., falls within the scope of the present invention and the scope of disclosure.
The preferred embodiments of the present invention have been described in detail above, but the present invention is not limited to the specific details of the above embodiments, and various simple modifications can be made to the technical solution of the present invention within the scope of the technical concept of the present invention, and all the simple modifications belong to the protection scope of the present invention.
In addition, the specific features described in the above embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, various possible combinations are not described further.

Claims (7)

1. The benzo-multi-ring thiazoline amide compound or pharmaceutically acceptable salt thereof is characterized by having a structure shown in the following formula I:
Figure QLYQS_1
wherein the A ring is selected from benzene ring or
Figure QLYQS_2
X is selected from-CH 2 -or-O-, n is selected from 0, 1 or 2, n is 0 indicating the absence of a group here, X is directly linked to the other end, -/->
Figure QLYQS_3
Represents the attachment site of the fused ring structure;
the B ring is selected from
Figure QLYQS_4
Any one of them;
R 1 selected from-H, -Me, -CD 3 、-Et、-i-either Pr or-Bn;
R 2 selected from the group consisting of
Figure QLYQS_5
Y is selected from-ch=or-n=;
R 3 selected from the group consisting of-H, -OH, -OMe, -OCD 3 F, -Cl or-CF 3 Any one of them;
R 4 selected from the group consisting of
Figure QLYQS_6
Any one of them;
z is selected from-ch=or-n=;
R e selected from-H, -CF 3 or-COOR f Any one of them;
R f selected from the group consisting of-H, -Me, -Eti-Pr、-n-Bu、-i-Bu、-CH 2 -CHEt 2 、-(CH 2 ) 2 -OMe、-(CH 2 ) 2 -NMe 2 Or (b)
Figure QLYQS_7
Any one of them;
R g selected from-Me or MeOCH 2 -,R h Selected from-Me, -Etn-Pr、-i-Bu、-CF 3 、-Bn、
Figure QLYQS_8
Or->
Figure QLYQS_9
Any one of them; r is R i Any one selected from-H, -Me, -F or-CN; />
Figure QLYQS_10
Representing the attachment site of the group.
2. The benzomulti-ring thiazoline amide compound or a pharmaceutically acceptable salt thereof according to claim 1, wherein the benzomulti-ring thiazoline amide compound is selected from any one of the following formulas 1 to 58:
Figure QLYQS_11
Figure QLYQS_12
Figure QLYQS_13
Figure QLYQS_14
Figure QLYQS_15
Figure QLYQS_16
Figure QLYQS_17
Figure QLYQS_18
3. a pharmaceutical composition comprising the benzomulti-ring thiazoline amide compound of claim 1 or 2 or a pharmaceutically acceptable salt thereof.
4. The pharmaceutical composition of claim 3, wherein, the pharmaceutical composition also comprises pharmaceutically acceptable pharmaceutical excipients.
5. Use of a benzopolycyclothiazoline amide compound according to claim 1 or 2 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to claim 3 or 4 for the manufacture of a medicament for the treatment and/or prevention of diseases associated with excessive activity of COUP-TFII or overexpression of COUP-TFII.
6. The use according to claim 5, wherein the disease associated with excessive activity of COUP-TFII or overexpression of COUP-TFII is prostate cancer.
7. Use of a benzomulti-ring thiazoline amide according to claim 1 or 2 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to claim 3 or 4 for the manufacture of a medicament for the treatment and/or prevention of a disease associated with NRF2 over-activity or NRF2 overexpression.
CN202310294139.0A 2023-03-24 2023-03-24 Benzo-polycyclic thiazoline amide compound and application thereof Active CN116003397B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310294139.0A CN116003397B (en) 2023-03-24 2023-03-24 Benzo-polycyclic thiazoline amide compound and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310294139.0A CN116003397B (en) 2023-03-24 2023-03-24 Benzo-polycyclic thiazoline amide compound and application thereof

Publications (2)

Publication Number Publication Date
CN116003397A CN116003397A (en) 2023-04-25
CN116003397B true CN116003397B (en) 2023-06-16

Family

ID=86030207

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310294139.0A Active CN116003397B (en) 2023-03-24 2023-03-24 Benzo-polycyclic thiazoline amide compound and application thereof

Country Status (1)

Country Link
CN (1) CN116003397B (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011017519A2 (en) * 2009-08-05 2011-02-10 The Johns Hopkins University Inhibitors of methionine aminopeptidases and methods of treating disorders
CN103827099B (en) * 2011-09-23 2015-09-09 柳韩洋行 Containing the benzothiazole derivant of phthalimide or its salt and the pharmaceutical composition comprising it
US20190209532A1 (en) * 2016-08-10 2019-07-11 Biohaven Pharmaceutical Holding Company Ltd. Acyl benzo[d]thiazol-2-amine and their methods of use
CN107033100B (en) * 2017-05-24 2019-03-01 济南大学 A kind of benzothiazole derivant, preparation method and its medical usage
WO2019222134A1 (en) * 2018-05-14 2019-11-21 Baylor College Of Medicine Coup-tfii receptor inhibitors and methods using same
AU2019326368B2 (en) * 2018-08-20 2021-11-11 Janssen Pharmaceutica Nv Inhibitors of KEAP1-Nrf2 protein-protein interaction

Also Published As

Publication number Publication date
CN116003397A (en) 2023-04-25

Similar Documents

Publication Publication Date Title
EP3070089B1 (en) Aminomethyl tryptanthrin derivative, preparation method and application thereof
CN104812749B (en) Indolecarboxamide derivant as P2X7 receptor antagonist
CN110099900B (en) Hedgehog pathway inhibitors against Smoothened mutants
EA022813B1 (en) METHOD FOR PURIFYING METHYL{4,6-DIAMINO-2-[1-(2-FLUOROBENZYL)-1H-PYRAZOLO[3,4-b]PYRIDINO-3-YL]PYRIMIDINO-5-YL}METHYL CARBAMATE AND A DERIVATIVE THEREOF AS SULFINYLDIMETHANE
JP2020097526A (en) Heterocyclic compound
CN112300153B (en) Heterocyclic compound, pharmaceutical composition and application
KR102530597B1 (en) Compounds that induces anti-aging gene klotho and use thereof
CN114409680B (en) PPAR agonists and uses thereof
WO2012037351A1 (en) Compounds
CN106905319B (en) Preparation method of substituted benzenesulfonyl kuhseng butane or hydrochloride thereof
CN116262736A (en) Novel oxo-pyridine compound and preparation method and application thereof
EP4323355A1 (en) Uracil derivatives as trpa1 inhibitors
CN116003397B (en) Benzo-polycyclic thiazoline amide compound and application thereof
CN108752412B (en) Boswellic acid derivatives and their use
CN114195776B (en) Preparation and application of novel FXR small molecule agonist
AU2009301797B2 (en) Indazole derivative
JP2014051463A (en) Compound transported by lst-1 and/or lst-2
CN116199682A (en) Phenyl thiazolyl benzamide compound and application thereof
US8536342B2 (en) Process for producing thiabenzoazulene-propionic acid derivative
JP2022551947A (en) Substituted tricyclic compounds as PRMT5 inhibitors and their applications
CN112209896A (en) Thiazolidinedione derivatives and pharmaceutical compositions containing the same
ES2297415T3 (en) INTERMEDIATE COMPOUND THAT IS USED FOR THE PREPARATION OF PIOGLITAZONA.
CN111116551A (en) 1-azaspiro [5.5] undecan-3-ones and 1-azaspiro [5.5] undecan-3-ols
PT1648444E (en) Indol-7 sulfonamide derivatives, their preparation and their use 5-ht-6 as modulators
CN104016878B (en) Aryl acetic acid derivative as well as preparation method and medical application thereof

Legal Events

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